Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 308
Filtrar
1.
EMBO J ; 43(8): 1388-1419, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38514807

RESUMO

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.


Assuntos
Epirregulina , Neocórtex , Animais , Humanos , Camundongos , Proliferação de Células , Epirregulina/genética , Epirregulina/metabolismo , Gorilla gorilla/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Neocórtex/citologia , Neocórtex/metabolismo , Primatas/fisiologia
2.
Int J Biol Sci ; 20(3): 818-830, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38250159

RESUMO

Insulin-like growth factor 2 mRNA binding protein 2 (IGF2BP2), with high affinity to a myriad of RNA transcripts, has been shown to elicit promotive effects on tumorigenesis and metastasis. Yet, the functional involvement of IGF2BP2 in the progression of oral squamous cell carcinoma (OSCC) remains poorly understood. In this study, we showed that IGF2BP2 was upregulated in head and neck cancer, and high levels of IGF2BP2 were associated with poor survival. In in vitro experiments, IGF2BP2 promoted migration and invasion responses of OSCC cells. Moreover, we identified an IGF2BP2-regulated gene, EREG, which functioned as a modulator of OSCC invasion downstream of IGF2BP2. In addition, EREG expression triggered the epithelia-mesenchymal transition (EMT) in OSCC, as evidenced by the observation that knockdown of EREG weakened the induction of EMT mediated by IFG2BP2, and replenishment of EREG favored the EMT in IGF2BP2-depleted cells. Such IGF2BP2-regulated EREG expression, EMT, and cell invasion were dependent on the activation of FAK/Src signaling pathway. Collectively, these findings suggest that EREG, serving as a functional mediator of IGF2BP2-regulated EMT and cell invasion in oral cancer, may be implicated as a potential target for antimetastatic therapies.


Assuntos
Neoplasias Bucais , Proteínas de Ligação a RNA , Carcinoma de Células Escamosas de Cabeça e Pescoço , Humanos , Epirregulina , Transição Epitelial-Mesenquimal/genética , Neoplasias Bucais/genética , Proteínas de Ligação a RNA/genética , Carcinoma de Células Escamosas de Cabeça e Pescoço/genética , Regulação para Cima/genética
3.
Clin Cancer Res ; 29(20): 4021-4023, 2023 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-37594733

RESUMO

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.


Assuntos
Neoplasias do Colo , Neoplasias Colorretais , Neoplasias Retais , Humanos , Anfirregulina/genética , Epirregulina/genética , Multiômica , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Receptores ErbB/genética , Peptídeos e Proteínas de Sinalização Intercelular , Anticorpos , Inteligência Artificial
4.
J Pathol ; 261(1): 28-42, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37345534

RESUMO

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.


Assuntos
Neoplasias da Vesícula Biliar , Proteínas Proto-Oncogênicas p21(ras) , Humanos , Animais , Camundongos , Proteínas Proto-Oncogênicas p21(ras)/genética , Epirregulina/genética , Epirregulina/metabolismo , Neoplasias da Vesícula Biliar/genética , Neoplasias da Vesícula Biliar/metabolismo , Neoplasias da Vesícula Biliar/patologia , Regulação para Baixo , Linhagem Celular Tumoral , Proliferação de Células/genética , Proteínas Proto-Oncogênicas c-ets/genética , Receptores ErbB/genética , Receptores ErbB/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Regulação Neoplásica da Expressão Gênica , Proteínas de Ligação a DNA/genética , Fatores de Transcrição/genética
5.
Acta Biochim Pol ; 70(2): 389-393, 2023 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-37329522

