molecules-logo

Journal Browser

Journal Browser

Targeting of Signaling Pathways for Cancer Therapy

A special issue of Molecules (ISSN 1420-3049). This special issue belongs to the section "Chemical Biology".

Deadline for manuscript submissions: closed (31 December 2021) | Viewed by 37997

Special Issue Editors


E-Mail Website
Guest Editor
Department of Pathology, Kazan State Medical University, Kazan, Russia
Interests: cancer, gastrointestinal stromal tumors (GIST); soft tissue sarcomas (STS); targeted-based therapy; resistance; apoptosis; receptor tyrosine kinase inhibitors (RTKi); DNA damage repair (DDR); FGF-signaling
Special Issues, Collections and Topics in MDPI journals

E-Mail Website
Guest Editor
Cytogenetics Laboratory, Carcinogenesis Institute, N.N. Blokhin National Medical Research Center of Oncology, 115478 Moscow, Russia
Interests: tumor progression; cell signaling; cancer stem cells; mesenchymal cells; actin isoforms

Special Issue Information

Dear Colleagues,

The cell signaling pathways are well-known regulators of cell growth, proliferation, migration, differentiation, and survival. Meanwhile, several signaling pathways have been identified as frequently genetically altered in cancer, including the p53-signaling pathway, RTK/RAS/MAP-kinase and PI3K/AKT signaling pathway, Notch and Wnt signaling pathways, GSK3-signaling pathway, actin dynamics signaling pathway, death receptor signaling pathway, autophagy pathway, and others. For example, activation of some of the pathways indicated above might be due to activating mutations of RTKs, Ras, PI3k, Akt, etc., or loss of function of tumor suppressors (e.g., Pten). Given that signal transduction in these pathways is tightly regulated by the protein kinases, and taking into account that several of the cell membrane receptor families activate the same downstream intracellular pathways, development of the specific kinase or other receptor inhibitors targeting the universal signaling molecules is considered as one of the most promising approaches for the personalized therapies for cancers addicted to a particular signaling pathway.

Prof. Dr. Sergei Boichuk
Dr. Pavel B. Kopnin
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Molecules is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2700 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • Cancer
  • kinase
  • receptor
  • signaling
  • pathway
  • growth
  • cell death
  • small molecule inhibitors

Related Special Issue

Published Papers (13 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

14 pages, 2952 KiB  
Article
Integrated Machine Learning and Chemoinformatics-Based Screening of Mycotic Compounds against Kinesin Spindle ProteinEg5 for Lung Cancer Therapy
by Priyanka Maiti, Priyanka Sharma, Mahesha Nand, Indra D. Bhatt, Muthannan Andavar Ramakrishnan, Shalini Mathpal, Tushar Joshi, Ragini Pant, Shafi Mahmud, Jesus Simal-Gandara, Sultan Alshehri, Mohammed M. Ghoneim, Maha Alruwaily, Ahmed Abdullah Al Awadh, Mohammed Merae Alshahrani and Subhash Chandra
Molecules 2022, 27(5), 1639; https://doi.org/10.3390/molecules27051639 - 02 Mar 2022
Cited by 6 | Viewed by 3072
Abstract
Among the various types of cancer, lung cancer is the second most-diagnosed cancer worldwide. The kinesin spindle protein, Eg5, is a vital protein behind bipolar mitotic spindle establishment and maintenance during mitosis. Eg5 has been reported to contribute to cancer cell migration and [...] Read more.
Among the various types of cancer, lung cancer is the second most-diagnosed cancer worldwide. The kinesin spindle protein, Eg5, is a vital protein behind bipolar mitotic spindle establishment and maintenance during mitosis. Eg5 has been reported to contribute to cancer cell migration and angiogenesis impairment and has no role in resting, non-dividing cells. Thus, it could be considered as a vital target against several cancers, such as renal cancer, lung cancer, urothelial carcinoma, prostate cancer, squamous cell carcinoma, etc. In recent years, fungal secondary metabolites from the Indian Himalayan Region (IHR) have been identified as an important lead source in the drug development pipeline. Therefore, the present study aims to identify potential mycotic secondary metabolites against the Eg5 protein by applying integrated machine learning, chemoinformatics based in silico-screening methods and molecular dynamic simulation targeting lung cancer. Initially, a library of 1830 mycotic secondary metabolites was screened by a predictive machine-learning model developed based on the random forest algorithm with high sensitivity (1) and an ROC area of 0.99. Further, 319 out of 1830 compounds screened with active potential by the model were evaluated for their drug-likeness properties by applying four filters simultaneously, viz., Lipinski’s rule, CMC-50 like rule, Veber rule, and Ghose filter. A total of 13 compounds passed from all the above filters were considered for molecular docking, functional group analysis, and cell line cytotoxicity prediction. Finally, four hit mycotic secondary metabolites found in fungi from the IHR were screened viz., (−)-Cochlactone-A, Phelligridin C, Sterenin E, and Cyathusal A. All compounds have efficient binding potential with Eg5, containing functional groups like aromatic rings, rings, carboxylic acid esters, and carbonyl and with cell line cytotoxicity against lung cancer cell lines, namely, MCF-7, NCI-H226, NCI-H522, A549, and NCI H187. Further, the molecular dynamics simulation study confirms the docked complex rigidity and stability by exploring root mean square deviations, root mean square fluctuations, and radius of gyration analysis from 100 ns simulation trajectories. The screened compounds could be used further to develop effective drugs against lung and other types of cancer. Full article
(This article belongs to the Special Issue Targeting of Signaling Pathways for Cancer Therapy)
Show Figures

