Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
1.
J Ovarian Res ; 17(1): 97, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38720330

RESUMEN

The epidermal growth factor (EGF)-like factors, comprising amphiregulin (AREG), betacellulin (BTC), and epiregulin (EREG), play a critical role in regulating the ovulatory process. Pentraxin 3 (PTX3), an essential ovulatory protein, is necessary for maintaining extracellular matrix (ECM) stability during cumulus expansion. The aim of this study was to investigate the impact of EGF-like factors, AREG, BTC, and EREG on the expression and production of PTX3 in human granulosa-lutein (hGL) cells and the molecular mechanisms involved. Our results demonstrated that AREG, BTC, and EREG could regulate follicular function by upregulating the expression and increasing the production of PTX3 in both primary (obtained from 20 consenting patients undergoing IVF treatment) and immortalized hGL cells. The upregulation of PTX3 expression was primarily facilitated by the activation of the extracellular signal-regulated kinase 1 and 2 (ERK1/2) signaling pathway, induced by these EGF-like factors. In addition, we found that the upregulation of PTX3 expression triggered by the EGF-like factors was completely reversed by either pretreatment with the epidermal growth factor receptor (EGFR) inhibitor, AG1478, or knockdown of EGFR, suggesting that EGFR is crucial for activating the ERK1/2 signaling pathway in hGL cells. Overall, our findings indicate that AREG, BTC, and EREG may modulate human cumulus expansion during the periovulatory stage through the upregulation of PTX3.


Asunto(s)
Anfirregulina , Betacelulina , Proteína C-Reactiva , Epirregulina , Células Lúteas , Componente Amiloide P Sérico , Regulación hacia Arriba , Femenino , Humanos , Anfirregulina/metabolismo , Anfirregulina/genética , Betacelulina/metabolismo , Proteína C-Reactiva/metabolismo , Proteína C-Reactiva/genética , Factor de Crecimiento Epidérmico/metabolismo , Factor de Crecimiento Epidérmico/farmacología , Epirregulina/metabolismo , Epirregulina/genética , Receptores ErbB/metabolismo , Células Lúteas/metabolismo , Sistema de Señalización de MAP Quinasas , Componente Amiloide P Sérico/metabolismo , Componente Amiloide P Sérico/genética
2.
Int J Mol Sci ; 25(8)2024 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-38673992

RESUMEN

Lipopolysaccharides (LPSs) have been reported to be important factors in promoting the progression of hepatocellular carcinoma (HCC), but the corresponding molecular mechanisms remain to be elucidated. We hypothesize that epiregulin (EREG), an epidermal growth factor (EGF) family member derived from hepatic stellate cells (HSCs) and activated by LPS stimulation, is a crucial mediator of HCC progression with epidermal growth factor receptor (EGFR) expression in the tumor microenvironment. We used a mouse xenograft model of Huh7 cells mixed with half the number of LX-2 cells, with/without intraperitoneal LPS injection, to elucidate the role of EREG in LPS-induced HCC. In the mouse model, LPS administration significantly enlarged the size of xenografted tumors and elevated the expression of EREG in tumor tissues compared with those in negative controls. Moreover, CD34 immunostaining and the gene expressions of angiogenic markers by a reverse transcription polymerase chain reaction revealed higher vascularization, with increased interleukin-8 (IL-8) expression in the tumors of the mice group treated with LPS compared to those without LPS. Our data collectively suggested that EREG plays an important role in the cancer microenvironment under the influence of LPS to increase not only the tumor cell growth and migration/invasion of EGFR-positive HCC cells but also tumor neovascularization via IL-8 signaling.


Asunto(s)
Carcinoma Hepatocelular , Epirregulina , Receptores ErbB , Lipopolisacáridos , Neoplasias Hepáticas , Transducción de Señal , Microambiente Tumoral , Epirregulina/metabolismo , Epirregulina/genética , Animales , Receptores ErbB/metabolismo , Receptores ErbB/genética , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/inducido químicamente , Neoplasias Hepáticas/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/inducido químicamente , Carcinoma Hepatocelular/genética , Ratones , Línea Celular Tumoral , Neovascularización Patológica/metabolismo , Carcinogénesis/metabolismo , Carcinogénesis/genética , Carcinogénesis/patología , Interleucina-8/metabolismo , Interleucina-8/genética , Proliferación Celular , Masculino , Células Estrelladas Hepáticas/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos
3.
EMBO J ; 43(8): 1388-1419, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38514807

RESUMEN

Neocortex expansion during evolution is linked to higher numbers of neurons, which are thought to result from increased proliferative capacity and neurogenic potential of basal progenitor cells during development. Here, we show that EREG, encoding the growth factor EPIREGULIN, is expressed in the human developing neocortex and in gorilla cerebral organoids, but not in the mouse neocortex. Addition of EPIREGULIN to the mouse neocortex increases proliferation of basal progenitor cells, whereas EREG ablation in human cortical organoids reduces proliferation in the subventricular zone. Treatment of cortical organoids with EPIREGULIN promotes a further increase in proliferation of gorilla but not of human basal progenitor cells. EPIREGULIN competes with the epidermal growth factor (EGF) to promote proliferation, and inhibition of the EGF receptor abrogates the EPIREGULIN-mediated increase in basal progenitor cells. Finally, we identify putative cis-regulatory elements that may contribute to the observed inter-species differences in EREG expression. Our findings suggest that species-specific regulation of EPIREGULIN expression may contribute to the increased neocortex size of primates by providing a tunable pro-proliferative signal to basal progenitor cells in the subventricular zone.


