scholarly journals Surface Engineering of Escherichia coli–Derived OMVs as Promising Nano-Carriers to Target EGFR-Overexpressing Breast Cancer Cells

2021 ◽  
Vol 12 ◽  
Author(s):  
Zahra Sepahdar ◽  
Mehran Miroliaei ◽  
Saeid Bouzari ◽  
Vahid Khalaj ◽  
Mona Salimi

Bacterial outer membrane vesicles (OMVs) have recently drawn a great deal of attention due to their therapeutic efficiency and ability to target specific cells. In the present study, we sought to probe engineered OMVs as novel and promising carriers to target breast cancer cells. Following the fusion of the affiEGFR-GALA structure to the C-terminal of ClyA as an anchor protein, the ClyA-affiEGFR-GALA construct was successfully expressed on the surface of ∆msbB/∆pagP E. coli W3110-derived OMVs. Morphological features of the engineered and wild-type OMVs were identical. The engineered OMVs induced no endotoxicity, cytotoxicity, or immunogenicity, indicating the safety of their application. These OMVs could specifically bind to EGF receptors of MDA-MB-468 cells expressing high levels of EGFR and not to those with low levels of EGFR (HEK293T cells). Interestingly, despite a lower binding affinity of the engineered OMVs relative to the positive control Cetuximab, it was strong enough to identify these cells. Moreover, confocal microscopy revealed no uptake of the modified OMVs by the EGFR-overexpressing cells in the presence of EGFR competitors. These results suggest that OMVs might internalize into the cells with EGF receptors, as no OMVs entered the cells with any EGFR expression or those pretreated with EGF or Cetuximab. Regarding the EGFR-binding affinity of the engineered OMVs and their cellular uptake, they are presented here as a potential carrier for cell-specific drug delivery to treat a wide variety of cancer cells. Interestingly, the engineered OMVs are capable of reaching the cytoplasm while escaping the endosome due to the incorporation of a fusogenic GALA peptide in the construct.

2021 ◽  
Author(s):  
Mingping Qian ◽  
Hui Xu ◽  
Hongming Song ◽  
Hao Xi ◽  
Lin Fang

Abstract Background : MiR-218-5p is a small non-coding RNA acting as either oncogenes or tumor suppressor genes in human cancer. The expression levels of some miRNAs in human breast cancer plays a potential role in disease pathogenesis. Methods : Thirty pairs of invasive ductal carcinoma and adjacent specimens were included in the study. Breast tissues cell lines MCF-7 and MDA-MB-231 were identified as a breast cancer research cell line. MiR-218-5p mimics, miR-218-5p inhibitor, or negative controls were transfected. Specific antibodies were probed with LRIG1, ErbB2, and EGFR. Proliferation, migration, cell cycle and apoptosis, dual-luciferase reporter assay and immunohistochemistry were used to analyze miR-218-5p、LRIG1 and so on. Results : It was shown that miR-218-5p expression was higher in 30 breast cancer specimens than adjacent normal breast tissues. In human breast cancer cells MCF-7 and MDA-MB-231, restoring miR-218-5p promoted cell proliferation and migration and inhibited cell apoptosis and cell cycle arrest in the G1 stage. Luciferase assays indicated miR-218-5p could bind with its putative target site in the 3'-untranslated region (3'-UTR) of LRIG1. RT-qPCR, western blot, and immunocytochemistry analyses all indicated miR-218-5p overexpression results in LRIG1 downregulation at the mRNA and protein levels. ErbB2 and EGFR were found to be downstream effectors of miR-218-5p. Conclusion : MiR-218-5p promotes ErbB2 and EGFR expression by inhibiting LRIG1 in breast cancer cells, which suggests miR-218-5p and LRIG1 may act as an oncogene in breast cancer and it could be used as a therapeutic target for breast cancer treatments. Keywords: Breast cancer; miR-218-5p; LRIG1; Oncogene