Graphical abstract

10 pages, 632 KiB  
Communication
Identification of New Regulators of Pancreatic Cancer Cell Sensitivity to Oxaliplatin and Cisplatin
by Vera Skripova, Ramilia Vlasenkova, Yan Zhou, Igor Astsaturov and Ramziya Kiyamova
Molecules 2022, 27(4), 1289; https://doi.org/10.3390/molecules27041289 - 14 Feb 2022
Cited by 8 | Viewed by 2858
Abstract
The chemoresistance of tumor cells is one of the most urgent challenges in modern oncology and in pancreatic cancer, in which this problem is the most prominent. Therefore, the identification of new chemosensitizing co-targets may be a path toward increasing chemotherapy efficacy. In [...] Read more.
The chemoresistance of tumor cells is one of the most urgent challenges in modern oncology and in pancreatic cancer, in which this problem is the most prominent. Therefore, the identification of new chemosensitizing co-targets may be a path toward increasing chemotherapy efficacy. In this work, we performed high-performance in vitro knockout CRISPR/Cas9 screening to find potential regulators of the sensitivity of pancreatic cancer. For this purpose, MIA PaCa-2 cells transduced with two sgRNA libraries (“cell cycle/nuclear proteins genes” and “genome-wide”) were screened by oxaliplatin and cisplatin. In total, 173 candidate genes were identified as potential regulators of pancreatic cancer cell sensitivity to oxaliplatin and/or cisplatin; among these, 25 genes have previously been reported, while 148 genes were identified for the first time as potential platinum drug sensitivity regulators. We found seven candidate genes involved in pancreatic cancer cell sensitivity to both cisplatin and oxaliplatin. Gene ontology enrichment analysis reveals the enrichment of single-stranded DNA binding, damaged DNA binding pathways, and four associated with NADH dehydrogenase activity. Further investigation and validation of the obtained results by in vitro, in vivo, and bioinformatics approaches, as well as literature analysis, will help to identify novel pancreatic cancer platinum sensitivity regulators. Full article
(This article belongs to the Special Issue Targeting of Signaling Pathways for Cancer Therapy)
Show Figures

Figure 1

20 pages, 6450 KiB  
Article
Negative Modulation of the Angiogenic Cascade Induced by Allosteric Kinesin Eg5 Inhibitors in a Gastric Adenocarcinoma In Vitro Model
by Alessia Ricci, Marialucia Gallorini, Donatella Del Bufalo, Amelia Cataldi, Ilaria D’Agostino, Simone Carradori and Susi Zara
Molecules 2022, 27(3), 957; https://doi.org/10.3390/molecules27030957 - 31 Jan 2022
Cited by 10 | Viewed by 2516
Abstract
Eg5 is a kinesin essential in bipolar spindle formation, overexpressed in tumours, thus representing a new target in cancer therapy. We aimed at evaluating the anti-cancer activity of Eg5 thiadiazoline inhibitors 2 and 41 on gastric adenocarcinoma cells (AGS), focusing on the modulation [...] Read more.
Eg5 is a kinesin essential in bipolar spindle formation, overexpressed in tumours, thus representing a new target in cancer therapy. We aimed at evaluating the anti-cancer activity of Eg5 thiadiazoline inhibitors 2 and 41 on gastric adenocarcinoma cells (AGS), focusing on the modulation of angiogenic signalling. Docking studies confirmed a similar interaction with Eg5 to that of the parent compound K858. Thiadiazolines were also tested in combination with Hesperidin (HSD). Cell cycle analysis reveals a reduction of G1 and S phase percentages when 41 is administered as well as HSD in combination with K858. Western blot reveals Eg5 inhibitors capability to reduce PI3K, p-AKT/Akt and p-Erk/Erk expressions; p-Akt/Akt ratio is even more decreased in HSD+2 sample than the p-Erk/Erk ratio in HSD+41 or K858. VEGF expression is reduced when HSD+2 and HSD+41 are administered with respect to compounds alone, after 72 h. ANGPT2 gene expression increases in cells treated with 41 and HSD+2 compared to K858. The wound-healing assay highlights a reduction in the cut in HSD+2 sample compared to 2 and HSD. Thus, Eg5 inhibitors appear to modulate angiogenic signalling by controlling VEGF activity even better if combined with HSD. Overall, Eg5 inhibitors can represent a promising starting point to develop innovative anti-cancer strategies. Full article
(This article belongs to the Special Issue Targeting of Signaling Pathways for Cancer Therapy)
Show Figures