Asunto(s)
Epirregulina , Neocórtex , Animales , Humanos , Ratones , Proliferación Celular , Epirregulina/genética , Epirregulina/metabolismo , Gorilla gorilla/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Neocórtex/citología , Neocórtex/metabolismo , Primates/fisiología
4.
Clin Cancer Res ; 29(20): 4021-4023, 2023 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-37594733

RESUMEN

Overexpression of the EGFR ligands amphiregulin (AREG)/epiregulin (EREG) may be a surrogate of EGFR dependency regardless of sidedness in metastatic colorectal cancer. High AREG/EREG may be coupled with negative hyper-selection (i.e., lack of genomic drivers of primary resistance beyond RAS and BRAF) to identify patients with right-sided tumors and potential sensitivity to EGFR blockade. See related article by Williams et al., p. 4153.


Asunto(s)
Neoplasias del Colon , Neoplasias Colorrectales , Neoplasias del Recto , Humanos , Anfirregulina/genética , Epirregulina/genética , Multiómica , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Receptores ErbB/genética , Péptidos y Proteínas de Señalización Intercelular , Anticuerpos , Inteligencia Artificial
5.
J Pathol ; 261(1): 28-42, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37345534

RESUMEN

The prognosis of gallbladder cancer (GBC) remains poor, and a better understanding of GBC molecular mechanisms is important. Genome sequencing of human GBC has demonstrated that loss-of-function mutations of E74-like ETS transcription factor 3 (ELF3) are frequently observed, with ELF3 considered to be a tumour suppressor in GBC. To clarify the underlying molecular mechanisms by which ELF3 suppresses GBC development, we performed in vivo analysis using a combination of autochthonous and allograft mouse models. We first evaluated the clinical significance of ELF3 expression in human GBC tissues and found that low ELF3 expression was associated with advanced clinical stage and deep tumour invasion. For in vivo analysis, we generated Pdx1-Cre; KrasG12D ; Trp53R172H ; Elf3f/f (KPCE) mice and Pdx1-Cre; KrasG12D ; Trp53R172H ; Elf3wt/wt (KPC) mice as a control and analysed their gallbladders histologically. KPCE mice developed larger papillary lesions in the gallbladder than those developed by KPC mice. Organoids established from the gallbladders of KPCE and KPC mice were analysed in vitro. RNA sequencing showed upregulated expression of epiregulin (Ereg) in KPCE organoids, and western blotting revealed that EGFR/mechanical targets of rapamycin complex 1 (mTORC1) were upregulated in KPCE organoids. In addition, ChIP assays on Elf3-overexpressing KPCE organoids showed that ELF3 directly regulated Ereg. Ereg deletion in KPCE organoids (using CRISPR/Cas9) induced EGFR/mTORC1 downregulation, indicating that ELF3 controlled EGFR/mTORC1 activity through regulation of Ereg expression. We also generated allograft mouse models using KPCE and KPC organoids and found that KPCE organoid allograft tumours exhibited poorly differentiated structures with mTORC1 upregulation and mesenchymal phenotype, which were suppressed by Ereg deletion. Furthermore, EGFR/mTORC1 inhibition suppressed cell proliferation and epithelial-mesenchymal transition in KPCE organoids. Our results suggest that ELF3 suppresses GBC development via downregulation of EREG/EGFR/mTORC1 signalling. EGFR/mTORC1 inhibition is a potential therapeutic option for GBC with ELF3 mutation. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Asunto(s)
Neoplasias de la Vesícula Biliar , Proteínas Proto-Oncogénicas p21(ras) , Humanos , Animales , Ratones , Proteínas Proto-Oncogénicas p21(ras)/genética , Epirregulina/genética , Epirregulina/metabolismo , Neoplasias de la Vesícula Biliar/genética , Neoplasias de la Vesícula Biliar/metabolismo , Neoplasias de la Vesícula Biliar/patología , Regulación hacia Abajo , Línea Celular Tumoral , Proliferación Celular/genética , Proteínas Proto-Oncogénicas c-ets/genética , Receptores ErbB/genética , Receptores ErbB/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteínas de Unión al ADN/genética , Factores de Transcripción/genética
6.
Cells ; 12(5)2023 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-36899869