2021 ◽  
Author(s):  
Abdolamir Ghadaksaz ◽  
Abbas Ali Imani Fooladi ◽  
Hamideh Mahmoodzadeh Hosseini ◽  
Taher Nejad Satari ◽  
Mohsen Amin

Abstract PurposeTargeted cancer therapies based on overexpressed receptors and the fractions containing immunotoxins and bacterial metabolites are one of the well-known methods to overcome the chemotherapy resistance of cancer cells. In this paper, we design ARA-linker-TGFαL3, using Arazyme, a Serratia proteamaculans metabolite, and a third loop segment of TGFα to target EGFR expressing breast cancer cells.MethodsAfter cloning in pET28a (+), the expression of recombinant protein was optimized in E. coli strain BL21 (DE3). MDA-MB-468 (EGFR positive) and MDA-MB-453 (EGFR negative) breast cancer cell lines were employed. Also, the chemotherapeutic drug, Taxotere (Docetaxel), was employed to compare cytotoxicity effects. Cell ELISA assessed the binding affinity of recombinant proteins to the receptor, and the cytotoxicity was detected by MTT and lactate dehydrogenase release assays. The interfacing with cancer cell adhesion was evaluated. Furthermore, the induction of apoptosis was examined by using flow cytometric analysis, and caspase-3 activity assay. Moreover, RT-PCR was conducted to study the expression of apoptosis (bax, bcl2, and casp3), angiogenesis (vegfr2), and metastasis (mmp2 and mmp9) genes. ResultsARA-linker-TGFαL3 revealed a higher binding affinity, cytotoxicity, and early apoptosis induction in MDA-MB-468 compared to the effects of Arazyme while both recombinant proteins showed similar effects on MDA-MB-453. In addition, the Taxotere caused the highest cytotoxicity on cancer cells through induction of late apoptosis. Meanwhile, the expression of angiogenesis and metastasis genes was decreased in both cell lines after treatment with either ARA-linker-TGFαL3 or Arazyme. ConclusionsOur in vitro results indicated the therapeutic effect of ARA-linker-TGFαL3 on breast cancer cells.


