Signaling Pathway of Cancer

A special issue of Genes (ISSN 2073-4425). This special issue belongs to the section "Molecular Genetics and Genomics".

Deadline for manuscript submissions: closed (10 November 2023) | Viewed by 23315

Special Issue Editor


E-Mail Website
Guest Editor
Centre for Genome Engineering and Maintenance, Brunel University London, Kingston Lane, Uxbridge UB8 3PH, UK
Interests: cell signalling; breast cancer; tumour development; therapeutic targets; therapeutic resistance; plant-based therapies

Special Issue Information

Dear Colleagues,

The scope of this Special Issue of the journal Genes covers the broad subject that is signalling dysregulation in cancer development and progression. Communication between cell signalling pathways is essential for maintaining cellular homeostasis and aberrant signalling results in tumour progression and increased disease burden for patients. Our advanced knowledge of the contribution of signalling pathways to cancer development has been central to the advancement of targeted treatments that are now available to those suffering from cancer. Insights into the mechanisms underlying the interaction between individual pathways and their contribution to networks, as well as an understanding of their ability to compensate for each other, is vital in tackling therapeutic resistance.

Dr. Amanda Harvey
Guest Editor

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. Genes is an international peer-reviewed open access monthly 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 2600 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

  • cell signalling
  • tumour development
  • disease progression
  • novel therapies
  • therapeutic targets
  • therapeutic resistance

Published Papers (9 papers)

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

Research

Jump to: Review

15 pages, 2833 KiB  
Article
Metformin Resistance Is Associated with Expression of Inflammatory and Invasive Genes in A549 Lung Cancer Cells
by Dong Soo Seo, Sungmin Joo, Seungwoo Baek, Jaehyeon Kang, Taeg Kyu Kwon and Younghoon Jang
Genes 2023, 14(5), 1014; https://doi.org/10.3390/genes14051014 - 29 Apr 2023
Cited by 1 | Viewed by 1647
Abstract
Metformin, the most commonly used drug for type 2 diabetes, has recently been shown to have beneficial effects in patients with cancer. Despite growing evidence that metformin can inhibit tumor cell proliferation, invasion, and metastasis, studies on drug resistance and its side effects [...] Read more.
Metformin, the most commonly used drug for type 2 diabetes, has recently been shown to have beneficial effects in patients with cancer. Despite growing evidence that metformin can inhibit tumor cell proliferation, invasion, and metastasis, studies on drug resistance and its side effects are lacking. Here, we aimed to establish metformin-resistant A549 human lung cancer cells (A549-R) to determine the side effects of metformin resistance. Toward this, we established A549-R by way of prolonged treatment with metformin and examined the changes in gene expression, cell migration, cell cycle, and mitochondrial fragmentation. Metformin resistance is associated with increased G1-phase cell cycle arrest and impaired mitochondrial fragmentation in A549 cells. We demonstrated that metformin resistance highly increased the expression of proinflammatory and invasive genes, including BMP5, CXCL3, VCAM1, and POSTN, using RNA-seq analysis. A549-R exhibited increased cell migration and focal adhesion formation, suggesting that metformin resistance may potentially lead to metastasis during anti-cancer therapy with metformin. Taken together, our findings indicate that metformin resistance may lead to invasion in lung cancer cells. Full article
(This article belongs to the Special Issue Signaling Pathway of Cancer)
Show Figures