RESUMO

MicroRNA-1179 (miRNA-1179) is an extensively studied tumor suppressor. however, the significance of miR-1179 in multiple myeloma has not been investigated previously. So, there is a need for research to find out about the significance of miR-1179 in multiple myeloma. However, current investigations have examined the significance of miRNA-1179 in multiple myeloma for the first time by targeting epiregulin (EREG). In this study, 26 multiple myeloma specimens and 16 healthy donor specimens were examined. Multiple myeloma cell lines (U266, RPMI-8226, KMS-11, JJN-3, and IM-9) were used. In this study, expression analysis, cell viability, colony formation assay, and transwell assay were carried out by standard methods. The outcomes revealed the downregulation of miRNA-1179 in multiple myeloma. Overexpression of miRNA-1179 promotes, while its inhibition suppresses, the survival ability and colony formation of the U266 multiple myeloma cells. Investigation of underlying mechanisms revealed apoptosis to be responsible for the tumour-suppressive effects of miRNA-1179. The proportion of apoptosis in U266 cells rose from 5.32% to 34.86% when miRNA-1179 was overexpressed. Additionally, it was discovered that miRNA-1179 directs its tumor-inhabiting activities toward EREG at the molecular level. While EREG knockdown was found to halt the proliferation of U266 cells, its overexpression could overcome the suppressive effects of miRNA-1179 on the survival ability, mobility, and invasion of the U266 cells. This research proves that miRNA-1179 can be used as a new treatment or drug for multiple myeloma.


Assuntos
Epirregulina , MicroRNAs , Mieloma Múltiplo , Humanos , Apoptose/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Epirregulina/metabolismo , MicroRNAs/genética , Mieloma Múltiplo/tratamento farmacológico
6.
Clin Cancer Res ; 29(20): 4153-4165, 2023 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-37363997

RESUMO

PURPOSE: High tumor production of the EGFR ligands, amphiregulin (AREG) and epiregulin (EREG), predicted benefit from anti-EGFR therapy for metastatic colorectal cancer (mCRC) in a retrospective analysis of clinical trial data. Here, AREG/EREG IHC was analyzed in a cohort of patients who received anti-EGFR therapy as part of routine care, including key clinical contexts not investigated in the previous analysis. EXPERIMENTAL DESIGN: Patients who received panitumumab or cetuximab ± chemotherapy for treatment of RAS wild-type mCRC at eight UK cancer centers were eligible. Archival formalin-fixed paraffin-embedded tumor tissue was analyzed for AREG and EREG IHC in six regional laboratories using previously developed artificial intelligence technologies. Primary endpoints were progression-free survival (PFS) and overall survival (OS). RESULTS: A total of 494 of 541 patients (91.3%) had adequate tissue for analysis. A total of 45 were excluded after central extended RAS testing, leaving 449 patients in the primary analysis population. After adjustment for additional prognostic factors, high AREG/EREG expression (n = 360; 80.2%) was associated with significantly prolonged PFS [median: 8.5 vs. 4.4 months; HR, 0.73; 95% confidence interval (CI), 0.56-0.95; P = 0.02] and OS [median: 16.4 vs. 8.9 months; HR, 0.66 95% CI, 0.50-0.86; P = 0.002]. The significant OS benefit was maintained among patients with right primary tumor location (PTL), those receiving cetuximab or panitumumab, those with an oxaliplatin- or irinotecan-based chemotherapy backbone, and those with tumor tissue obtained by biopsy or surgical resection. CONCLUSIONS: High tumor AREG/EREG expression was associated with superior survival outcomes from anti-EGFR therapy in mCRC, including in right PTL disease. AREG/EREG IHC assessment could aid therapeutic decisions in routine practice. See related commentary by Randon and Pietrantonio, p. 4021.


Assuntos
Neoplasias do Colo , Neoplasias Colorretais , Neoplasias Retais , Humanos , Anfirregulina/metabolismo , Epirregulina/metabolismo , Epirregulina/uso terapêutico , Cetuximab/uso terapêutico , Panitumumabe , Estudos Retrospectivos , Neoplasias Colorretais/patologia , Inteligência Artificial , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Neoplasias do Colo/tratamento farmacológico , Neoplasias Retais/tratamento farmacológico , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Receptores ErbB/metabolismo
7.
Atherosclerosis ; 375: 45-58, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37245426

