FIGURE 2 - available via license: CC BY
Content may be subject to copyright.
| HB-EGF activates EGFR in arsenic transformed cells. (A) As-T cells and A549 cells were treated with 10 µg/mL CAM197 (an HB-EGF inhibitor). After 48 h, p-EGFR(Y1068) and EGFR expression were analyzed via Western blotting. CAM197, cross-reacting material 197; p-EGFR (Y1068), activated EGFR at phospho-Tyr1068. *p < 0.05, compared with As-T/PBS. # p < 0.05, compared with A549/PBS. (B) B2B cells were treated with or without 1 µM NaAsO 2 and 10 µg/mL CRM197, as indicated for 48 h; and levels of p-EGFR protein were determined via Western blotting. *p < 0.05, compared with B2B/PBS. (C) B2B cells were transfected with siHB-NC and siHB-EGF as per the manufacturer's instructions. Twelve hours after transfection, As-T cells were treated with or without NaAsO 2, as indicated for 36 h, and expression of HB-EGF and p-EGFR was analyzed by Western blotting. The data are presented as the mean±SD. * ,# p < 0.05, ***p < 0.001.

| HB-EGF activates EGFR in arsenic transformed cells. (A) As-T cells and A549 cells were treated with 10 µg/mL CAM197 (an HB-EGF inhibitor). After 48 h, p-EGFR(Y1068) and EGFR expression were analyzed via Western blotting. CAM197, cross-reacting material 197; p-EGFR (Y1068), activated EGFR at phospho-Tyr1068. *p < 0.05, compared with As-T/PBS. # p < 0.05, compared with A549/PBS. (B) B2B cells were treated with or without 1 µM NaAsO 2 and 10 µg/mL CRM197, as indicated for 48 h; and levels of p-EGFR protein were determined via Western blotting. *p < 0.05, compared with B2B/PBS. (C) B2B cells were transfected with siHB-NC and siHB-EGF as per the manufacturer's instructions. Twelve hours after transfection, As-T cells were treated with or without NaAsO 2, as indicated for 36 h, and expression of HB-EGF and p-EGFR was analyzed by Western blotting. The data are presented as the mean±SD. * ,# p < 0.05, ***p < 0.001.

Source publication
Article
Full-text available
Arsenic was recently identified as a pollutant that is a major cause of lung cancer. Since heparin-binding EGF-like growth factor (HB-EGF) was reported to be a promising therapeutic target for lung cancer, we investigated the role and mechanism of HB-EGF during arsenic-induced carcinogenesis and development of lung cancer. HB-EGF expression were up...

Contexts in source publication

Context 1
... in lung cancer cells, arsenic exposure upregulates the levels of heparin binding-EGF (an EGFR ligand) and stimulates an activating phosphorylation of EGFR (p-EGFR at Tyr 1173). To test whether arsenic activates EGFR through HB-EGF, As-T and A549 cells were first treated with 10 µg/mL of CRM197 (an HB-EGF inhibitor; as indicated in Figure 2A) for 48 h, and p-EGFR and EGFR protein levels were evaluated by Western blotting. Under these conditions, CRM197 inhibited p-EGFR protein levels, in both As-T and A549 cells, but it did not affect EGFR protein level in As-T cells. ...
Context 2
... these conditions, CRM197 inhibited p-EGFR protein levels, in both As-T and A549 cells, but it did not affect EGFR protein level in As-T cells. When B2B cells were treated for 48 h with 1 µM NaAsO 2 and 10 µg/mL CRM197, as indicated in Figure 2B, the arsenic-induced increases in p-EGFR expression were reversed, suggesting arsenic activated p-EGFR via the HB-EGF ligand. Confirmatory results (shown in Figure 2C) showed transfecting an siHB-EGF similarly inhibited p-EGFR protein levels. ...
Context 3
... B2B cells were treated for 48 h with 1 µM NaAsO 2 and 10 µg/mL CRM197, as indicated in Figure 2B, the arsenic-induced increases in p-EGFR expression were reversed, suggesting arsenic activated p-EGFR via the HB-EGF ligand. Confirmatory results (shown in Figure 2C) showed transfecting an siHB-EGF similarly inhibited p-EGFR protein levels. ...