Figure 1

13 pages, 2893 KiB  
Article
SODD Promotes Lung Cancer Tumorigenesis by Activating the PDK1/AKT and RAF/MEK/ERK Signaling
by Fan Bao, Su An, Yang Yang and Tian-Rui Xu
Genes 2023, 14(4), 829; https://doi.org/10.3390/genes14040829 - 30 Mar 2023
Viewed by 1366
Abstract
Background: The Bcl2-associated athanogene4 (BAG4/SODD) protein could be identified as a tumor marker for several malignancies and plays a major role in the occurrence, development, and drug resistance of tumors. However, the role of Silencer of death domains (SODD) in lung carcinogenesis is [...] Read more.
Background: The Bcl2-associated athanogene4 (BAG4/SODD) protein could be identified as a tumor marker for several malignancies and plays a major role in the occurrence, development, and drug resistance of tumors. However, the role of Silencer of death domains (SODD) in lung carcinogenesis is still elusive. Objective: To illuminate the effect of SODD on the proliferation, migration, invasion, and apoptosis of lung cancer cells and tumor growth in vivo and explore the corresponding mechanism. Methods: The expression of SODD in tumor and normal tissues was determined and compared via western blot. SODD gene knockout lung cancer cells (H1299 cells) were established through a CRISPR/Cas9 gene deleting system, and a transient SODD overexpression of H1299 cells was also constructed. Then, cell proliferation and invasion were assessed through colony formation and cell counting kit-8 assays, transwell migration assays, and wound healing assays. Cell drug sensitivity is also analyzed by Cell Counting Kit-8 assay. The flow cytometer was used to perform cell circle and apoptosis analysis. The interaction of SODD and RAF-1 was confirmed by co-immunoprecipitation, and the phosphorylated level of Phosphatidylinositol 3-kinase (PI3K), Serine/threonine-protein kinase (AKT), Rapidly accelerated fibrosarcoma (RAF)-1,and extracellular signal regulated kinase (ERK) in cells was examined by western blot to evaluate the activation of PI3K/PDK1/AKT and RAF/MEK/ERK pathways. In vivo, Xenograft tumor assay of SODD knockout H1299 cells was used to evaluate further the role of SODD on the proliferation of H1299 cells. Results: SODD binds to RAF-1 and is over-expressed in lung tissues, and promotes the proliferation, migration, invasion, and drug sensitivity of H1299 cells. The reduced cells in the S phase and increased cells arrested in the G2/M phase were found in SODD knockout H1299 cells, and more cells got apoptosis. The expression of 3-phosphoinositide-dependent protein kinase 1(PDK1) protein in SODD knockout H1299 cells decreases distinctively, and the phosphorylated level of AKT, RAF-1, and ERK-1 kinase in SODD knockout H1299 cells is also less than that in normal H1299 cells. In contrast, SODD overexpression significantly increases the phosphorylation of AKT. In vivo, SODD promotes the tumorigenicity of H1299 cells in nude mice. Conclusions: SODD is overexpressed in lung tissues and plays a considerable role in the development and progression of lung cancer by regulating the PI3K/PDK1/AKT and RAF/MEK/ERK pathways. Full article
(This article belongs to the Special Issue Signaling Pathway of Cancer)
Show Figures

Figure 1

10 pages, 2316 KiB  
Article
Casticin Impacts Key Signaling Pathways in Colorectal Cancer Cells Leading to Cell Death with Therapeutic Implications
by Michael Kowalski, Ashley Assa, Ketki Patil, Courtney Terrell, Nathan Holliday and S. Balakrishna Pai
Genes 2022, 13(5), 815; https://doi.org/10.3390/genes13050815 - 03 May 2022
Cited by 2 | Viewed by 1670
Abstract
Colorectal cancer is the third most frequently encountered cancer worldwide. While current chemotherapeutics help to manage the disease to some extent, they have eluded achieving complete remission and are limited by their severe side effects. This warrants exploration of novel agents that are [...] Read more.
Colorectal cancer is the third most frequently encountered cancer worldwide. While current chemotherapeutics help to manage the disease to some extent, they have eluded achieving complete remission and are limited by their severe side effects. This warrants exploration of novel agents that are efficacious with anticipation of minimal adverse effects. In the current study, casticin, a tetramethoxyflavone, was tested for its ability to inhibit the viability of three human colorectal cancer cells: adenocarcinoma (DLD-1, Caco-2 cell lines) and human colorectal carcinoma cells (HCT116 cell line). Casticin showed potent inhibition of viability of DLD-1 and HCT116 cells. Clonogenic assay performed in DLD-1 cells revealed that casticin impeded the colony-forming efficiency of the cells, suggesting its impact on the proliferation of these cells. Further, a sustained effect of the inhibitory action upon withdrawal of the treatment was observed. Elucidation of the mechanism of action revealed that casticin impacted the extrinsic programmed cell death pathway, leading to an increase in apoptotic cells. Further, Bcl-2, the key moiety of cell survival, was affected. Notably, a significant number of cells were arrested in the G2/M phase of the cell cycle in DLD-1 cells. Due to the multifaceted action of casticin, we envision that treatment with casticin could provide an efficacious treatment option for colorectal adenocarcinomas with minimal side effects. Full article
(This article belongs to the Special Issue Signaling Pathway of Cancer)
Show Figures