RESUMO

BACKGROUND AND AIMS: Lactylation, a recently identified post-translational modification (PTM), plays a central role in the regulation of multiple physiological and pathological processes. Exercise is known to provide protection against cardiovascular disease. However, whether exercise-generated lactate changes lactylation and is involved in the exercise-induced attenuation of atherosclerotic cardiovascular disease (ASCVD) remains unclear. The purpose of this study was to investigate the effects and mechanisms of exercise-induced lactylation on ASCVD. METHODS AND RESULTS: Using the high-fat diet-induced apolipoprotein-deficient mouse model of ASCVD, we found that exercise training promoted Mecp2 lysine lactylation (Mecp2k271la); it also decreased the expression of vascular cell adhesion molecule 1 (Vcam-1), intercellular adhesion molecule 1 (Icam-1), monocyte chemoattractant protein 1 (Mcp-1), interleukin (IL)-1ß, IL-6, and increased the level of endothelial nitric oxide synthase (Enos) in the aortic tissue of mice. To explore the underlying mechanisms, mouse aortic endothelial cells (MAECs) were subjected to RNA-sequencing and CHIP-qPCR, which confirmed that Mecp2k271la repressed the expression of epiregulin (Ereg) by binding to its chromatin, demonstrating Ereg as a key downstream molecule for Mecp2k271la. Furthermore, Ereg altered the mitogen-activated protein kinase (MAPK) signalling pathway through regulating the phosphorylation level of epidermal growth factor receptor, thereby affecting the expression of Vcam-1, Icam-1, Mcp-1, IL-1ß, IL-6, and Enos in ECs, which in turn promoted the regression of atherosclerosis. In addition, increasing the level of Mecp2k271la by exogenous lactate administration in vivo also inhibits the expression of Ereg and the MAPK activity in ECs, resulting in repressed atherosclerotic progression. CONCLUSIONS: In summary, this study provides a mechanistic link between exercise and lactylation modification, offering new insight into the anti-atherosclerotic effects of exercise-induced PTM.


Assuntos
Aterosclerose , Doenças Cardiovasculares , Camundongos , Animais , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/farmacologia , Células Endoteliais/metabolismo , Molécula 1 de Adesão Intercelular/metabolismo , Interleucina-6/metabolismo , Epirregulina/metabolismo , Epirregulina/farmacologia , Molécula 1 de Adesão de Célula Vascular/metabolismo , Doenças Cardiovasculares/metabolismo , Aterosclerose/genética , Aterosclerose/prevenção & controle , Aterosclerose/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteína 2 de Ligação a Metil-CpG/genética , Proteína 2 de Ligação a Metil-CpG/metabolismo , Proteína 2 de Ligação a Metil-CpG/farmacologia
8.
Cells ; 12(5)2023 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-36899869

RESUMO

(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.


Assuntos
Ferroptose , Neoplasias de Cabeça e Pescoço , Humanos , Cetuximab/farmacologia , Epirregulina/genética , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Peptídeos e Proteínas de Sinalização Intercelular , Carcinoma de Células Escamosas de Cabeça e Pescoço/tratamento farmacológico , Microambiente Tumoral
9.
Reprod Sci ; 30(8): 2537-2546, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-36881337

RESUMO

The release of epidermal growth factor ligand epiregulin (EREG) by human ovarian granulosa cells, its direct action on basic ovarian cell functions, and interrelationships with gonadotropins were investigated. We examined (1) the ovarian production of EREG (the time-dependent accumulation of EREG in the medium incubated with human ovarian granulosa cells, and (2) the effect of the addition of EREG (0, 1, 10, and 100 ng.ml-1) given alone or in combination with FSH or LH (100 ng.ml-1) on basic granulosa cells functions. Viability, proliferation (accumulation of PCNA and cyclin B1) and apoptosis (accumulation of bax and caspase 3), the release of steroid hormones (progesterone, testosterone, and estradiol), and prostaglandin E2 (PGE2) were analyzed by using the Trypan blue exclusion test, quantitative immunocytochemistry, and ELISA. A significant time-dependent accumulation of EREG in a medium cultured with human granulosa cells with a peak at 3 and 4 days was observed. The addition of EREG alone increased cell viability, proliferation, progesterone, testosterone, and estradiol release, decreased apoptosis, bud did not affect PGE2 release. The addition of either FSH or LH alone increased cell viability, proliferation, progesterone, testosterone, estradiol, and PGE2 release and decreased apoptosis. Furthermore, both FSH and LH mostly promoted the stimulatory action of EREG on granulosa cell functions. These results demonstrated, that EREG produced by ovarian cells can be an autocrine/paracrine stimulator of human ovarian cell functions. Furthermore, they demonstrate the functional interrelationship between EREG and gonadotropins in the control of ovarian functions.