Graphical abstract

19 pages, 1617 KiB  
Article
Molecular Mechanisms of Anticancer Activity of N-Glycosides of Indolocarbazoles LCS-1208 and LCS-1269
by Roman G. Zenkov, Olga A. Vlasova, Varvara P. Maksimova, Timur I. Fetisov, Natalia Y. Karpechenko, Lidiya V. Ektova, Vera A. Eremina, Valeriia G. Popova, Olga G. Usalka, Ekaterina A. Lesovaya, Gennady A. Belitsky, Marianna G. Yakubovskaya and Kirill I. Kirsanov
Molecules 2021, 26(23), 7329; https://doi.org/10.3390/molecules26237329 - 02 Dec 2021
Cited by 5 | Viewed by 2021
Abstract
Novel indolocarbazole derivatives named LCS were synthesized by our research group. Two of them were selected as the most active anticancer agents in vivo. We studied the mechanisms of anticancer activity in accordance with the previously described effects of indolocarbazoles. Cytotoxicity was estimated [...] Read more.
Novel indolocarbazole derivatives named LCS were synthesized by our research group. Two of them were selected as the most active anticancer agents in vivo. We studied the mechanisms of anticancer activity in accordance with the previously described effects of indolocarbazoles. Cytotoxicity was estimated by MTT assay. We analyzed LCS-DNA interactions by circular dichroism in cholesteric liquid crystals and fluorescent indicator displacement assay. The effect on the activity of topoisomerases I and II was studied by DNA relaxation assay. Expression of interferon signaling target genes was estimated by RT-PCR. Chromatin remodeling was analyzed–the effect on histone H1 localization and reactivation of epigenetically silenced genes. LCS-induced change in the expression of a wide gene set was counted by means of PCR array. Our study revealed the cytotoxic activity of the compounds against 11 cancer cell lines and it was higher than in immortalized cells. Both compounds bind DNA; binding constants were estimated—LCS-1208 demonstrated higher affinity than LCS-1269; it was shown that LCS-1208 intercalates into DNA that is typical for rebeccamycin derivatives. LCS-1208 also inhibits topoisomerases I and IIα. Being a strong intercalator and topoisomerase inhibitor, LCS-1208 upregulates the expression of interferon-induced genes. In view of LCSs binding to DNA we analyzed their influence on chromatin stability and revealed that LCS-1269 displaces histone H1. Our analysis of chromatin remodeling also included a wide set of epigenetic experiments in which LCS-1269 demonstrated complex epigenetic activity. Finally, we revealed that the antitumor effect of the compounds is based not only on binding to DNA and chromatin remodeling but also on alternative mechanisms. Both compounds induce expression changes in genes involved in neoplastic transformation and target genes of the signaling pathways in cancer cells. Despite of being structurally similar, each compound has unique biological activities. The effects of LCS-1208 are associated with intercalation. The mechanisms of LCS-1269 include influence on higher levels such as chromatin remodeling and epigenetic effects. Full article
(This article belongs to the Special Issue Targeting of Signaling Pathways for Cancer Therapy)
Show Figures