Figure 1

13 pages, 2682 KiB  
Article
Breast Tumour Kinase (Brk/PTK6) Contributes to Breast Tumour Xenograft Growth and Modulates Chemotherapeutic Responses In Vitro
by Rajpal S. Burmi, Gary M. Box, Umar Wazir, Haroon A. Hussain, Julie A. Davies, William J. Court, Suzanne A. Eccles, Wen G. Jiang, Kefah Mokbel and Amanda J. Harvey
Genes 2022, 13(3), 402; https://doi.org/10.3390/genes13030402 - 24 Feb 2022
Cited by 2 | Viewed by 2032
Abstract
Breast tumour kinase (Brk/PTK6) is overexpressed in up to 86% of breast cancers and is associated with poorer patient outcomes. It is considered a potential therapeutic target in breast cancer, even though the full spectrum of its kinase activity is not known. This [...] Read more.
Breast tumour kinase (Brk/PTK6) is overexpressed in up to 86% of breast cancers and is associated with poorer patient outcomes. It is considered a potential therapeutic target in breast cancer, even though the full spectrum of its kinase activity is not known. This study investigated the role of the kinase domain in promoting tumour growth and its potential in sensitising triple negative breast cancer cells to standard of care chemotherapy. Triple negative human xenograft models revealed that both kinase-inactive and wild-type Brk promoted xenograft growth. Suppression of Brk activity in cells subsequently co-treated with the chemotherapy agents doxorubicin or paclitaxel resulted in an increased cell sensitivity to these agents. In triple negative breast cancer cell lines, the inhibition of Brk kinase activity augmented the effects of doxorubicin or paclitaxel. High expression of the alternatively spliced isoform, ALT-PTK6, resulted in improved patient outcomes. Our study is the first to show a role for kinase-inactive Brk in human breast tumour xenograft growth; therefore, it is unlikely that kinase inhibition of Brk, in isolation, would halt tumour growth in vivo. Breast cancer cell responses to chemotherapy in vitro were kinase-dependent, indicating that treatment with kinase inhibitors could be a fruitful avenue for combinatorial treatment. Of particular prognostic value is the ratio of ALT-PTK6:Brk expression in predicating patient outcomes. Full article
(This article belongs to the Special Issue Signaling Pathway of Cancer)
Show Figures

Figure 1

12 pages, 2883 KiB  
Article
The Adenoviral E1B-55k Protein Present in HEK293 Cells Mediates Abnormal Accumulation of Key WNT Signaling Proteins in Large Cytoplasmic Aggregates
by Petter Angell Olsen and Stefan Krauss
Genes 2021, 12(12), 1920; https://doi.org/10.3390/genes12121920 - 29 Nov 2021
Cited by 3 | Viewed by 2458
Abstract
HEK293 cells are one of the most widely used cell lines in research, and HEK293 cells are frequently used as an in vitro model for studying the WNT signaling pathway. The HEK293 cell line was originally established by transfection of human embryonic kidney [...] Read more.
HEK293 cells are one of the most widely used cell lines in research, and HEK293 cells are frequently used as an in vitro model for studying the WNT signaling pathway. The HEK293 cell line was originally established by transfection of human embryonic kidney cells with sheared adenovirus 5 DNA, and it is known that that HEK293 cells stably express the adenoviral E1A and E1B-55k proteins. Here, we show that HEK293 cells display an unexpected distribution of key components of the WNT/β-catenin signaling pathway where AXIN1, APC, DVL2 and tankyrase are all co-localized in large spherical cytoplasmic aggregates. The cytoplasmic aggregates are enclosed by a narrow layer of the adenoviral E1B-55k protein. The reduction of E1B-55k protein levels leads to the disappearance of the cytoplasmic aggregates thus corroborating an essential role of the E1B-55k protein in mediating the formation of the aggregates. Furthermore, HEK293 cells with reduced E1B-55k protein levels display reduced levels of transcriptional activation of WNT/β-catenin signaling upon stimulation by the Wnt3A agonist. The demonstrated influence of the E1B-55k protein on the cellular localization of WNT/β-catenin signaling components and on transcriptional regulation of WNT/β-catenin signaling asks for caution in the interpretation of data derived from the HEK293 cell line. Full article
(This article belongs to the Special Issue Signaling Pathway of Cancer)
Show Figures