RESUMEN

(1) Background: Epiregulin (EREG) is a ligand of EGFR and ErB4 involved in the development and the progression of various cancers including head and neck squamous cell carcinoma (HNSCC). Its overexpression in HNSCC is correlated with short overall survival and progression-free survival but predictive of tumors responding to anti-EGFR therapies. Besides tumor cells, macrophages and cancer-associated fibroblasts shed EREG in the tumor microenvironment to support tumor progression and to promote therapy resistance. Although EREG seems to be an interesting therapeutic target, no study has been conducted so far on the consequences of EREG invalidation regarding the behavior and response of HNSCC to anti-EGFR therapies and, more specifically, to cetuximab (CTX); (2) Methods: EREG was silenced in various HNSCC cell lines. The resulting phenotype (growth, clonogenic survival, apoptosis, metabolism, ferroptosis) was assessed in the absence or presence of CTX. The data were confirmed in patient-derived tumoroids; (3) Results: Here, we show that EREG invalidation sensitizes cells to CTX. This is illustrated by the reduction in cell survival, the alteration of cell metabolism associated with mitochondrial dysfunction and the initiation of ferroptosis characterized by lipid peroxidation, iron accumulation and the loss of GPX4. Combining ferroptosis inducers (RSL3 and metformin) with CTX drastically reduces the survival of HNSCC cells but also HNSCC patient-derived tumoroids; (4) Conclusions: The loss of EREG might be considered in clinical settings as a predictive biomarker for patients that might undergo ferroptosis in response to CTX and that might benefit the most from the combination of ferroptosis inducers and CTX.


Asunto(s)
Ferroptosis , Neoplasias de Cabeza y Cuello , Humanos , Cetuximab/farmacología , Epirregulina/genética , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Péptidos y Proteínas de Señalización Intercelular , Carcinoma de Células Escamosas de Cabeza y Cuello/tratamiento farmacológico , Microambiente Tumoral
7.
Anticancer Res ; 42(8): 3873-3878, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35896250

RESUMEN

BACKGROUND/AIM: Epiregulin (EREG) is a ligand of the epidermal growth factor receptor (EGFR) and promotes tumour progression mainly by stimulating the EGF pathway. We investigated the clinical significance of EREG mRNA expression in cancer tissues from patients with gastric cancer (GC) in pathological (p) Stage II/III who have undergone curative surgery. PATIENTS AND METHODS: Expression of EREG mRNA was measured in cancer tissues obtained from 253 patients with pStage II/III GC who underwent curative surgery. Patients were divided into groups based on high or low expression of EREG mRNA. We examined the relationship between EREG mRNA expression levels and clinicopathological features and survival. RESULTS: Clinicopathological features did not vary between the high and low EREG mRNA expression groups. Overall survival was significantly lower in the high-expression group compared to that in the low-expression group (5-year survival probability: 55.0% vs. 73.0%; p=0.005). Multivariate analysis showed EREG mRNA expression to be an independent predictor of poor survival (hazard ratio=1.794; 95% confidence interval=1.186-2.712; p=0.006). CONCLUSION: Expression of EREG mRNA in cancer tissue from patients with pStage II/III GC may be a useful prognostic marker after curative surgery.


Asunto(s)
Neoplasias Gástricas , Epirregulina/genética , Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , ARN Mensajero/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/cirugía
8.
Stem Cell Res Ther ; 13(1): 197, 2022 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-35551652

RESUMEN

BACKGROUND: Chemoresistance often causes the failure of treatment and death of patients with advanced non-small-cell lung cancer. However, there is still no resistance genes signature and available enriched signaling derived from a comprehensive RNA-Seq data analysis of lung cancer patients that could act as a therapeutic target to re-sensitize the acquired resistant cancer cells to chemo-drugs. Hence, in this study, we aimed to identify the resistance signature for clinical lung cancer patients and explore the regulatory mechanism. METHOD: Analysis of RNA-Seq data from clinical lung cancer patients was conducted in R studio to identify the resistance signature. The resistance signature was validated by survival time of lung cancer patients and qPCR in chemo-resistant cells. Cytokine application, small-interfering RNA and pharmacological inhibition approaches were applied to characterize the function and molecular mechanism of EREG and downstream signaling in chemoresistance regulation via stemness. RESULTS: The RTK and vitamin D signaling were enriched among resistance genes, where 6 genes were validated as resistance signature and associated with poor survival in patients. EREG/ERK signaling was activated by chemo-drugs in NSCLC cells. EREG protein promoted the NSCLC resistance to chemo-drugs by increasing stemness genes expression. Additionally, inhibition of EREG/ErbB had downregulated ERK signaling, resulting in decreased expression of stemness-associated genes and subsequently re-sensitized the resistant NSCLC cells and spheres to chemo-drugs. CONCLUSIONS: These findings revealed 6 resistance genes signature and proved that EREG/ErbB regulated the stemness to maintain chemoresistance of NSCLC via ERK signaling. Therefore, targeting EREG/ErbB might significantly and effectively resolve the chemoresistance issue.


Asunto(s)
Antineoplásicos , Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Línea Celular Tumoral , Resistencia a Antineoplásicos/genética , Epirregulina/genética , Epirregulina/metabolismo , Epirregulina/farmacología , Humanos , Péptidos y Proteínas de Señalización Intercelular/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Transducción de Señal
9.
J Obstet Gynaecol Res ; 48(7): 1848-1858, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35491469