Graphical abstract

16 pages, 2490 KiB  
Article
Plasma Exosomes of Patients with Breast and Ovarian Tumors Contain an Inactive 20S Proteasome
by Natalia Yunusova, Elena Kolegova, Elena Sereda, Larisa Kolomiets, Alisa Villert, Marina Patysheva, Irina Rekeda, Alina Grigor’eva, Natalia Tarabanovskaya, Irina Kondakova and Svetlana Tamkovich
Molecules 2021, 26(22), 6965; https://doi.org/10.3390/molecules26226965 - 18 Nov 2021
Cited by 8 | Viewed by 1877
Abstract
Exosomes are directly involved in governing of physiological and pathological conditions of an organism through the transfer of information from producing to receiving cells. It can be assumed that exosomes are one of the key players of tumor dissemination since they are very [...] Read more.
Exosomes are directly involved in governing of physiological and pathological conditions of an organism through the transfer of information from producing to receiving cells. It can be assumed that exosomes are one of the key players of tumor dissemination since they are very stable and small enough to penetrate from various tissues into biological fluids and then back, thus interacting with tissue target cells. We evaluated the enzymatic activity and the level of 20S proteasome in tissue and exosomes of healthy females (n = 39) and patients with ovarian (n = 50) and breast (n = 108) tumors to reveal the critical role of exosomal cargo in the mediation of different types of metastases. Exosomes from plasma and ascites were isolated and characterized in according to International Society for Extracellular Vesicles guidelines. The level of 20S proteasome in tissue and exosomes was determined using Western blot analysis. Chymotrypsin- and caspase-like (ChTL and CL, respectively) peptidase activities of the proteasomes were determined using fluorogenic Suc-LLVY-AMC and Cbz-LLG-AMC substrates, respectively. We observed increased levels of 20S proteasome in ovarian cancer tissue and luminal B subtype breast cancer tissue as well as in plasma exosomes from cancer patients. Moreover, the level of the 20S proteasome in plasma exosomes and ascites exosomes in patients with ovarian tumors is comparable and higher in ovarian cancer patients with low volume ascites than in patients with moderate and high-volume ascites. We also found increased ChTL and CL activities in breast cancer and ovarian cancer tissues, as well as in peritoneal metastases in ovarian cancer, while proteasomal activity in exosomes from plasma of healthy females and all patients, as well as from ascites of ovarian tumor patients were lower than detection limit of assay. Thus, regardless of the type of tumor metastasis (lymphogenous or peritoneal), the exosomes of cancer patients were characterized by an increased level of 20S proteasome, which do not exhibit enzymatic activity. Full article
(This article belongs to the Special Issue Targeting of Signaling Pathways for Cancer Therapy)
Show Figures

Figure 1

19 pages, 6861 KiB  
Article
Nifuroxazide Mitigates Angiogenesis in Ehlrich’s Solid Carcinoma: Molecular Docking, Bioinformatic and Experimental Studies on Inhibition of Il-6/Jak2/Stat3 Signaling
by Mohamed El-Sherbiny, Rehab M. El-Sayed, Mohamed A. Helal, Afaf T. Ibrahiem, Hoda S. Elmahdi, Mohamed Ahmed Eladl, Shymaa E. Bilay, Asma M. Alshahrani, Mona K. Tawfik, Ziad E. Hamed, Amany O. Mohamed and Sawsan A. Zaitone
Molecules 2021, 26(22), 6858; https://doi.org/10.3390/molecules26226858 - 13 Nov 2021
Cited by 11 | Viewed by 2879
Abstract
Nifuroxazide is an antidiarrheal medication that has promising anticancer activity against diverse types of tumors. The present study tested the anticancer activity of nifuroxazide against Ehrlich’s mammary carcinoma grown in vivo. Furthermore, we investigated the effect of nifuroxazide on IL-6/jak2/STAT3 signaling and the [...] Read more.
Nifuroxazide is an antidiarrheal medication that has promising anticancer activity against diverse types of tumors. The present study tested the anticancer activity of nifuroxazide against Ehrlich’s mammary carcinoma grown in vivo. Furthermore, we investigated the effect of nifuroxazide on IL-6/jak2/STAT3 signaling and the possible impact on tumor angiogenesis. The biological study was supported by molecular docking and bioinformatic predictions for the possible effect of nifuroxazide on this signaling pathway. Female albino mice were injected with Ehrlich carcinoma cells to produce Ehrlich’s solid tumors (ESTs). The experimental groups were as follows: EST control, EST + nifuroxazide (5 mg/kg), and EST + nifuroxazide (10 mg/kg). Nifuroxazide was found to reduce tumor masses (730.83 ± 73.19 and 381.42 ± 109.69 mg vs. 1099.5 ± 310.83) and lessen tumor pathologies. Furthermore, nifuroxazide downregulated IL-6, TNF-α, NFk-β, angiostatin, and Jak2 proteins, and it also reduced tumoral VEGF, as indicated by ELISA and immunohistochemical analysis. Furthermore, nifuroxazide dose-dependently downregulated STAT3 phosphorylation (60% and 30% reductions, respectively). Collectively, the current experiment shed light on the antitumor activity of nifuroxazide against mammary solid carcinoma grown in vivo. The antitumor activity was at least partly mediated by inhibition of IL-6/Jak2/STAT3 signaling that affected angiogenesis (low VEGF and high angiostatin) in the EST. Therefore, nifuroxazide might be a promising antitumor medication if appropriate human studies will be conducted. Full article
(This article belongs to the Special Issue Targeting of Signaling Pathways for Cancer Therapy)
Show Figures