Figure 1

Review

Jump to: Research

26 pages, 3106 KiB  
Review
Sex Differences in Colon Cancer: Genomic and Nongenomic Signalling of Oestrogen
by Brian J. Harvey and Harry M. Harvey
Genes 2023, 14(12), 2225; https://doi.org/10.3390/genes14122225 - 16 Dec 2023
Viewed by 1841
Abstract
Colon cancer (CRC) is a prevalent malignancy that exhibits distinct differences in incidence, prognosis, and treatment responses between males and females. These disparities have long been attributed to hormonal differences, particularly the influence of oestrogen signalling. This review aims to provide a comprehensive [...] Read more.
Colon cancer (CRC) is a prevalent malignancy that exhibits distinct differences in incidence, prognosis, and treatment responses between males and females. These disparities have long been attributed to hormonal differences, particularly the influence of oestrogen signalling. This review aims to provide a comprehensive analysis of recent advances in our understanding of the molecular mechanisms underlying sex differences in colon cancer and the protective role of membrane and nuclear oestrogen signalling in CRC development, progression, and therapeutic interventions. We discuss the epidemiological and molecular evidence supporting sex differences in colon cancer, followed by an exploration of the impact of oestrogen in CRC through various genomic and nongenomic signalling pathways involving membrane and nuclear oestrogen receptors. Furthermore, we examine the interplay between oestrogen receptors and other signalling pathways, in particular the Wnt/β-catenin proliferative pathway and hypoxia in shaping biological sex differences and oestrogen protective actions in colon cancer. Lastly, we highlight the potential therapeutic implications of targeting oestrogen signalling in the management of colon cancer and propose future research directions to address the current gaps in our understanding of this complex phenomenon. Full article
(This article belongs to the Special Issue Signaling Pathway of Cancer)
Show Figures

Graphical abstract

31 pages, 1433 KiB  
Review
JAK/STAT Signaling and Cervical Cancer: From the Cell Surface to the Nucleus
by Arturo Valle-Mendiola, Adriana Gutiérrez-Hoya and Isabel Soto-Cruz
Genes 2023, 14(6), 1141; https://doi.org/10.3390/genes14061141 - 24 May 2023
Cited by 5 | Viewed by 3592
Abstract
The Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signaling pathway constitutes a rapid signaling module from the cell surface to the nucleus, and activates different cellular responses, such as proliferation, survival, migration, invasion, and inflammation. When the JAK/STAT pathway is altered, [...] Read more.
The Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signaling pathway constitutes a rapid signaling module from the cell surface to the nucleus, and activates different cellular responses, such as proliferation, survival, migration, invasion, and inflammation. When the JAK/STAT pathway is altered, it contributes to cancer progression and metastasis. STAT proteins play a central role in developing cervical cancer, and inhibiting the JAK/STAT signaling may be necessary to induce tumor cell death. Several cancers show continuous activation of different STATs, including cervical cancer. The constitutive activation of STAT proteins is associated with a poor prognosis and overall survival. The human papillomavirus (HPV) oncoproteins E6 and E7 play an essential role in cervical cancer progression, and they activate the JAK/STAT pathway and other signals that induce proliferation, survival, and migration of cancer cells. Moreover, there is a crosstalk between the JAK/STAT signaling cascade with other signaling pathways, where a plethora of different proteins activate to induce gene transcription and cell responses that contribute to tumor growth. Therefore, inhibition of the JAK/STAT pathway shows promise as a new target in cancer treatment. In this review, we discuss the role of the JAK/STAT pathway components and the role of the HPV oncoproteins associated with cellular malignancy through the JAK/STAT proteins and other signaling pathways to induce tumor growth. Full article
(This article belongs to the Special Issue Signaling Pathway of Cancer)
Show Figures