RESUMEN

AIMS: Cervical squamous cell carcinoma (SCC) is one of the most frequent malignancies of the female reproductive system. The malignant mechanism of SCC has not been totally clarified. We aimed to discover a list of differentially expressed genes (DEGs) to identify the malignant mechanism of cervical SCC. METHOD: Three expression chips (GSE7803, GSE9750, and GSE64217) were downloaded from gene expression omnibus (GEO) datasets. After standardization, 50 cervical SCC tumor tissues and 33 normal cervical tissues (NCTs) were included for DEGs and clustering analysis. RobustRankAggreg (RRA) algorithm was used to extract the overlapping DEGs. Gene function and signaling pathway analysis was implemented based on Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway databases. Protein-protein interaction (PPI) analysis and prognostic analysis were also carried out to identify the DEGs as prognostic markers for cervical SCC. RESULTS: Totally 100 DEGs were obtained from GSE7803, 319 DEGs from GSE9750, and 1639 DEGs from GSE64217. RRA analysis uncovered 17 upregulated DEGs and 25 downregulated DEGs. GO and KEGG analysis showed DEGs were involved in the mediation of extracellular functions, cell-cell interactions, and cell metabolism. PPI network showed a close interaction among the integrated DEGs. Prognostic analysis showed gene secreted phosphoprotein 1 (SPP1) and epiregulin (EREG) genes were independent prognostic predictors of cervical SCC. CONCLUSION: The gene expression profile was changed in cervical SCC tumor tissues compared to NCTs. SPP1 and EREG were postulated as prognostic markers for cervical SCC, which might be potential targets for clinical therapy of cervical SCC.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias del Cuello Uterino , Biomarcadores de Tumor/genética , Carcinoma de Células Escamosas/genética , Biología Computacional , Epirregulina/genética , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Ontología de Genes , Redes Reguladoras de Genes , Humanos , Osteopontina/genética , Pronóstico , Mapas de Interacción de Proteínas/genética , Neoplasias del Cuello Uterino/genética
10.
Cell Biol Toxicol ; 38(5): 865-887, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-34036453

RESUMEN

Exposure to environmental and occupational contaminants leads to lung cancer. 3-Nitrobenzanthrone (3-nitro-7H-benz[de]anthracen-7-one, 3-NBA) is a potential carcinogen in ambient air or diesel particulate matter. Studies have revealed that short-term exposure to 3-NBA induces cell death, reactive oxygen species activation, and DNA adduct formation and damage. However, details of the mechanism by which chronic exposure to 3-NBA influences lung carcinogenesis remain largely unknown. In this study, human lung epithelial BEAS-2B cells were continuously exposed to 0-10-µM 3-NBA for 6 months. NanoString analysis was conducted to evaluate gene expression in the cells, revealing that 3-NBA-mediated transformation results in a distinct gene expression signature including carbon cancer metabolism, metastasis, and angiogenesis. Alterations in tumor-promoting genes such as EREG (epiregulin), SOX9, E-cadherin, TWIST, and IL-6 were involved in epithelial cell aggressiveness. Kaplan-Meier plotter analyses indicated that increased EREG and IL-6 expressions in early-stage lung cancer cells are correlated with poor survival. In vivo xenografts on 3-NBA-transformed cells exhibited prominent tumor formation and metastasis. EREG knockout cells exposed to 3-NBA for a short period exhibited high apoptosis and low colony formation. By contrast, overexpression of EREG in 3-NBA-transformed cells markedly activated the PI3K/AKT and MEK/ERK signaling pathways, resulting in tumorigenicity. Furthermore, elevated IL-6 and EREG expressions synergistically led to STAT3 signaling activation, resulting in clonogenic cell survival and migration. Taken together, chronic exposure of human lung epithelial cells to 3-NBA leads to malignant transformation, in which the EREG signaling pathway plays a pivotal mediating role. • Short-term exposure of lung epithelial cells to 3-NBA can lead to ROS production and cell apoptosis. • Long-term chronic exposure to 3-NBA upregulates the levels of tumor-promoting genes such as EREG and IL-6. • Increased EREG expression in 3-NBA-transformed cells markedly contributes to tumorigenesis through PI3K/AKT and MEK/ERK activation and synergistically enhances the IL-6/STAT3 signaling pathway, which promotes tumorigenicity.


Asunto(s)
Aductos de ADN , Neoplasias Pulmonares , Benzo(a)Antracenos , Cadherinas/metabolismo , Carbono/metabolismo , Carbono/farmacología , Carcinogénesis/metabolismo , Carcinógenos , Transformación Celular Neoplásica/metabolismo , Aductos de ADN/metabolismo , Aductos de ADN/farmacología , Epirregulina/genética , Epirregulina/metabolismo , Epirregulina/farmacología , Células Epiteliales/metabolismo , Humanos , Interleucina-6/metabolismo , Pulmón/metabolismo , Neoplasias Pulmonares/inducido químicamente , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/farmacología , Material Particulado/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal
11.
Int J Mol Sci ; 22(23)2021 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-34884633