Figure 1

27 pages, 6259 KiB  
Article
The Design, Synthesis, and Biological Activities of Pyrrole-Based Carboxamides: The Novel Tubulin Inhibitors Targeting the Colchicine-Binding Site
by Sergei Boichuk, Aigul Galembikova, Kirill Syuzov, Pavel Dunaev, Firuza Bikinieva, Aida Aukhadieva, Svetlana Zykova, Nazim Igidov, Ksenia Gankova, Maria Novikova and Pavel Kopnin
Molecules 2021, 26(19), 5780; https://doi.org/10.3390/molecules26195780 - 24 Sep 2021
Cited by 13 | Viewed by 2559
Abstract
Microtubule targeting agents (MTAs) that interfere with the dynamic state of the mitotic spindle are well-known and effective chemotherapeutic agents. These agents interrupt the microtubule network via polymerization or depolymerization, halting the cell cycle progression and leading to apoptosis. We report two novel [...] Read more.
Microtubule targeting agents (MTAs) that interfere with the dynamic state of the mitotic spindle are well-known and effective chemotherapeutic agents. These agents interrupt the microtubule network via polymerization or depolymerization, halting the cell cycle progression and leading to apoptosis. We report two novel pyrrole-based carboxamides (CAs) (CA-61 and -84) as the compounds exhibiting potent anti-cancer properties against a broad spectrum of epithelial cancer cell lines, including breast, lung, and prostate cancer. The anti-cancer activity of CAs is due to their ability to interfere with the microtubules network and inhibit tubulin polymerization. Molecular docking demonstrated an efficient binding between these ligands and the colchicine-binding site on the tubulin. CA-61 formed two hydrogen bond interactions with THR 179 (B) and THR 353 (B), whereas two hydrogen bonds with LYS 254 (B) and 1 with ASN 101 (A) were identified for CA-84. The binding energy for CA-84 and CA-61 was −9.910 kcal/mol and −9.390 kcal/mol. A tubulin polymerization assay revealed a strong inhibition of tubulin polymerization induced by CA-61 and -84. The immunofluorescence data revealed the disruption of the tubulin assembly in CA-treated cancer cells. As an outcome of the tubulin inhibition, these compounds halted the cell cycle progression in the G2/M phase, leading to the accumulation of the mitotic cells, and further induced apoptosis. Lastly, the in vivo study indicated that CAs significantly inhibited the HCC1806 breast cancer xenograft tumor growth in a nude mouse model. Collectively, we identified the novel CAs as potent MTAs, inhibiting tubulin polymerization via binding to the colchicine-binding site, disrupting the microtubule network, and exhibiting potent pro-apoptotic activities against the epithelial cancer cell lines both in vitro and in vivo. Full article
(This article belongs to the Special Issue Targeting of Signaling Pathways for Cancer Therapy)
Show Figures

Figure 1

7 pages, 1665 KiB  
Communication
Secretion of Mutant DNA and mRNA by the Exosomes of Breast Cancer Cells
by Olga E. Andreeva, Yuri Y. Shchegolev, Alexander M. Scherbakov, Ekaterina I. Mikhaevich, Danila V. Sorokin, Margarita V. Gudkova, Irina V. Bure, Ekaterina B. Kuznetsova, Dmitry S. Mikhaylenko, Marina V. Nemtsova, Dmitry V. Bagrov and Mikhail A. Krasil’nikov
Molecules 2021, 26(9), 2499; https://doi.org/10.3390/molecules26092499 - 25 Apr 2021
Cited by 12 | Viewed by 2250
Abstract
Exosomes are the small vesicles that are secreted by different types of normal and tumour cells and can incorporate and transfer their cargo to the recipient cells. The main goal of the present work was to study the tumour exosomes’ ability to accumulate [...] Read more.
Exosomes are the small vesicles that are secreted by different types of normal and tumour cells and can incorporate and transfer their cargo to the recipient cells. The main goal of the present work was to study the tumour exosomes’ ability to accumulate the parent mutant DNA or RNA transcripts with their following transfer to the surrounding cells. The experiments were performed on the MCF7 breast cancer cells that are characterized by the unique coding mutation in the PIK3CA gene. Using two independent methods, Sanger sequencing and allele-specific real-time PCR, we revealed the presence of the fragments of the mutant DNA and RNA transcripts in the exosomes secreted by the MCF7 cells. Furthermore, we demonstrated the MCF7 exosomes’ ability to incorporate into the heterologous MDA-MB-231 breast cancer cells supporting the possible transferring of the exosomal cargo into the recipient cells. Sanger sequencing of the DNA from MDA-MB-231 cells (originally bearing a wild type of PIK3CA) treated with MCF7 exosomes showed no detectable amount of mutant DNA or RNA; however, using allele-specific real-time PCR, we revealed a minor signal from amplification of a mutant allele, showing a slight increase of mutant DNA in the exosome-treated MDA-MB-231 cells. The results demonstrate the exosome-mediated secretion of the fragments of mutant DNA and mRNA by the cancer cells and the exosomes’ ability to transfer their cargo into the heterologous cells. Full article
(This article belongs to the Special Issue Targeting of Signaling Pathways for Cancer Therapy)
Show Figures