Similar publications

Article
Full-text available
Oral mucositis refers to lesions of the oral mucosa observed in patients with cancer being treated with radiation with or without chemotherapy, and can significantly affect quality of life. There is a large unmet medical need to prevent oral mucositis that can occur with radiation either alone or in combination with chemotherapy. We investigated th...

Citations

... CD68+ TAMs were also an adverse prognostic factor that predicted a lower OS in ES, and worse progression-free survival (PFS) in LMS [33,51]. Furthermore, higher levels of CD68+ TAMs were associated with a poorer OS in myxoid liposarcoma that promoted invasiveness through the heparin-binding EGF-like growth factor (HB-EGF)-EGFR-PI3K/Akt pathway [65]. A study of almost 200 STSs indicated that CD68+ macrophages were independently associated with an increased risk of LR, regardless of margin status, age, sex, or the number of CD20+ B cells [61]. ...
Article
Full-text available
Simple Summary Soft tissue and bone sarcomas belong to a group of rare and malignant tumors. The treatment of these tumors is very complex and depends on their specific subtypes. Therefore, there is a great need to develop novel therapeutic options for patients with sarcomas. One such option may be to target tumor-associated macrophages (TAMs), which are involved in immunosuppression during tumor growth. High levels of TAMs are widely recognized as a poor prognostic factor in many tumors, including sarcomas, making them a promising target for future targeted therapies. In this review, we highlight the role of TAMs in the microenvironment of sarcomas, along with their clinical relevance, potential targetable markers on their surface, and the molecular pathways involved. We also discuss currently ongoing clinical trials with TAMs. Abstract Sarcomas are a heterogeneous group of malignant mesenchymal tumors, including soft tissue and bone sarcomas. Macrophages in the tumor microenvironment, involved in immunosuppression and leading to tumor development, are called tumor-associated macrophages (TAMs). TAMs are very important in modulating the microenvironment of sarcomas by expressing specific markers and secreting factors that influence immune and tumor cells. They are involved in many signaling pathways, such as p-STAT3/p-Erk1/2, PI3K/Akt, JAK/MAPK, and JAK/STAT3. TAMs also significantly impact the clinical outcomes of patients suffering from sarcomas and are mainly related to poor overall survival rates among bone and soft tissue sarcomas, for example, chondrosarcoma, osteosarcoma, liposarcoma, synovial sarcoma, and undifferentiated pleomorphic sarcoma. This review summarizes the current knowledge on TAMs in sarcomas, focusing on specific markers on sarcoma cells, cell–cell interactions, and the possibly involved molecular pathways. Furthermore, we discuss the clinical significance of macrophages in sarcomas as a potential target for new therapies, presenting clinical relevance, possible new treatment options, and ongoing clinical trials using TAMs in sarcoma treatment.
... Besides acting as a key enzyme of glycolysis, PKM2 also acts as a co-transcription factor to regulate the expression of GLUT1 and LDHA, contributing to glycolysis in various cells [18,19]. Wang et al. [20] found that the expression of PKM2 was significantly increased in arsenic-induced lung epithelial cell lines, but the role of PKM2 in the Warburg effect and cell proliferation induced by arsenic needs to be further explored. ...