RESUMEN

Aberrant activation of the epidermal growth factor receptor (EGFR/ERBB1) by erythroblastic leukemia viral oncogene homolog (ERBB) ligands contributes to various tumor malignancies, including lung cancer and colorectal cancer (CRC). Epiregulin (EREG) is one of the EGFR ligands and is low expressed in most normal tissues. Elevated EREG in various cancers mainly activates EGFR signaling pathways and promotes cancer progression. Notably, a higher EREG expression level in CRC with wild-type Kirsten rat sarcoma viral oncogene homolog (KRAS) is related to better efficacy of therapeutic treatment. By contrast, the resistance of anti-EGFR therapy in CRC was driven by low EREG expression, aberrant genetic mutation and signal pathway alterations. Additionally, EREG overexpression in non-small cell lung cancer (NSCLC) is anticipated to be a therapeutic target for EGFR-tyrosine kinase inhibitor (EGFR-TKI). However, recent findings indicate that EREG derived from macrophages promotes NSCLC cell resistance to EGFR-TKI treatment. The emerging events of EREG-mediated tumor promotion signals are generated by autocrine and paracrine loops that arise from tumor epithelial cells, fibroblasts, and macrophages in the tumor microenvironment (TME). The TME is a crucial element for the development of various cancer types and drug resistance. The regulation of EREG/EGFR pathways depends on distinct oncogenic driver mutations and cell contexts that allows specific pharmacological targeting alone or combinational treatment for tailored therapy. Novel strategies targeting EREG/EGFR, tumor-associated macrophages, and alternative activation oncoproteins are under development or undergoing clinical trials. In this review, we summarize the clinical outcomes of EREG expression and the interaction of this ligand in the TME. The EREG/EGFR pathway may be a potential target and may be combined with other driver mutation targets to combat specific cancers.


Asunto(s)
Neoplasias del Colon/metabolismo , Epirregulina/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Epirregulina/genética , Receptores ErbB/genética , Receptores ErbB/metabolismo , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Macrófagos/metabolismo , Terapia Molecular Dirigida , Mutación , Transducción de Señal , Microambiente Tumoral
12.
Int J Mol Sci ; 22(22)2021 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-34830244

RESUMEN

p130 Crk-associated substrate (p130Cas) is associated with poor prognosis and treatment resistance in breast and lung cancers. To elucidate p130Cas functional and clinical role in colorectal cancer (CRC) progression/therapy resistance, we performed cell culture experiments and bioinformatic/statistical analyses of clinical data sets. p130Cas expression was associated with poor survival in the cancer genome atlas (TCGA) data set. Knockdown/reconstitution experiments showed that p130Cas drives migration but, unexpectedly, inhibits proliferation in CRC cells. TCGA data analyses identified the growth factor epiregulin (EREG) as inversely correlated with p130Cas. p130Cas knockdown and simultaneous EREG treatment further enhanced proliferation. RNA interference and EREG treatment experiments suggested that p130Cas/EREG limit each other's expression/activity. Inverse p130Cas/EREG Spearman correlations were prominent in right-sided and earlier stage CRC. p130Cas was inducible by 5-fluorouracil (5-FU) and FOLFIRI (folinic acid, 5-FU, irinotecan), and p130Cas and EREG were upregulated in distant metastases (GSE121418). Positive p130Cas/EREG correlations were observed in metastases, preferentially in post-treatment samples (especially pulmonary metastases). p130Cas knockdown sensitized CRC cells to FOLFIRI independent of EREG treatment. RNA sequencing and gene ontology analyses revealed that p130Cas is involved in cytochrome P450 drug metabolism and epithelial-mesenchymal transition. p130Cas expression was associated with poor survival in right-sided, stage I/II, MSS (microsatellite stable), or BRAF-mutated CRC. In summary, p130Cas represents a prognostic factor and potential therapeutic target in CRC.


Asunto(s)
Neoplasias de la Mama/diagnóstico , Neoplasias Colorrectales/diagnóstico , Proteína Sustrato Asociada a CrK/genética , Epirregulina/genética , Transición Epitelial-Mesenquimal/genética , Adulto , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Atlas como Asunto , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/mortalidad , Camptotecina/análogos & derivados , Camptotecina/uso terapéutico , Línea Celular Tumoral , Proliferación Celular , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/mortalidad , Proteína Sustrato Asociada a CrK/antagonistas & inhibidores , Proteína Sustrato Asociada a CrK/metabolismo , Epirregulina/metabolismo , Femenino , Fluorouracilo/uso terapéutico , Regulación Neoplásica de la Expresión Génica , Ontología de Genes , Humanos , Leucovorina/uso terapéutico , Masculino , Persona de Mediana Edad , Anotación de Secuencia Molecular , Mutación , Metástasis de la Neoplasia , Estadificación de Neoplasias , Pronóstico , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Análisis de Supervivencia
13.
Tissue Cell ; 73: 101669, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34715618

