Engineering BinB Pore-Forming Toxin for Selective Killing of Breast Cancer Cells

Breast cancer is one of the most common cancers in women worldwide. Conventional cancer chemotherapy always has adverse side effects on the patient’s healthy tissues. Consequently, combining pore-forming toxins with cell-targeting peptides (CTPs) is a promising anticancer strategy for selectively de...

Full description

Bibliographic Details
Main Authors: Tipaporn Kumkoon, Chalongrat Noree, Panadda Boonserm
Format: Article
Language:English
Published: MDPI AG 2023-04-01
Series:Toxins
Subjects:
Online Access:https://www.mdpi.com/2072-6651/15/4/297
_version_ 1797603300368973824
author Tipaporn Kumkoon
Chalongrat Noree
Panadda Boonserm
author_facet Tipaporn Kumkoon
Chalongrat Noree
Panadda Boonserm
author_sort Tipaporn Kumkoon
collection DOAJ
description Breast cancer is one of the most common cancers in women worldwide. Conventional cancer chemotherapy always has adverse side effects on the patient’s healthy tissues. Consequently, combining pore-forming toxins with cell-targeting peptides (CTPs) is a promising anticancer strategy for selectively destroying cancer cells. Here, we aim to improve the target specificity of the BinB toxin produced from <i>Lysinibacillus sphaericus</i> (Ls) by fusing a luteinizing hormone-releasing hormone (LHRH) peptide to its pore-forming domain (BinB<sub>C</sub>) to target MCF-7 breast cancer cells as opposed to human fibroblast cells (Hs68). The results showed that LHRH-BinB<sub>C</sub> inhibited MCF-7 cell proliferation in a dose-dependent manner while leaving Hs68 cells unaffected. BinB<sub>C</sub>, at any concentration tested, did not affect the proliferation of MCF-7 or Hs68 cells. In addition, the LHRH-BinB<sub>C</sub> toxin caused the efflux of the cytoplasmic enzyme lactate dehydrogenase (LDH), demonstrating the efficacy of the LHRH peptide in directing the BinB<sub>C</sub> toxin to damage the plasma membranes of MCF-7 cancer cells. LHRH-BinB<sub>C</sub> also caused MCF-7 cell apoptosis by activating caspase-8. In addition, LHRH-BinB<sub>C</sub> was predominantly observed on the cell surface of MCF-7 and Hs68 cells, without colocalization with mitochondria. Overall, our findings suggest that LHRH-BinB<sub>C</sub> could be investigated further as a potential cancer therapeutic agent.
first_indexed 2024-03-11T04:28:14Z
format Article
id doaj.art-947274fca4104cbdb4162d0526953293
institution Directory Open Access Journal
issn 2072-6651
language English
last_indexed 2024-03-11T04:28:14Z
publishDate 2023-04-01
publisher MDPI AG
record_format Article
series Toxins
spelling doaj.art-947274fca4104cbdb4162d05269532932023-11-17T21:39:12ZengMDPI AGToxins2072-66512023-04-0115429710.3390/toxins15040297Engineering BinB Pore-Forming Toxin for Selective Killing of Breast Cancer CellsTipaporn Kumkoon0Chalongrat Noree1Panadda Boonserm2Institute of Molecular Biosciences, Mahidol University, Salaya, Phuttamonthon, Nakhon Pathom 73170, ThailandInstitute of Molecular Biosciences, Mahidol University, Salaya, Phuttamonthon, Nakhon Pathom 73170, ThailandInstitute of Molecular Biosciences, Mahidol University, Salaya, Phuttamonthon, Nakhon Pathom 73170, ThailandBreast cancer is one of the most common cancers in women worldwide. Conventional cancer chemotherapy always has adverse side effects on the patient’s healthy tissues. Consequently, combining pore-forming toxins with cell-targeting peptides (CTPs) is a promising anticancer strategy for selectively destroying cancer cells. Here, we aim to improve the target specificity of the BinB toxin produced from <i>Lysinibacillus sphaericus</i> (Ls) by fusing a luteinizing hormone-releasing hormone (LHRH) peptide to its pore-forming domain (BinB<sub>C</sub>) to target MCF-7 breast cancer cells as opposed to human fibroblast cells (Hs68). The results showed that LHRH-BinB<sub>C</sub> inhibited MCF-7 cell proliferation in a dose-dependent manner while leaving Hs68 cells unaffected. BinB<sub>C</sub>, at any concentration tested, did not affect the proliferation of MCF-7 or Hs68 cells. In addition, the LHRH-BinB<sub>C</sub> toxin caused the efflux of the cytoplasmic enzyme lactate dehydrogenase (LDH), demonstrating the efficacy of the LHRH peptide in directing the BinB<sub>C</sub> toxin to damage the plasma membranes of MCF-7 cancer cells. LHRH-BinB<sub>C</sub> also caused MCF-7 cell apoptosis by activating caspase-8. In addition, LHRH-BinB<sub>C</sub> was predominantly observed on the cell surface of MCF-7 and Hs68 cells, without colocalization with mitochondria. Overall, our findings suggest that LHRH-BinB<sub>C</sub> could be investigated further as a potential cancer therapeutic agent.https://www.mdpi.com/2072-6651/15/4/297pore-forming toxin<i>Lysinibacillus sphaericus</i>cell-targeting peptidebreast cancer cellscell death
spellingShingle Tipaporn Kumkoon
Chalongrat Noree
Panadda Boonserm
Engineering BinB Pore-Forming Toxin for Selective Killing of Breast Cancer Cells
Toxins
pore-forming toxin
<i>Lysinibacillus sphaericus</i>
cell-targeting peptide
breast cancer cells
cell death
title Engineering BinB Pore-Forming Toxin for Selective Killing of Breast Cancer Cells
title_full Engineering BinB Pore-Forming Toxin for Selective Killing of Breast Cancer Cells
title_fullStr Engineering BinB Pore-Forming Toxin for Selective Killing of Breast Cancer Cells
title_full_unstemmed Engineering BinB Pore-Forming Toxin for Selective Killing of Breast Cancer Cells
title_short Engineering BinB Pore-Forming Toxin for Selective Killing of Breast Cancer Cells
title_sort engineering binb pore forming toxin for selective killing of breast cancer cells
topic pore-forming toxin
<i>Lysinibacillus sphaericus</i>
cell-targeting peptide
breast cancer cells
cell death
url https://www.mdpi.com/2072-6651/15/4/297
work_keys_str_mv AT tipapornkumkoon engineeringbinbporeformingtoxinforselectivekillingofbreastcancercells
AT chalongratnoree engineeringbinbporeformingtoxinforselectivekillingofbreastcancercells
AT panaddaboonserm engineeringbinbporeformingtoxinforselectivekillingofbreastcancercells