Graphical abstract

14 pages, 4061 KiB  
Article
Impaired Expression of Cytoplasmic Actins Leads to Chromosomal Instability of MDA-MB-231 Basal-Like Mammary Gland Cancer Cell Line
by Vera Dugina, Galina Shagieva, Mariya Novikova, Svetlana Lavrushkina, Olga Sokova, Igor Kireev and Pavel Kopnin
Molecules 2021, 26(8), 2151; https://doi.org/10.3390/molecules26082151 - 08 Apr 2021
Cited by 1 | Viewed by 2387
Abstract
We have shown previously that two cytoplasmic actin isoforms play different roles in neoplastic cell transformation. Namely, β-cytoplasmic actin acts as a tumor suppressor, whereas γ-cytoplasmic actin enhances malignant features of tumor cells. The distinct participation of each cytoplasmic actin in the cell [...] Read more.
We have shown previously that two cytoplasmic actin isoforms play different roles in neoplastic cell transformation. Namely, β-cytoplasmic actin acts as a tumor suppressor, whereas γ-cytoplasmic actin enhances malignant features of tumor cells. The distinct participation of each cytoplasmic actin in the cell cycle driving was also observed. The goal of this study was to describe the diverse roles of cytoplasmic actins in the progression of chromosomal instability of MDA-MB-231 basal-like human carcinoma cell line. We performed traditional methods of chromosome visualization, as well as 3D-IF microscopy and western blotting for CENP-A detection/quantification, to investigate chromosome morphology. Downregulation of cytoplasmic actin isoforms alters the phenotype and karyotype of MDA-MB-231 breast cancer cells. Moreover, β-actin depletion leads to the progression of chromosomal instability with endoreduplication and aneuploidy increase. On the contrary, γ-actin downregulation results not only in reduced percentage of mitotic carcinoma cells, but leads to chromosome stability, reduced polyploidy, and aneuploidy. Full article
(This article belongs to the Special Issue Targeting of Signaling Pathways for Cancer Therapy)
Show Figures

Figure 1

19 pages, 4734 KiB  
Article
Metformin Inhibits the Urea Cycle and Reduces Putrescine Generation in Colorectal Cancer Cell Lines
by Tao Zhang, Ling Hu, Jia-Feng Tang, Hang Xu, Kuan Tian, Meng-Na Wu, Shi-Ying Huang, Yu-Mei Du, Peng Zhou, Rui-Jin Lu, Shuang He, Jia-Mei Xu, Jian-Jun Si, Jing Li, Di-Long Chen and Jian-Hua Ran
Molecules 2021, 26(7), 1990; https://doi.org/10.3390/molecules26071990 - 01 Apr 2021
Cited by 14 | Viewed by 4965
Abstract
The urea cycle (UC) removes the excess nitrogen and ammonia generated by nitrogen-containing compound composites or protein breakdown in the human body. Research has shown that changes in UC enzymes are not only related to tumorigenesis and tumor development but also associated with [...] Read more.
The urea cycle (UC) removes the excess nitrogen and ammonia generated by nitrogen-containing compound composites or protein breakdown in the human body. Research has shown that changes in UC enzymes are not only related to tumorigenesis and tumor development but also associated with poor survival in hepatocellular, breast, and colorectal cancers (CRC), etc. Cytoplasmic ornithine, the intermediate product of the urea cycle, is a specific substrate for ornithine decarboxylase (ODC, also known as ODC1) for the production of putrescine and is required for tumor growth. Polyamines (spermidine, spermine, and their precursor putrescine) play central roles in more than half of the steps of colorectal tumorigenesis. Given the close connection between polyamines and cancer, the regulation of polyamine metabolic pathways has attracted attention regarding the mechanisms of action of chemical drugs used to prevent CRC, as the drug most widely used for treating type 2 diabetes (T2D), metformin (Met) exhibits antitumor activity against a variety of cancer cells, with a vaguely defined mechanism. In addition, the influence of metformin on the UC and putrescine generation in colorectal cancer has remained unclear. In our study, we investigated the effect of metformin on the UC and putrescine generation of CRC in vivo and in vitro and elucidated the underlying mechanisms. In nude mice bearing HCT116 tumor xenografts, the administration of metformin inhibited tumor growth without affecting body weight. In addition, metformin treatment increased the expression of monophosphate (AMP)-activated protein kinase (AMPK) and p53 in both HCT116 xenografts and colorectal cancer cell lines and decreased the expression of the urea cycle enzymes, including carbamoyl phosphate synthase 1 (CPS1), arginase 1 (ARG1), ornithine trans-carbamylase (OTC), and ODC. The putrescine levels in both HCT116 xenografts and HCT116 cells decreased after metformin treatment. These results demonstrate that metformin inhibited CRC cell proliferation via activating AMPK/p53 and that there was an association between metformin, urea cycle inhibition and a reduction in putrescine generation. Full article
(This article belongs to the Special Issue Targeting of Signaling Pathways for Cancer Therapy)
Show Figures