RESUMEN

Gouty arthritis (GA) is a chronic inflammatory disease characterized by the deposition of monosodium urate (MSU) crystals within joints. MiR-192-5p is shown to be low-expressed in GA patients. However, the potential mechanism involving miR-192-5p in GA remains unclear. In the current study, a significant reduction in miR-192-5p and an increase in epiregulin (EREG) were observed in serum of GA patients, suggesting that miR-192-5p and EREG were involved in the pathogenic process of GA. A mouse GA model was established via 0.5 mg/20 µL MSU crystal administration. To investigate the effect of miR-192-5p on GA, mice were injected with miR-192-5p agomir or NC agomir before modeling. We found that miR-192-5p overexpression induced by miR-192-5p agomir reduced EREG expression, attenuated ankle joint swelling and synovial inflammatory cell infiltration and improved bone erosion in MSU-induced GA mice. MiR-192-5p decreased CD16/32+ (M1 marker) macrophages, but increased CD206 (M2 marker) expression in synovium of GA models. In vitro, RAW264.7 macrophages were stimulated with miR-192-5p mimic or NC mimic under IFNγ plus LPS-stimulated M1 polarization condition. MiR-192-5p reduced the release of inflammatory cytokines TNF-α and IL-1ß, decreased iNOS expression, and inhibited CD16/32 expression, indicating the blockade of M1 macrophage activation. Luciferase reporter system revealed the target interaction between miR-192-5p and EREG. Further rescue experiments demonstrated that EREG overexpression partly reversed the inhibitory role of miR-192-5p on M1 macrophage polarization manifested by elevated iNOS and CD16/32 levels. Collectively, miR-192-5p ameliorates inflammatory response in GA by inhibiting M1 macrophage activation via inhibiting EREG protein.


Asunto(s)
Artritis Gotosa/genética , Polaridad Celular/genética , Regulación hacia Abajo/genética , Epirregulina/genética , Macrófagos/metabolismo , MicroARNs/metabolismo , Animales , Artritis Gotosa/sangre , Artritis Gotosa/patología , Secuencia de Bases , Epirregulina/sangre , Epirregulina/metabolismo , Inflamación/genética , Inflamación/patología , Activación de Macrófagos/genética , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , Células RAW 264.7
14.
J Mol Biol ; 433(21): 167240, 2021 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-34508725

RESUMEN

Receptor tyrosine kinases (RTK) bind growth factors and are critical for cell proliferation and differentiation. Their dysregulation leads to a loss of growth control, often resulting in cancer. Epidermal growth factor receptor (EGFR) is the prototypic RTK and can bind several ligands exhibiting distinct mitogenic potentials. Whereas the phosphorylation on individual EGFR sites and their roles for downstream signaling have been extensively studied, less is known about ligand-specific ubiquitination events on EGFR, which are crucial for signal attenuation and termination. We used a proteomics-based workflow for absolute quantitation combined with mathematical modeling to unveil potentially decisive ubiquitination events on EGFR from the first 30 seconds to 15 minutes of stimulation. Four ligands were used for stimulation: epidermal growth factor (EGF), heparin-binding-EGF like growth factor, transforming growth factor-α and epiregulin. Whereas only little differences in the order of individual ubiquitination sites were observed, the overall amount of modified receptor differed depending on the used ligand, indicating that absolute magnitude of EGFR ubiquitination, and not distinctly regulated ubiquitination sites, is a major determinant for signal attenuation and the subsequent cellular outcomes.


Asunto(s)
Factor de Crecimiento Epidérmico/metabolismo , Epirregulina/metabolismo , Factor de Crecimiento Similar a EGF de Unión a Heparina/metabolismo , Transducción de Señal/genética , Factor de Crecimiento Transformador alfa/metabolismo , Secuencia de Aminoácidos , Línea Celular Tumoral , Factor de Crecimiento Epidérmico/química , Factor de Crecimiento Epidérmico/genética , Epirregulina/química , Epirregulina/genética , Células Epiteliales/citología , Células Epiteliales/metabolismo , Receptores ErbB/química , Receptores ErbB/genética , Receptores ErbB/metabolismo , Expresión Génica , Factor de Crecimiento Similar a EGF de Unión a Heparina/química , Factor de Crecimiento Similar a EGF de Unión a Heparina/genética , Humanos , Ligandos , Modelos Moleculares , Mutación , Fosforilación , Conformación Proteica , Procesamiento Proteico-Postraduccional , Proteómica , Factor de Crecimiento Transformador alfa/química , Factor de Crecimiento Transformador alfa/genética , Ubiquitinación
15.
Purinergic Signal ; 17(4): 681-691, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34351588

RESUMEN

The ability of cardiac adipose-derived stem cells (cADSC) to differentiate into multiple cell types has opened new perspectives in cardiac cell-based regenerative therapies. P2Y nucleotide receptors have already been described as regulators of adipogenic differentiation of cADSC and bone marrow-derived stem cells. In this study, we defined UTP as a regulator of cADSC endothelial differentiation. A daily UTP stimulation of cADSC during endothelial predifferentiation increased their capacity to form an endothelial network in matrigel. Additionally, pro-angiogenic UTP target genes such as epiregulin and hyaluronan synthase-1 were identified in predifferentiated cADSC by RNA sequencing experiments. Their regulation by UTP was confirmed by qPCR and ELISA experiments. We then evaluated the capacity of UTP-treated predifferentiated cADSC to increase post-ischemic revascularization in mice subjected to left anterior descending artery ligation. Predifferentiated cADSC treated or not with UTP were injected in the periphery of the infarcted zone, 3 days after ligation. We observed a significant increase of capillary density 14 and 30 days after UTP-treated predifferentiated cADSC injection, correlated with a reduction of cardiac fibrosis. This revascularization increase was not observed after injection of UTP-treated cADSC deficient for UTP and ATP nucleotide receptor P2Y2. The present study highlights the P2Y2 receptor as a regulator of cADSC endothelial differentiation and as a potential target for the therapeutic use of cADSC in post-ischemic heart revascularization.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Células Madre Multipotentes/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Uridina Trifosfato/farmacología , Animales , Epirregulina/genética , Epirregulina/metabolismo , Ratones , Ratones Noqueados , Células Madre Multipotentes/metabolismo , Receptores Purinérgicos P2Y2/genética , Receptores Purinérgicos P2Y2/metabolismo
16.
J Cell Sci ; 134(18)2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34406412