Assuntos
Dinoprostona , Progesterona , Feminino , Humanos , Progesterona/metabolismo , Epirregulina/metabolismo , Epirregulina/farmacologia , Dinoprostona/metabolismo , Proliferação de Células , Gonadotropinas/metabolismo , Células da Granulosa/metabolismo , Apoptose , Fator de Crescimento Epidérmico/farmacologia , Estradiol/farmacologia , Estradiol/metabolismo , Hormônio Foliculoestimulante/metabolismo , Testosterona/metabolismo , Células Cultivadas
11.
Int J Mol Sci ; 24(4)2023 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-36834576

RESUMO

Decidualization is necessary for the successful establishment of early pregnancy in rodents and humans. Disturbed decidualization results in recurrent implantation failure, recurrent spontaneous abortion, and preeclampsia. Tryptophan (Trp), one of the essential amino acids in humans, has a positive effect on mammalian pregnancy. Interleukin 4-induced gene 1 (IL4I1) is a recently identified enzyme that can metabolize L-Trp to activate aryl hydrocarbon receptor (AHR). Although IDO1-catalyzed kynurenine (Kyn) from Trp has been shown to enhance human in vitro decidualization via activating AHR, whether IL4I1-catalyzed metabolites of Trp are involved in human decidualization is still unknown. In our study, human chorionic gonadotropin stimulates IL4I1 expression and secretion from human endometrial epithelial cells through ornithine decarboxylase-induced putrescine production. Either IL4I1-catalyzed indole-3-pyruvic acid (I3P) or its metabolite indole-3-aldehyde (I3A) from Trp is able to induce human in vitro decidualization by activating AHR. As a target gene of AHR, Epiregulin induced by I3P and I3A promotes human in vitro decidualization. Our study indicates that IL4I1-catalyzed metabolites from Trp can enhance human in vitro decidualization through AHR-Epiregulin pathway.


Assuntos
Interleucina-4 , Receptores de Hidrocarboneto Arílico , Animais , Humanos , Epirregulina , Receptores de Hidrocarboneto Arílico/metabolismo , Triptofano/metabolismo , Cinurenina/metabolismo , Gonadotropina Coriônica , Mamíferos/metabolismo , L-Aminoácido Oxidase
12.
JCI Insight ; 8(6)2023 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-36821371

RESUMO

Epithelial organoids derived from intestinal tissue, called enteroids, recapitulate many aspects of the organ in vitro and can be used for biological discovery, personalized medicine, and drug development. Here, we interrogated the cell signaling environment within the developing human intestine to identify niche cues that may be important for epithelial development and homeostasis. We identified an EGF family member, EPIREGULIN (EREG), which is robustly expressed in the developing human crypt. Enteroids generated from the developing human intestine grown in standard culture conditions, which contain EGF, are dominated by stem and progenitor cells and feature little differentiation and no spatial organization. Our results demonstrate that EREG can replace EGF in vitro, and EREG leads to spatially resolved enteroids that feature budded and proliferative crypt domains and a differentiated villus-like central lumen. Multiomic (transcriptome plus epigenome) profiling of native crypts, EGF-grown enteroids, and EREG-grown enteroids showed that EGF enteroids have an altered chromatin landscape that is dependent on EGF concentration, downregulate the master intestinal transcription factor CDX2, and ectopically express stomach genes, a phenomenon that is reversible. This is in contrast to EREG-grown enteroids, which remain intestine like in culture. Thus, EREG creates a homeostatic intestinal niche in vitro, enabling interrogation of stem cell function, cellular differentiation, and disease modeling.