Graphical abstract

16 pages, 2233 KiB  
Article
EGCG Inhibits Adipose-Derived Mesenchymal Stem Cells Differentiation into Adipocytes and Prevents a STAT3-Mediated Paracrine Oncogenic Control of Triple-Negative Breast Cancer Cell Invasive Phenotype
by Narjara Gonzalez Suarez, Sahily Rodriguez Torres, Amira Ouanouki, Layal El Cheikh-Hussein and Borhane Annabi
Molecules 2021, 26(6), 1506; https://doi.org/10.3390/molecules26061506 - 10 Mar 2021
Cited by 23 | Viewed by 3261
Abstract
Obese subjects have an increased risk of developing triple-negative breast cancer (TNBC), in part associated with the chronic low-grade inflammation state. On the other hand, epidemiological data indicates that increased consumption of polyphenol-rich fruits and vegetables plays a key role in reducing incidence [...] Read more.
Obese subjects have an increased risk of developing triple-negative breast cancer (TNBC), in part associated with the chronic low-grade inflammation state. On the other hand, epidemiological data indicates that increased consumption of polyphenol-rich fruits and vegetables plays a key role in reducing incidence of some cancer types. Here, we tested whether green tea-derived epigallocatechin-3-gallate (EGCG) could alter adipose-derived mesenchymal stem cell differentiation into adipocytes, and how this impacts the secretome profile and paracrine regulation of the TNBC invasive phenotype. Here, cell differentiation was performed and conditioned media (CM) from preadipocytes and mature adipocytes harvested. Human TNBC-derived MDA-MB-231 real-time cell migration was performed using the exCELLigence system. Differential gene arrays and RT-qPCR were used to assess gene expression levels. Western blotting was used to assess protein expression and phosphorylation status levels. In vitro vasculogenic mimicry (VM) was assessed with Matrigel. EGCG was found to inhibit the induction of key adipogenic biomarkers, including lipoprotein lipase, adiponectin, leptin, fatty acid synthase, and fatty acid binding protein 4. Increased TNBC-derived MDA-MB-231 cell chemotaxis and vasculogenic mimicry were observed in response to mature adipocytes secretome, and this was correlated with increased STAT3 phosphorylation status. This invasive phenotype was prevented by EGCG, the JAK/STAT inhibitors Tofacitinib and AG490, as well as upon STAT3 gene silencing. In conclusion, dietary catechin-mediated interventions could, in part through the inhibition of adipogenesis and modulation of adipocytes secretome profile, prevent the onset of an obesogenic environment that favors TNBC development. Full article
(This article belongs to the Special Issue Targeting of Signaling Pathways for Cancer Therapy)
Show Figures