Article
Full-text available
This study aimed to investigate the potential role of pyruvate kinase M2 (PKM2) and extracellular regulated protein kinase (ERK) in arsenic-induced cell proliferation. L-02 cells were treated with 0.2 and 0.4 μmol/L As3+, glycolysis inhibitor (2-deoxy-D-glucose,2-DG), ERK inhibitor [1,4-diamino-2,3-dicyano-1,4-bis(2-aminophenylthio)-butadiene, U0126] or transfected with PKM2 plasmid. Cell viability, proliferation, lactate acid production, and glucose intake capacity were determined by CCK-8 assay, EdU assay, lactic acid kit and 2-deoxy-2-[(7-nitro-2,1,3-benzoxadiazol-4-yl) amino]-D-glucose (2-NBDG) uptake kit, respectively. Also, levels of PKM2, phospho-PKM2S37, glucose transporter protein 1 (GLUT1), lactate dehydrogenase A (LDHA), ERK, and phospho-ERK were detected using Western blot and the subcellular localization of PKM2 in L-02 cells was detected by immunocytochemistry (ICC). Treatment with 0.2 and 0.4 μmol/L As3+ for 48 h increased the viability and proliferation of L-02 cells, the proportion of 2-NBDG+ cell and lactic acid in the culture medium, and GLUT1, LDHA, PKM2, phospho-PKM2S37, and phospho-ERK levels and PKM2 in nucleus. Compared with the 0.2 μmol/L As3+ treatment group, the lactic acid in the culture medium, cell proliferation and cell viability, and the expression of GLUT1 and LDHA were reduced in the group co-treated with siRNA-PKM2 and arsenic or in the group co-treated with U0126. Moreover, the arsenic-increased phospho-PKM2S37/PKM2 was decreased by U0126. Therefore, ERK/PKM2 plays a key role in the Warburg effect and proliferation of L-02 cells induced by arsenic, and also might be involved in arsenic-induced upregulation of GLUT1 and LDHA. This study provides a theoretical basis for further elucidating the carcinogenic mechanism of arsenic.
... It has been reported that EGFR activation is induced by arsenic exposure [20][21][22], and long-term exposure to arsenic at low doses results in increased EGFR expression due to inducing TGFα [11]. We also found that chronic arsenic exposure activates the EGFR/p-ERK/HIF-1α pathway [69]. Consistent with the studies above, we found that long-term exposure to arsenic dramatically upregulated EGFR expression levels, activated PKM2, ERK1/2, and p65 of the NF-κB subunit, and promoted HIF-1α expression, indicating that long-term exposure to arsenic induces the expression of EGFR and its downstream targets. ...
Article
Full-text available
Background: Arsenic is a well-known carcinogen inducing lung, skin, bladder, and liver cancer. Abnormal epidermal growth factor receptor (EGFR) expression is common in lung cancer; it is involved in cancer initiation, development, metastasis, and treatment resistance. However, the underlying mechanism for arsenic-inducing EGFR upregulation remains unclear. Methods: RT-PCR and immunoblotting assays were used to detect the levels of miR-218-5p and EGFR expression. The Luciferase assay was used to test the transcriptional activity of EGFR mediated by miR-218-5p. Cell proliferation, colony formation, wound healing, migration assays, tube formation assays, and tumor growth assays were used to study the function of miR-218-5p/EGFR signaling. Results: EGFR and miR-218-5p were dramatically upregulated and downregulated in arsenic-induced transformed (As-T) cells, respectively. MiR-218-5p acted as a tumor suppressor to inhibit cell proliferation, migration, colony formation, tube formation, tumor growth, and angiogenesis. Furthermore, miR-218-5p directly targeted EGFR by binding to its 3'-untranslated region (UTR). Finally, miR-218-5p exerted its antitumor effect by inhibiting its direct target, EGFR. Conclusion: Our study highlights the vital role of the miR-218-5p/EGFR signaling pathway in arsenic-induced carcinogenesis and angiogenesis, which may be helpful for the treatment of lung cancer induced by chronic arsenic exposure in the future.
... Therefore, our data suggested that ARAP1 could interact with CIN85 in high glucose-induced HRMCs and diabetic db/db mouse glomeruli. EGFR activation promotes dimer PKM2 expression and nuclear translocation to activate HIF-1α, contributing to abnormal glycolysis and tumor cell proliferation (Yang et al., 2011;Yang et al., 2018;Wang et al., 2020). EGFR activation promotes HIF-1α activation and ECM accumulation in mesangial cells in response to high glucose, which finally resulted in matrix upregulation and glomerular fibrosis (Uttarwar et al., 2011;Li et al., 2015), but the exact regulatory mechanism of ARAP1 on EGFR activation and PKM2/HIF-1ɑ pathway in mesangial cells has not been reported. ...