Assuntos
Fator de Crescimento Epidérmico , Intestinos , Humanos , Epirregulina , Mucosa Intestinal , Diferenciação Celular
13.
J Transl Med ; 21(1): 28, 2023 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-36647156

RESUMO

BACKGROUND: Glioma is the most prevalent primary tumor of the central nervous system. Glioblastoma multiforme (GBM) is the most malignant form of glioma with an extremely poor prognosis. A novel, regulated cell death form of copper-induced cell death called "cuproptosis" provides a new prospect for cancer treatment by regulating cuproptosis. METHODS: Data from bulk RNA sequencing (RNA-seq) analysis (The Cancer Genome Atlas cohort and Chinese Glioma Genome Atlas cohort) and single cell RNA-seq (scRNA-seq) analysis were integrated to reveal their relationships. A scoring system was constructed according to the cuproptosis-related gene expression, and core genes were experimentally verified using real-time quantitative reverse transcription polymerase chain reaction (qRT-PCR), Western blot (WB), immunohistochemistry (IHC), and immunofluorescence (IF). Moreover, cell counting kit-8 (CCK8), colony formation, 5-ethynyl-2'-deoxyuridine (EdU) incorporation, transwell, and flow cytometry cell cycle were performed to evaluate cell proliferation, invasion, and migration. RESULTS: The Cuproptosis Activation Scoring (CuAS) Model has stable and independent prognostic efficacy, as verified by two CGGA datasets. Epiregulin (EREG), the gene of the model has the most contributions in the principal component analysis (PCA), is an onco-immunological gene that can affect immunity by influencing the expression of programmed death-ligand 1 (PD-L1) and mediate the process of cuproptosis by influencing the expression of ferredoxin 1 (FDX1). Single cell transcriptome analysis revealed that high CuAS GBM cells are found in vascular endothelial growth factor A (VEGFA) + malignant cells. Oligodendrocyte transcription factor 1 (OLIG1) + malignant is the original clone, and VEGF and CD99 are the differential pathways of specific cell communication between the high and low CuAS groups. This was also demonstrated by immunofluorescence in the tissue sections. Furthermore, CuAS has therapeutic potential for immunotherapy, and we predict that many drugs (methotrexate, NU7441, KU -0063794, GDC-0941, cabozantinib, and NVP-BEZ235) may be used in patients with high CuAS. CONCLUSION: EREG is the core onco-immunological biomarker of CuAS and modulates the cross-talk between VEGF and CD99 signaling in glioblastoma, and CuAS may provide support for immunotherapy and chemotherapy.


Assuntos
Apoptose , Epirregulina , Glioblastoma , Glioma , Humanos , Antígeno 12E7 , Biomarcadores , Glioblastoma/genética , Glioma/genética , Peptídeos e Proteínas de Sinalização Intercelular , Fator A de Crescimento do Endotélio Vascular
14.
Mol Biotechnol ; 65(9): 1453-1464, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-36645579

RESUMO

It is well established that circular RNAs (circRNAs) play a role in tumor initiation and tumorigenesis. The goal of this study was to reveal the detailed functions and regulatory mechanisms of circ_0000078 in cervical cancer (CC). Circ_0000078, miR-205-5p, and epiregulin (EREG) mRNA expression levels were examined using RT-qPCR. Western blotting was performed to quantify EREG protein. Cell proliferation, apoptosis, migration, and invasion were examined by performing CCK-8, caspase 3 activity, wound healing, and transwell assays, respectively. The effect of circ_0000078 on tumor growth in vivo was confirmed in a xenograft model. The putative relationship between miR-205-5p and circ_0000078 or EREG, as predicted by bioinformatics analysis, was evaluated by dual-luciferase and RNA immunoprecipitation assays. Aberrant downregulation of circ_0000078 and EREG as well as upregulation of miR-205-5p were observed in cervical tumor samples and cancer cells. Ectopic expression of circ _0000078 not only restrained cancer cell growth, survival, migration, and invasiveness, but also decelerated tumor formation and development in a mouse model. miR-205-5p, acts as a target of circ_0000078 and directly binds to EREG to repress its expression. Overexpression of miR-205-5p reversed the inhibitory effects of circ_0000078 upregulation on cancer cell behavior and also partially abolished the anti-cancer effects of EREG upregulation in vitro. Circ_0000078 inhibits the growth of cancer by interfering with the miR-205-5p/EREG network, acting as a tumor suppressor in CC. These results provide a better understanding of the pathogenesis of this disease.