RESUMEN

In polarized MDCK cells, disruption of the tyrosine-based YXXΦ basolateral trafficking motif (Y156A) in the epidermal growth factor receptor (EGFR) ligand epiregulin (EREG), results in its apical mistrafficking and transformation in vivo. However, the mechanisms underlying these dramatic effects are unknown. Using a doxycycline-inducible system in 3D Matrigel cultures, we now show that induction of Y156A EREG in fully formed MDCK cysts results in direct and complete delivery of mutant EREG to the apical cell surface. Within 3 days of induction, ectopic lumens were detected in mutant, but not wild-type, EREG-expressing cysts. Of note, these structures resembled histological features found in subcutaneous xenografts of mutant EREG-expressing MDCK cells. These ectopic lumens formed de novo rather than budding from the central lumen and depended on metalloprotease-mediated cleavage of EREG and subsequent EGFR activity. Moreover, the most frequent EREG mutation in human cancer (R147stop) resulted in its apical mistrafficking in engineered MDCK cells. Thus, induction of EREG apical mistrafficking is sufficient to disrupt selective aspects of polarity of a preformed polarized epithelium. This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Receptores ErbB , Transducción de Señal , Epirregulina/genética , Epirregulina/metabolismo , Receptores ErbB/genética , Receptores ErbB/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular , Fosforilación
17.
Gynecol Endocrinol ; 37(9): 836-840, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34223781

RESUMEN

OBJECTIVE: Ovarian hyperstimulation syndrome (OHSS) is mainly caused by human chorionic gonadotropin (hCG) through vasoactive mediators such as vascular endothelial growth factor (VEGF) and various inflammatory factors. Our previous study showed that soluble receptor for advanced glycation end products (sRAGE) played a protective role in PCOS by inhibiting VEGF, so wanted to explore the role of sRAGE in OHSS. METHODS: Two sets of experiments were performed in this study. In part one, sRAGE protein levels in follicular fluid (FF) samples from 60 patients with OHSS and 60 non-OHSS patients were measured by ELISA. In part two, ovarian granulosa cells were isolated from an additional 25 patients with OHSS and cultured. Then, ovarian granulosa cells were treated with different concentrations of sRAGE. Granulosa cells cultured without sRAGE stimulation were used as the control group. The levels of VEGF, amphiregulin (AREG), betacellulin (BTC), and epiregulin (EREG) mRNA were examined by quantitative RT-PCR. The protein levels of VEGF, AREG, BTC, and EREG were measured by ELISA. RESULTS: Compared with non-OHSS patients, patients with OHSS exhibited lower sRAGE levels in both serum and FF (p < .05). Treatment with sRAGE decreased the production of VEGF, and the effects were dependent on the concentration of sRAGE (p < .05). Simultaneously, the expression of the EGF-like growth factors AREG, BTC and EREG was decreased, and their expression was dependent on the concentration of sRAGE (p < .05). CONCLUSIONS: sRAGE downregulate VEGF expression in OHSS ovarian granulosa cells, in which EGF-like growth factor pathway may be involved, and sRAGE may play a potential protective role in OHSS.


Asunto(s)
Regulación hacia Abajo/efectos de los fármacos , Células de la Granulosa/metabolismo , Síndrome de Hiperestimulación Ovárica/metabolismo , Receptor para Productos Finales de Glicación Avanzada/administración & dosificación , Factores de Crecimiento Endotelial Vascular/genética , Adulto , Anfirregulina/análisis , Anfirregulina/genética , Betacelulina/análisis , Betacelulina/genética , Células Cultivadas , Epirregulina/análisis , Epirregulina/genética , Femenino , Líquido Folicular/química , Humanos , ARN Mensajero/análisis , Receptor para Productos Finales de Glicación Avanzada/análisis , Receptor para Productos Finales de Glicación Avanzada/sangre , Factores de Crecimiento Endotelial Vascular/análisis
18.
J Dermatol ; 48(9): 1433-1438, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34128258

RESUMEN

Psoriasis, an immune-mediated inflammatory disease, is characterized by keratinocyte hyperproliferation. Tumor necrosis factor (TNF)-α, interleukin (IL)-23, and IL-17A play critical roles in the pathogenesis of psoriasis. IL-17A secreted by T-helper 17 acts more directly against keratinocytes than TNF-α or IL-23 do. Regarding the receptors of cytokines, fibroblasts also express receptors against IL-17A and TNF-α, and induce the production of growth factors. Epiregulin (EREG), an epidermal growth factor receptor ligand, is produced by both keratinocytes and fibroblasts. EREG enhances keratinocyte proliferation and differentiation. We hypothesized that fibroblasts stimulated with IL-17A and/or TNF-α may play a role in epidermal hyperproliferation through the production of epidermal growth factors in psoriasis. The mRNA expression of EREG was found to be significantly upregulated by co-stimulation with IL-17A and TNF-α (mean, 49.2-fold). Furthermore, the stimulation with TNF-α alone, but not IL-17A alone, induced significant increases. Immunofluorescent staining demonstrated that the protein expression level of EREG was also increased in fibroblasts stimulated with these cytokines. Stimulation with EREG significantly enhanced keratinocyte proliferation in vitro. In human psoriatic patients' skin, immunofluorescence staining of EREG showed significantly high intensity in the dermis of lesional skin. In conclusion, cytokine stimulation with TNF-α and IL-17A induces the overexpression of EREG from dermal fibroblasts in the lesional skin of psoriasis, and plays a role in epidermal hyperproliferation.