Article
Full-text available
Objectives: Dimeric pyruvate kinase (PK) M2 (PKM2) plays an important role in promoting the accumulation of hypoxia-inducible factor (HIF)-1α, mediating aberrant glycolysis and inducing fibrosis in diabetic kidney disease (DKD). The aim of this work was to dissect a novel regulatory mechanism of Yin and Yang 1 (YY1) on lncRNA-ARAP1-AS2/ARAP1 to regulate EGFR/PKM2/HIF-1α pathway and glycolysis in DKD. Materials and methods: We used adeno-associated virus (AAV)-ARAP1 shRNA to knocked down ARAP1 in diabetic mice and overexpressed or knocked down YY1, ARAP1-AS2 and ARAP1 expression in human glomerular mesangial cells. Gene levels were assessed by Western blotting, RT-qPCR, immunofluorescence staining and immunohistochemistry. Molecular interactions were determined by RNA pull-down, co-immunoprecipitation, ubiquitination assay and dual-luciferase reporter analysis. Results: YY1, ARAP1-AS2, ARAP1, HIF-1α, glycolysis and fibrosis genes expressions were upregulated and ARAP1 knockdown could inhibit dimeric PKM2 expression and partly restore tetrameric PKM2 formation, while downregulate HIF-1α accumulation and aberrant glycolysis and fibrosis in in-vivo and in-vitro DKD models. ARAP1 knockdown attenuates renal injury and renal dysfunction in diabetic mice. ARAP1 maintains EGFR overactivation in-vivo and in-vitro DKD models. Mechanistically, YY1 transcriptionally upregulates ARAP1-AS2 and indirectly regulates ARAP1 and subsequently promotes EGFR activation, HIF-1α accumulation and aberrant glycolysis and fibrosis. Conclusion: Our results first highlight the role of the novel regulatory mechanism of YY1 on ARAP1-AS2 and ARAP1 in promoting aberrant glycolysis and fibrosis by EGFR/PKM2/HIF-1α pathway in DKD and provide potential therapeutic strategies for DKD treatments.
... Some scholars propose that arsenic may induce tumorigenesis and development by participating in cell proliferation, apoptosis, oxidative stress, DNA damage, and chromosomal aberrations (Mar Wai et al. 2019;Medda et al. 2021;Wang et al. 2020;Wu et al. 2019;Zang et al. 2020). As an essential tumor suppressor regulatory gene, the p53 regulatory network is closely related to the known mechanisms of arsenic toxicity. ...
Article
Full-text available
As a class I carcinogen, arsenic has been reported to cause diseases accompanied by circRNAs regulating proliferation and apoptosis at the molecular level, but whether circP50 (circBase ID: hsa_circ_0008012) does the same has not been demonstrated. The aim of this study is to provide the basis for anti-lung cancer mechanism research, by studying the expression of circP50 under arsenic-induced conditions, and the effect and mechanism on the proliferation and apoptosis of A549 cells based on the circP50 knockdown models. To explore whether the circP50 is responsive to arsenic exposure, the qRT-PCR was applied to discover that the relative expression of circP50 in A549 cells increased only with increasing NaAsO2 dose and independent of its metabolites. We further determined the mechanism of circP50 by establishing circP50 knockdown models. The results of cell viability and EdU assays indicated the proliferation of A549 cells. According to the western blotting, phosphorylation of p53 at Ser15, Ser376, and Ser392 and acetylation of p53 at Lys370 and Lys382 were inhibited, resulting in the deficiency of p53 expression. Subsequently, the expression of genes downstream of p53 was reduced, including p21, PUMA, Caspase3, and Bcl-xS. Furthermore, the expressions of IKB-α, p65, and p50 decreased, but C-myc expression did not change significantly, referring to the NF-κB pathway was not dominant. The results suggest that circP50 mainly functions through the p53 pathway to mediate apoptosis in response to arsenic exposure.