Assuntos
MicroRNAs , Neoplasias do Colo do Útero , Animais , Feminino , Humanos , Camundongos , Linhagem Celular Tumoral , Proliferação de Células/genética , Regulação para Baixo , Epirregulina , Peptídeos e Proteínas de Sinalização Intercelular , MicroRNAs/genética , Neoplasias do Colo do Útero/genética , Neoplasias do Colo do Útero/patologia
15.
Cancer Res Commun ; 2(9): 1061-1074, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-36506869

RESUMO

Preclinical and clinical studies have evidenced that effective targeted therapy treatment against receptor tyrosine kinases (RTKs) in different solid tumor paradigms is predicated on simultaneous inhibition of both the PI3K and MEK intracellular signaling pathways. Indeed, re-activation of either pathway results in resistance to these therapies. Recently, oncogenic phosphatase SHP2 inhibitors have been developed with some now reaching clinical trials. To expand on possible indications for SHP099, we screened over 800 cancer cell lines covering over 25 subsets of cancer. We found HNSCC was the most sensitive adult subtype of cancer to SHP099. We found that, in addition to the MEK pathway, SHP2 inhibition blocks the PI3K pathway in sensitive HNSCC, resulting in downregulation of mTORC signaling and anti-tumor effects across several HNSCC mouse models, including an HPV+ patient-derived xenograft (PDX). Importantly, we found low levels of the RTK ligand epiregulin identified HNSCCs that were sensitive to SHP2 inhibitor, and, adding exogenous epiregulin mitigated SHP099 efficacy. Mechanistically, epiregulin maintained SHP2-GAB1 complexes in the presence of SHP2 inhibition, preventing downregulation of the MEK and PI3K pathways. We demonstrate HNSCCs were highly dependent on GAB1 for their survival and knockdown of GAB1 is sufficient to block the ability of epiregulin to rescue MEK and PI3K signaling. These data connect the sensitivity of HNSCC to SHP2 inhibitors and to a broad reliance on GAB1-SHP2, revealing an important and druggable signaling axis. Overall, SHP2 inhibitors are being heavily developed and may have activity in HNSCCs, and in particular those with low levels of epiregulin.


Assuntos
Neoplasias de Cabeça e Pescoço , Fosfatidilinositol 3-Quinases , Animais , Camundongos , Humanos , Fosfatidilinositol 3-Quinases/metabolismo , Carcinoma de Células Escamosas de Cabeça e Pescoço/tratamento farmacológico , Epirregulina/metabolismo , Inibidores Enzimáticos/farmacologia , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo
16.
Oncogene ; 41(45): 4941-4959, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36202915

RESUMO

The tumor microenvironment (TME) represents a milieu enabling cancer cells to develop malignant properties, while concerted interactions between cancer and stromal cells frequently shape an "activated/reprogramed" niche to accelerate pathological progression. Here we report that a soluble factor epiregulin (EREG) is produced by senescent stromal cells, which non-cell-autonomously develop the senescence-associated secretory phenotype (SASP) upon DNA damage. Genotoxicity triggers EREG expression by engaging NF-κB and C/EBP, a process supported by elevated chromatin accessibility and increased histone acetylation. Stromal EREG reprograms the expression profile of recipient neoplastic cells in a paracrine manner, causing upregulation of MARCHF4, a membrane-bound E3 ubiquitin ligase involved in malignant progression, specifically drug resistance. A combinational strategy that empowers EREG-specific targeting in treatment-damaged TME significantly promotes cancer therapeutic efficacy in preclinical trials, achieving response indices superior to those of solely targeting cancer cells. In clinical oncology, EREG is expressed in tumor stroma and handily measurable in circulating blood of cancer patients post-chemotherapy. This study establishes EREG as both a targetable SASP factor and a new noninvasive biomarker of treatment-damaged TME, thus disclosing its substantial value in translational medicine.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Microambiente Tumoral , Epirregulina/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular , NF-kappa B
17.
Eur J Med Res ; 27(1): 208, 2022 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-36271425