Asunto(s)
Epirregulina , Queratinocitos , Psoriasis , Proliferación Celular , Epirregulina/genética , Fibroblastos , Humanos , Psoriasis/genética , Piel
19.
Clin Cancer Res ; 27(12): 3422-3431, 2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-33888518

RESUMEN

PURPOSE: High tumor mRNA levels of the EGFR ligands amphiregulin (AREG) and epiregulin (EREG) are associated with anti-EGFR agent response in metastatic colorectal cancer (mCRC). However, ligand RNA assays have not been adopted into routine practice due to issues with analytic precision and practicality. We investigated whether AREG/EREG IHC could predict benefit from the anti-EGFR agent panitumumab. EXPERIMENTAL DESIGN: Artificial intelligence algorithms were developed to assess AREG/EREG IHC in 274 patients from the PICCOLO trial of irinotecan with or without panitumumab (Ir vs. IrPan) in RAS wild-type mCRC. The primary endpoint was progression-free survival (PFS). Secondary endpoints were RECIST response rate (RR) and overall survival (OS). Models were repeated adjusting separately for BRAF mutation status and primary tumor location (PTL). RESULTS: High ligand expression was associated with significant PFS benefit from IrPan compared with Ir [8.0 vs. 3.2 months; HR, 0.54; 95% confidence interval (CI), 0.37-0.79; P = 0.001]; whereas low ligand expression was not (3.4 vs. 4.4 months; HR, 1.05; 95% CI, 0.74-1.49; P = 0.78). The ligand-treatment interaction was significant (P interaction = 0.02) and remained significant after adjustment for BRAF-mutation status and PTL. Likewise, RECIST RR was significantly improved in patients with high ligand expression (IrPan vs. Ir: 48% vs. 6%; P < 0.0001) but not those with low ligand expression (25% vs. 14%; P = 0.10; P interaction = 0.01). The effect on OS was similar but not statistically significant. CONCLUSIONS: AREG/EREG IHC identified patients who benefitted from the addition of panitumumab to irinotecan chemotherapy. IHC is a practicable assay that may be of use in routine practice.


Asunto(s)
Inteligencia Artificial , Neoplasias Colorrectales , Anfirregulina/genética , Anfirregulina/metabolismo , Anfirregulina/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Epirregulina/genética , Epirregulina/metabolismo , Receptores ErbB/genética , Humanos , Panitumumab , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo
20.
Exp Mol Pathol ; 117: 104549, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33007298

RESUMEN

BACKGROUND: Human papillomavirus (HPV) etiology has become evident in head and neck cancers (HNCs) and HPV positivity showed a strong association with its malignant progression. Since aberrant DNA methylation is known to drive carcinogenesis and progression in HNCs, we investigated to determine target gene(s) associated with this modification. METHODS: We characterized epigenetic changes in tumor-related genes (TRGs) that are known to be associated with HNC development and its progression. RESULTS: The expression levels of 42 candidate HNC-associated genes were analyzed. Of these, 7 TGRs (CHFR, RARß, GRB7, EREG, RUNX2, RUNX3, and SMG-1) showed decreased expressions in HPV-positive (+) HNC cells compared with HPV-negative (-) HNC cells. When gene expression levels were compared corresponding to the DNA methylation conditions, GRB7 and EREG showed significant differential expression between HPV+ and HPV- cells, which suggested these genes as primary targets of epigenetic regulation in HPV-induced carcinogenesis. Furthermore, treatment with a demethylation agent, 5-aza-2'-deoxycytidine (5-aza-dc), caused restoration of EREG expression and was associated with hypomethylation of its promoter in HPV+ cells, while no changes was noted in HPV- cells. EREG promoter hypermethylation in HPV+ cells was confirmed using methylation-specific PCR (MS-PCR). CONCLUSION: We conclude that EREG is the target of epigenetic regulation in HPV+ HNCs and its suppressed expression through promoter hypermethylation is associated with the development of HPV-associated HNCs.


Asunto(s)
Alphapapillomavirus/genética , Epigénesis Genética , Epirregulina/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Alphapapillomavirus/patogenicidad , Azacitidina , Carcinogénesis/genética , Línea Celular Tumoral , ADN (Citosina-5-)-Metiltransferasa 1/genética , ADN (Citosina-5-)-Metiltransferasas/genética , Metilación de ADN/genética , ADN Metiltransferasa 3A , Decitabina , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Proteínas de Neoplasias/genética , Regiones Promotoras Genéticas/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Carcinoma de Células Escamosas de Cabeza y Cuello/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...