Figure 1

22 pages, 6995 KiB  
Article
2-APCAs, the Novel Microtubule Targeting Agents Active against Distinct Cancer Cell Lines
by Sergei Boichuk, Aigul Galembikova, Firuza Bikinieva, Pavel Dunaev, Aida Aukhadieva, Kirill Syuzov, Svetlana Zykova, Nazim Igidov, Alexander Ksenofontov and Pavel Bocharov
Molecules 2021, 26(3), 616; https://doi.org/10.3390/molecules26030616 - 25 Jan 2021
Cited by 8 | Viewed by 2562
Abstract
Microtubules are known as the most attractive molecular targets for anti-cancer drugs. However, the number of serious limitations of the microtubule targeting agents (MTAs) including poor bioavailability, adverse effects (e.g., systemic and neural toxicity), and acquired resistance after initiation of MTA-based therapy remain [...] Read more.
Microtubules are known as the most attractive molecular targets for anti-cancer drugs. However, the number of serious limitations of the microtubule targeting agents (MTAs) including poor bioavailability, adverse effects (e.g., systemic and neural toxicity), and acquired resistance after initiation of MTA-based therapy remain the driving forces to develop the novel therapeutic agents effectively targeting microtubules and exhibiting potent anti-tumor activities. Here, we report the discovery of 2-amino-pyrrole-carboxamides (2-APCAs), a novel class of MTA, which effectively inhibited the growth of the broad spectrum of cancer cell lines in vitro, including various types of breast, prostate, and non-small lung cancer (NSLC), soft tissue sarcomas (STS) (e.g., leio-, rhabdomyo-, and fibrosarcomas), osteosarcomas and gastrointestinal stromal tumors (GISTs). Importantly, 2-APCAs were also effective in cancer cell lines exhibiting resistance to certain chemotherapeutic agents, including MTAs and topoisomerase II inhibitors. The anti-proliferative effect of 2-APCAs was due to their ability to interfere with the polymerization of tubulin and thereby leading to the accumulation of tumor cells in the M-phase. As an outcome of the mitotic arrest, cancer cells underwent apoptotic cell death which was evidenced by increased expression of cleaved forms of the poly-ADP-ribose polymerase (PARP) and caspase-3 and the increased numbers of Annexin V-positive cells, as well. Among the compounds exhibiting the potent anti-cancer activities against the various cancer cell lines indicated above, 2-APCA-III was found the most active. Importantly, its cytotoxic activities correlated with its highest potency to interfere with the dynamics of tubulin polymerization and inducement of cell cycle arrest in the G2/M phase. Interestingly, the cytotoxic and tubulin polymerization activities of 2-APCAs correlated with the stability of the «tubulin—2-АРСА» complexes, illustrating the “tubulin-2-APCA-III” complex as the most stable. Molecular docking showed that the binding site for 2-АРСА-III is located in α tubulin by forming a hydrogen bond with Leu23. Of note, single-cell electrophoresis (Comet assay) data illustrated the low genotoxic activities of 2-APCAs when compared to certain anti-cancer chemotherapeutic agents. Taken together, our study describes the novel MTAs with potent anti-proliferative and pro-apoptotic activities, thereby illustrating them as a scaffold for the development of successful chemotherapeutic anti-cancer agent targeting microtubules. Full article
(This article belongs to the Special Issue Targeting of Signaling Pathways for Cancer Therapy)
Show Figures

Figure 1

20 pages, 4629 KiB  
Article
Rab GTPase Mediating Regulation of NALP3 in Colorectal Cancer
by Gülçin Tezcan, Ekaterina E. Garanina, Margarita N. Zhuravleva, Shaimaa Hamza, Albert A. Rizvanov and Svetlana F. Khaiboullina
Molecules 2020, 25(20), 4834; https://doi.org/10.3390/molecules25204834 - 20 Oct 2020
Cited by 7 | Viewed by 3200
Abstract
The NALP3 inflammasome signaling contributes to inflammation within tumor tissues. This inflammation may be promoted by the vesicle trafficking of inflammasome components and cytokines. Rab5, Rab7 and Rab11 regulate vesicle trafficking. However, the role of these proteins in the regulation of inflammasomes remains [...] Read more.
The NALP3 inflammasome signaling contributes to inflammation within tumor tissues. This inflammation may be promoted by the vesicle trafficking of inflammasome components and cytokines. Rab5, Rab7 and Rab11 regulate vesicle trafficking. However, the role of these proteins in the regulation of inflammasomes remains largely unknown. To elucidate the role of these Rab proteins in inflammasome regulation, HCT-116, a colorectal cancer (CRC) cell line expressing pDsRed-Rab5 wild type (WT), pDsRed-Rab5 dominant-negative (DN), pDsRed-Rab7 WT, pDsRed-Rab7 DN, pDsRed-Rab11 WT and pDsRed-Rab11 DN were treated with lipopolysaccharide (LPS)/nigericin. Inflammasome activation was analyzed by measuring the mRNA expression of NLRP3, Pro-CASP1, RAB39A and Pro-IL-1β, conducting immunofluorescence imaging and western blotting of caspase-1 and analysing the secretion levels of IL-1β using enzyme-linked immunosorbent assay (ELISA). The effects of Rabs on cytokine release were evaluated using MILLIPLEX MAP Human Cytokine/Chemokine Magnetic Bead Panel-Premixed 41 Plex. The findings showed that LPS/nigericin-treated cells expressing Rab5-WT indicated increased NALP3 expression and secretion of the IL-1β as compared to Rab5-DN cells. Caspase-1 was localized in the nucleus and cytosol of Rab5-WT cells but was localized in the cytosol in Rab5-DN cells. There were no any effects of Rab7 and Rab11 expression on the regulation of inflammasomes. Our results suggest that Rab5 may be a potential target for the regulation of NALP3 in the treatment of the CRC inflammation. Full article
(This article belongs to the Special Issue Targeting of Signaling Pathways for Cancer Therapy)
Show Figures

Figure 1

Back to TopTop