RESUMO

BACKGROUND: The recovery from cardiac surgery involves resolving inflammation and remodeling with significant connective tissue turnover. Dynamics of smoldering inflammation and injury (white blood cells, platelets, CRP, IL-8, IL-6), vascular inflammation (IL-15, VEGF, RANTES), connective tissue remodeling (tenascin, MMP-9), cardiac injury and remodeling (YKL-40), and vascular remodeling (epiregulin, MCP-1, VEGF) were assessed up to 3 months after cardiac surgery. We hypothesize that at 3 months, studied markers will return to pre-surgical levels. METHODS: Patients (n = 139) scheduled for non-emergent heart surgery were included, except for patients with pre-existing immunological aberrancies. Blood was collected before surgery(tbaseline), 24 h later(t24h) after the first sample, 7 days(t7d), and 3 months(t3m) after tbaseline. Serum markers were measured via multiplex or ELISA. Electronic medical records (EMR) were used to extract demographical, pre-existing conditions and clinical data. Disposition (discharge home, discharge to facility, death, re-admission) was determined at 28 days and 3 months from admission. RESULTS: Not all inflammatory markers returned to baseline (CRP↑↑, leukocytosis, thrombocytosis, IL-8↓, IL-6↓). Tenascin and YKL-40 levels remained elevated even at t3m. YKL-40 serum levels were significantly elevated at t24h and t7d while normalized at t3m. VEGF returned to the baseline, yet MCP-1 remained elevated at 3 months. CCL28 increased at 3 months, while RANTES and IL-15 declined at the same time. Disposition at discharge was determined by serum MMP-9, while YKL-40 correlated with duration of surgery and APACHE II24h. CONCLUSIONS: The data demonstrated an ongoing extracellular matrix turnover at 3 months, while acute inflammation and vascular remodeling resolved only partially.


Assuntos
Procedimentos Cirúrgicos Cardíacos , Metaloproteinase 9 da Matriz , Humanos , Tenascina , Interleucina-15 , Epirregulina , Proteína 1 Semelhante à Quitinase-3 , Interleucina-6 , Fator A de Crescimento do Endotélio Vascular , Remodelação Vascular , Interleucina-8 , Biomarcadores , Inflamação , Procedimentos Cirúrgicos Cardíacos/efeitos adversos
18.
Cell Death Dis ; 13(10): 882, 2022 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-36266264

RESUMO

Superoxide dismutase 1 (SOD1) modulates intestinal barrier integrity and intestinal homeostasis as an antioxidant enzyme. Intestinal homeostasis is maintained by the intestinal stem cells (ISCs). However, whether and how SOD1 regulates ISCs is unknown. In this study, we established intestinal organoids from tamoxifen-inducible intestinal epithelial cell-specific Sod1 knockout (Sod1f/f; Vil-creERT2) mice. We found that loss of Sod1 in organoids suppressed the proliferation and survival of cells and Lgr5 gene expression. SOD1 is known for nearly half a century for its canonical role as an antioxidant enzyme. We identified its enzyme-independent function in ISC: inhibition of SOD1 enzymatic activity had no impact on organoid growth, and enzymatically inactive Sod1 mutants could completely rescue the growth defects of Sod1 deficient organoids, suggesting that SOD1-mediated ISC growth is independent of its enzymatic activity. Moreover, Sod1 deficiency did not affect the ROS levels of the organoid, but induced the elevated WNT signaling and excessive Paneth cell differentiation, which mediates the occurrence of growth defects in Sod1 deficient organoids. In vivo, epithelial Sod1 loss induced a higher incidence of apoptosis in the stem cell regions and increased Paneth cell numbers, accompanied by enhanced expression of EGFR ligand Epiregulin (EREG) in the stromal tissue, which may compensate for Sod1 loss and maintain intestinal structure in vivo. Totally, our results show a novel enzyme-independent function of SOD1 in ISC growth under homeostasis.