Article
Full-text available
Background Angiogenesis stands as a pivotal hallmark in lung adenocarcinoma (LUAD), intricately shaping the tumor microenvironment (TME) and influencing LUAD progression. It emerges as a promising therapeutic target for LUAD, affecting patients’ prognosis. However, its role in TME, LUAD prognosis, and its clinical applicability remain shrouded in mystery. Methods We employed integrated single-cell and bulk transcriptome sequencing to unravel the heterogeneity of angiogenesis within LUAD cells. Through “consensus clustering”, we delineated distinct angiogenic clusters and deciphered their TME features. “Monocle2” was used to unravel divergent trajectories within malignant cell subpopulations of LUAD. Additionally, regulon submodules and specific cellular communication patterns of cells in different angiogenic states were analyzed by “pyscenic” and “Cellchat” algorithms. The “univariate Cox” and “LASSO” algorithms were applied to build angiogenic prognostic models. Immunohistochemistry (IHC) on clinical samples validated the role of model factors in LUAD angiogenesis. We utilized CTRP 2.0 and PRISM databases for pinpointing sensitive drugs against lung adenocarcinoma. Results Two clusters for the activation of angiogenesis were identified, with Cluster 1 showing a poor prognosis and a pro-cancerous TME. Three differentiated states of malignant epithelial LUAD cells were identified, which had different degrees of angiogenic activation, were regulated by three different regulon submodules, and had completely different crosstalk from other cells in TME. The experiments validate that SLC2A1 promotes angiogenesis in LUAD. ARS (Angiogenesis related score) had a high prognostic value; low ARSs showed immunotherapy benefits, whereas high ARSs were sensitive to 15 chemotherapeutic agents. Conclusion The assessment of angiogenic clusters helps to determine the prognostic and TME characteristics of LUAD. Angiogenic prognostic models can be used to assess the prognosis, immunotherapeutic response, and chemotherapeutic drug sensitivity of LUAD.
Article
Full-text available
Background Despite the high prevalence of lung cancer, with a five-year survival rate of only 23%, the underlying molecular mechanisms of non-small cell lung cancer (NSCLC) remain unknown. There is a great need to identify reliable candidate biomarker genes for early diagnosis and targeted therapeutic strategies to prevent cancer progression. Methods In this study, four datasets obtained from the Gene Expression Omnibus were evaluated for NSCLC- associated differentially expressed genes (DEGs) using bioinformatics analysis. About 10 common significant DEGs were shortlisted based on their p-value and FDR (DOCK4, ID2, SASH1, NPR1, GJA4, TBX2, CD24, HBEGF, GATA3, and DDR1). The expression of significant genes was validated using experimental data obtained from TCGA and the Human Protein Atlas database. The human proteomic data for post- translational modifications was used to interpret the mutations in these genes. Results Validation of DEGs revealed a significant difference in the expression of hub genes in normal and tumor tissues. Mutation analysis revealed 22.69%, 48.95%, and 47.21% sequence predicted disordered regions of DOCK4, GJA4, and HBEGF, respectively. The gene-gene and drug-gene network analysis revealed important interactions between genes and chemicals suggesting they could act as probable drug targets. The system-level network showed important interactions between these genes, and the drug interaction network showed that these genes are affected by several types of chemicals that could serve as potential drug targets. Conclusions The study demonstrates the importance of systemic genetics in identifying potential drug- targeted therapies for NSCLC. The integrative system- level approach should contribute to a better understanding of disease etiology and may accelerate drug discovery for many cancer types.
Chapter
Tungsten is an emerging contaminant in the environment. Research has demonstrated that humans are exposed to high levels of tungsten in certain settings, primarily due to increased use of tungsten in industrial applications. However, our understanding of the potential human health risks of tungsten exposure is still limited. An important point we have learned about the toxicity profile of tungsten is that it is complex because tungsten can often augment the effects of other co-exposures or co-stressors, which could result in greater toxicity or more severe disease. This has shaped the tungsten toxicology field and the types of research questions being investigated. This has particularly been true when evaluating the toxicity profile of tungsten metal alloys in combination with cobalt. In this chapter, the current state of the tungsten toxicology field will be discussed focusing on data investigating tungsten carcinogenicity and other major toxicities including pulmonary, cardiometabolic, bone, and immune endpoints, either alone or in combination with other metals. Environmental and human monitoring data will also be discussed to highlight human populations most at risk of exposure to high concentrations of tungsten, the forms of tungsten present in each setting, and exposure levels in each population.