Figure 1

20 pages, 1320 KiB  
Review
Crosstalk between Hypoxia and Extracellular Matrix in the Tumor Microenvironment in Breast Cancer
by Yasmin Dekker, Sylvia E. Le Dévédec, Erik H. J. Danen and Qiuyu Liu
Genes 2022, 13(9), 1585; https://doi.org/10.3390/genes13091585 - 03 Sep 2022
Cited by 19 | Viewed by 3464
Abstract
Even though breast cancer is the most diagnosed cancer among women, treatments are not always successful in preventing its progression. Recent studies suggest that hypoxia and the extracellular matrix (ECM) are important in altering cell metabolism and tumor metastasis. Therefore, the aim of [...] Read more.
Even though breast cancer is the most diagnosed cancer among women, treatments are not always successful in preventing its progression. Recent studies suggest that hypoxia and the extracellular matrix (ECM) are important in altering cell metabolism and tumor metastasis. Therefore, the aim of this review is to study the crosstalk between hypoxia and the ECM and to assess their impact on breast cancer progression. The findings indicate that hypoxic signaling engages multiple mechanisms that directly contribute to ECM remodeling, ultimately increasing breast cancer aggressiveness. Second, hypoxia and the ECM cooperate to alter different aspects of cell metabolism. They mutually enhance aerobic glycolysis through upregulation of glucose transport, glycolytic enzymes, and by regulating intracellular pH. Both alter lipid and amino acid metabolism by stimulating lipid and amino acid uptake and synthesis, thereby providing the tumor with additional energy for growth and metastasis. Third, YAP/TAZ signaling is not merely regulated by the tumor microenvironment and cell metabolism, but it also regulates it primarily through its target c-Myc. Taken together, this review provides a better understanding of the crosstalk between hypoxia and the ECM in breast cancer. Additionally, it points to a role for the YAP/TAZ mechanotransduction pathway as an important link between hypoxia and the ECM in the tumor microenvironment, driving breast cancer progression. Full article
(This article belongs to the Special Issue Signaling Pathway of Cancer)
Show Figures

Figure 1

16 pages, 996 KiB  
Review
Synthetic Lethality Targeting Polθ
by Małgorzata Drzewiecka, Gabriela Barszczewska-Pietraszek, Piotr Czarny, Tomasz Skorski and Tomasz Śliwiński
Genes 2022, 13(6), 1101; https://doi.org/10.3390/genes13061101 - 20 Jun 2022
Cited by 9 | Viewed by 3880
Abstract
Research studies regarding synthetic lethality (SL) in human cells are primarily motivated by the potential of this phenomenon to be an effective, but at the same time, safe to the patient’s anti-cancer chemotherapy. Among the factors that are targets for the induction of [...] Read more.
Research studies regarding synthetic lethality (SL) in human cells are primarily motivated by the potential of this phenomenon to be an effective, but at the same time, safe to the patient’s anti-cancer chemotherapy. Among the factors that are targets for the induction of the synthetic lethality effect, those involved in DNA repair seem to be the most relevant. Specifically, when mutation in one of the canonical DNA double-strand break (DSB) repair pathways occurs, which is a frequent event in cancer cells, the alternative pathways may be a promising target for the elimination of abnormal cells. Currently, inhibiting RAD52 and/or PARP1 in the tumor cells that are deficient in the canonical repair pathways has been the potential target for inducing the effect of synthetic lethality. Unfortunately, the development of resistance to commonly used PARP1 inhibitors (PARPi) represents the greatest obstacle to working out a successful treatment protocol. DNA polymerase theta (Polθ), encoded by the POLQ gene, plays a key role in an alternative DSB repair pathway—theta-mediated end joining (TMEJ). Thus, it is a promising target in the treatment of tumors harboring deficiencies in homologous recombination repair (HRR), where its inhibition can induce SL. In this review, the authors discuss the current state of knowledge on Polθ as a potential target for synthetic lethality-based anticancer therapies. Full article
(This article belongs to the Special Issue Signaling Pathway of Cancer)
Show Figures

Figure 1

Back to TopTop