Assuntos
Neoplasias Intestinais , Superóxido Dismutase , Camundongos , Animais , Superóxido Dismutase-1/genética , Superóxido Dismutase-1/metabolismo , Epirregulina/metabolismo , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Ligantes , Antioxidantes/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Células-Tronco/metabolismo , Celulas de Paneth/metabolismo , Organoides/metabolismo , Neoplasias Intestinais/metabolismo , Receptores ErbB/metabolismo , Tamoxifeno/farmacologia , Mucosa Intestinal/metabolismo , Proliferação de Células
19.
Mol Cancer ; 21(1): 181, 2022 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-36117171

RESUMO

BACKGROUND: Although, micropeptides encoded by non-coding RNA have been shown to have an important role in a variety of tumors processes, there have been no reports on micropeptide in renal cell carcinoma (RCC). Based on the micropeptide MIAC (micropeptide inhibiting actin cytoskeleton) discovered and named in the previous work, this study screened its tumor spectrum, and explored its mechanism of action and potential diagnosis and treatment value in the occurrence and development of renal carcinoma. METHODS: The clinical significance of MIAC in RCC was explored by bioinformatics analysis through high-throughput RNA-seq data from 530 patients with kidney renal clear cell carcinoma (KIRC) in the TCGA database, and the detection of clinical samples of 70 cases of kidney cancer. In vitro and in vivo experiments to determine the role of MIAC in renal carcinoma cell growth and metastasis; High-throughput transcriptomics, western blotting, immunoprecipitation, molecular docking, affinity experiments, and Streptavidin pulldown experiments identify MIAC direct binding protein and key regulatory pathways. RESULTS: The analysis of 600 renal carcinoma samples from different sources revealed that the expression level of MIAC is significantly decreased, and corelated with the prognosis and clinical stage of tumors in patients with renal carcinoma. Overexpression of MIAC in renal carcinoma cells can significantly inhibit the proliferation and migration ability, promote apoptosis of renal carcinoma cells, and affect the distribution of cells at various stages. After knocking down MIAC, the trend is reversed. In vivo experiments have found that MIAC overexpression inhibit the growth and metastasis of RCC, while the synthetized MIAC peptides can significantly inhibit the occurrence and development of RCC in vitro and in vivo. Further mechanistic studies have demonstrated that MIAC directly bind to AQP2 protein, inhibit EREG/EGFR expression and activate downstream pathways PI3K/AKT and MAPK to achieve anti-tumor effects. CONCLUSIONS: This study revealed for the first time the tumor suppressor potential of the lncRNA-encoded micropeptide MIAC in RCC, which inhibits the activation of the EREG/EGFR signaling pathway by direct binding to AQP2 protein, thereby inhibiting renal carcinoma progression and metastasis. This result emphasizes that the micropeptide MIAC can provide a new strategy for the diagnosis and treatment of RCC.


Assuntos
Carcinoma de Células Renais , Neoplasias Renais , RNA Longo não Codificante , Aquaporina 2/genética , Aquaporina 2/metabolismo , Carcinoma de Células Renais/patologia , Linhagem Celular Tumoral , Epirregulina , Receptores ErbB/genética , Receptores ErbB/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Renais/patologia , Simulação de Acoplamento Molecular , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Longo não Codificante/genética , Transdução de Sinais , Estreptavidina/genética , Estreptavidina/metabolismo , Estreptavidina/uso terapêutico
20.
Anticancer Res ; 42(8): 3873-3878, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35896250

RESUMO

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.


Assuntos
Neoplasias Gástricas , Epirregulina/genética , Expressão Gênica , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , RNA Mensageiro/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/cirurgia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...