2006 ◽  
Author(s):  
Αικατερίνη Ζώμπρα

Leuprolide is a synthetic nonapeptide, analogue of Luteinizing Hormone Releasing Hormone (LHRH or GnRH). Is a super agonist of LHRH and as an active pharmaceutical ingredient has been used in the treatment of many sex hormone-related disorders. Regulation of hypothalamic-gonadal-axis by LHRH analogues is used for the treatment of hormone-dependent diseases, such as endometriosis, benign prostate hyperplasia, fertility disorders, as well as prostate, ovarian and breast cancer to induce reversible chemical castration. The currently available LHRH analogues as drugs and their applications in the treatment of the above diseases lead the scientists to the development of new more potent LHRH analogues with better pharmacological profile. In recent years the biology of GnRH has been revised, due to accumulating evidence that peripheral, extrapituitary, normal and malignant tissues locally produce GnRH and express GnRH binding sites. An autocrine/paracrine role of GnRH at these normal and malignant tissues, including the mammary gland, has been suggested. The direct antiproliferative effect of the LHRH analogues in many malignant tissues inaugurates the field and leads to the design of effective LHRH analogues with new perspectives. To-date, only a few studies have investigated the effects of LHRH analogues in pituitary and cancer cells in parallel. Information concerning the impact of systematic single amino acid modifications of LHRH on pituitary and malignant cells is still lacking. The structural investigation of this field seems to be a very important issue. The aim of this study was the contribution in the field of the structure-activity relationships based in the rational design (bibliographic data, conformational analysis), development (modern synthesis) and biological characterization of the new analogues of Leuprolide. Particularly, with the biological characterization, we tried to compare the binding affinities of the new Leuprolide analogues in the LHRH hypophysial receptor with their direct antiproliferative activity on breast cancer cells. Furthermore, the development of a new synthetic protocol which leads to high yield and purities of the synthesized products was a requisition. Important structural data obtained by conformational analysis of LHRH and Leuprolide using NMR spectroscopy. This conformational study in combination with the bibliographic data led us to design and synthesize new analogues of Leuprolide with main modifications in positions 3 and 6. Trp3 of peptide sequence of Leuprolide is substituted by non natural aromatic amino/imino acids. D-Leu6 is substituted by α,α-dialkyl amino acids L, D-Gly(tBu), D-Lys and D-Dab. In the ε- and γ-amino group of Lysine and diaminoisobutyric acid in position 6 several modifications have been performed by conjugation of natural amino acids (Gly, Ala, Sar) and 2-amino-4-pyrrolidinothieno[2,3-d]pyrimidine-6-carboxylic acid (ATPC). ATPC was also incorporated in the N-terminal segment replacing the final dipeptide of LHRH and Leuprolide. Like in the peptide sequence of Leuprolide, most of the new LHRH analogues lack the carboxy-terminal Gly10-amide of LHRH and an ethylamide residue is added to Pro9 (Fujino modification). The new analogues of LHRH were assembled on a [3-((Ethyl-Fmoc-amino)-methyl)-1-indol-1-yl]-acetyl AM resin to provide the peptide amide using Fmoc/tBu methodology. The affinities for the LHRH receptor of the new analogues were determined by competition binding experiments in mouse anterior pituitary αT3-1 cells while their antiproliferative activity was performed with human breast cancer cell lines (MCF-7). Modern peptide synthesis afforded us peptides in high yields which could be easily purified. This study provides binding requirements which can be used in the design of new Leuprolide analogues with high pituitary binding affinity and high antiproliferative activity in breast cancer cells (MCF-7). Results show in many cases differential impact on pituitary binding affinity and direct antiproliferative effect on breast cancer cells which is in accordance with bibliographic reports that mainly support this dissimilarity to different signal transduction mechanisms in hypothalamic and extra hypothalamic malignant tissues. Final this study provides new Leuprolide analogues with higher binding affinity and antiproliferative activity than it has exhibited and they deserve further pharmaceutical/biological investigation. This structure-activity relationship study for LHRH analogues at pituitary and breast cancer cell lines contributes to the on-going research of LHRH binding requirements of breast cancer cells and provides new perspectives for the design of effective GnRH analogues.


2020 ◽  
Vol 21 (23) ◽  
pp. 9008
Author(s):  
Florian Weinberg ◽  
Diana B. Peckys ◽  
Niels de Jonge

The epidermal growth factor receptor HER2 is overexpressed in 20% of breast cancer cases. HER2 is an orphan receptor that is activated ligand-independently by homodimerization. In addition, HER2 is able to heterodimerize with EGFR, HER3, and HER4. Heterodimerization has been proposed as a mechanism of resistance to therapy for HER2 overexpressing breast cancer. Here, a method is presented for the simultaneous detection of individual EGFR and HER2 receptors in the plasma membrane of breast cancer cells via specific labeling with quantum dot nanoparticles (QDs). Correlative fluorescence microscopy and liquid phase electron microscopy were used to analyze the plasma membrane expression levels of both receptors in individual intact cells. Fluorescent single-cell analysis of SKBR3 breast cancer cells dual-labeled for EGFR and HER2 revealed a heterogeneous expression for receptors within both the cell population as well as within individual cells. Subsequent electron microscopy of individual cells allowed the determination of individual receptors label distributions. QD-labeled EGFR was observed with a surface density of (0.5–5) × 101 QDs/µm2, whereas labeled HER2 expression was higher ranging from (2–10) × 102 QDs/µm2. Although most SKBR3 cells expressed low levels of EGFR, an enrichment was observed at large plasma membrane protrusions, and amongst a newly discovered cellular subpopulation termed EGFR-enriched cells.


2015 ◽  
Vol 77 (31) ◽  
Author(s):  
Nadhira Atiqah Khiru Nasir ◽  
Siti Pauliena Mohd Bohari

Cancers are diseases that can cause death and breast cancer is one of the prevalence cancers. Conventional treatments have been used to treat cancer. However, these treatments produced inefficient effects and low survival rate. Therefore, most cancer patients began to consume complementary and utilized alternative treatments to alleviate their pain. This study is to identify the cytotoxicity effect of methanolic leaves extract Typhonium flagelliforme and Clinacanthus nutans on breast cancer cells (MDA-MB-231). MTT assay has been used to determine the cytotoxicity effect of both plants on MDA-MB-231 cells and CHO cells (non-cancerous cell), which acted as the positive control cells. Result revealed that T. flagelliforme extract has shown higher cytotoxic effect on MDA-MB-231cells (IC50: 0.11 mg/mL) compared to C. nutans extract (IC50: 0.17 mg/mL). Then, the IC50 values of T. flagelliforme and C. nutans extract on CHO cells were 0.10 mg/mL and 0.24 mg/mL respectively. Based on these results, T. flagelliforme represented higher toxicity effect on CHO compared to the MDA-MB-231 cells. Thus, in future CHO cell (as a positive control) can be replaced with a normal breast cell line such as HCC1395 (epithelial mammary duct of normal breast cells) to understand clearly the toxicity effect of T. flagelliforme extract towards the normal breast cell. Moreover, identification of potential compounds that can inhibit MDA-MB-231 cells growth  is also important for future research.


2008 ◽  
Vol 7 (7) ◽  
pp. 1846-1850 ◽  
Author(s):  
Dongwei Zhang ◽  
Ashutosh Pal ◽  
William G. Bornmann ◽  
Fumiyuki Yamasaki ◽  
Francisco J. Esteva ◽  
...  

Molecules ◽  
2020 ◽  
Vol 25 (20) ◽  
pp. 4709
Author(s):  
Fatema Al-Rashed ◽  
Reeby Thomas ◽  
Areej Al-Roub ◽  
Fahd Al-Mulla ◽  
Rasheed Ahmad

Granulocyte–macrophage colony-stimulating factor (GM-CSF) is a monomeric glycoprotein that has been implicated in the tumor growth and progression of different types of cancer. GM-CSF is produced by various non-immune cells including MDA-MB-231 in response to various stimuli. However, the role of lipopolysaccharide (LPS) in the regulation of GM-CSF in MDA-MB-231 breast cancer cells so far remains unclear. Herein, we asked whether LPS could induce GM-CSF production in MDA-MB-231 cells, and if so, which signaling pathway was involved. MDA-MB-231 cells were treated with LPS or tumor necrosis factor alpha (TNF-α; positive control), and GM-CSF expression levels were determined by qRT-PCR, ELISA, and confocal microscopy. Phosphorylation of the mitogen-activated protein kinases (MAPKs) and nuclear factor-κB (NF-kB) signaling proteins were evaluated by flow cytometry. Our results show that LPS induces GM-CSF expression at both mRNA and protein levels in MDA-MBA-231 cells. Inhibition of acyl-CoA synthetase 1 (ACSL1) activity in the cells with triacsin C significantly reduces the secretion of GM-CSF. Furthermore, the inhibition of ACSL1 activity significantly blocks the LPS-mediated phosphorylation of p38 MAPK, MEK1/2, extracellular signal-regulated kinase (ERK)1/2, c-Jun NH2-terminal kinase (JNK), and nuclear factor-κB (NF-kB) in the cells. These findings provide the first evidence that LPS induces ACSL1-dependent GM-CSF gene expression in MDA-MB-231 breast cancer cells, which requires the activation of p38 MAPK, MEK1/2, ERK1/2, JNK, and NF-kB.


Sign in / Sign up

Export Citation Format

Share Document