Molecular Mechanisms of Tumor Development and Progression - A Themed Honorary Issue to Prof. Miguel Quintanilla

A special issue of Cells (ISSN 2073-4409). This special issue belongs to the section "Cellular Pathology".

Deadline for manuscript submissions: closed (15 January 2022) | Viewed by 30838

Special Issue Editors


E-Mail Website
Guest Editor
Molecular Biology Department, Science Center, School of Osteopathic Medicine, Rowan University, 2 Medical Center Dr., Stratford, NJ 08084, USA
Interests: cell communication; signal transduction; cancer; inflammation

E-Mail Website
Guest Editor
Department of Biotechnology, Universidad Francisco de Vitoria, Carretera Pozuelo a Majadahonda, Km 1.800, 28223 Madrid, Spain
Interests: carcinogenesis; tumour invasion; skin homeostasis

Special Issue Information

Dear Colleagues,

Cancer is still considered one of the leading causes of death worldwide due to tumour progression triggering metastatic dissemination. Although this continues to be a sad scenario, looking back at past decades, one realises that huge improvements have been made in the field of cancer research. These advances include the discovery of oncogenes in the early 1970s, genetic and epigenetic mechanisms associated with carcinogenesis, the collaboration of tumour and host cells during the metastatic process, nanotechnology, immunotherapy and cancer organoid technology development for personalised patient treatment design. Many dedicated cancer researchers have made and are making outstanding contributions to understand and fight this complex multifactorial disease.

In 1986, Miguel Quintanilla, then as a postdoc in the lab of Allan Balmain, published a seminal paper identifying a genomic mutation in a “cancer driver” gene, which has an important role in tumour development. He described the well-known A→T transversion at the second nucleotide of codon 61 of the Harvey-ras (Ha-ras) gene, which was promoted by the exposure of mouse skin to the chemical carcinogen dimethylbenz-anthracene (DMBA). Using this classical model of mouse skin chemical carcinogenesis, Miguel Quintanilla has also made other significant contributions in the field of epithelial carcinogenesis. His findings describe the anti-invasive role of the cell–cell adhesion protein E-cadherin, whose loss is associated with tumor progression (alongside Amparo Cano and Carlos Gamallo). He also performed pioneering work to find that TGF-beta acts as a stimulator of epithelial-mesenchymal transition (EMT) associated with progression to poorly differentiated carcinomas, and the identification of the TGF-beta co-receptor endoglin as a suppressor of malignancy in epithelial carcinogenesis by a mechanism involving inhibition of TGF-beta signaling. Miguel´s work has also been a driving force towards the discovery of podoplanin (PA2.26), a small mucin-type transmembrane glycoprotein, as a promoter of malignancy in epithelial cancer by a variety of strategies including EMT induction, cooperating with CD44 to stimulate directional migration, and facilitating an efficient degradation of the extracellular matrix and stabilising invadopodia (alongside Gareth E. Jones an Ester Martin-Villar).

This Special Issue of Cells focuses on the molecular mechanisms underlying carcinogenesis and tumour progression related to the pioneering work of Miguel Quintanilla. Although genomic mutations and epigenetic changes are indisputably important during carcinogenesis, Miguel´s contributions have also brought out the importance of invasion promoting factors that facilitate tumour progression through mechanisms that do not necessarily involve DNA damage. This Special Issue intends to serve as a tribute to his work, gathering original research and timely reviews that consolidate our current knowledge of molecular mechanisms underlying cancer and open new fronts in the fight against this devastating disease.

text

Miguel Quintanilla, born in the monumental city of Toledo (Spain), earned his bachelor’s degree in Chemistry from the Complutense University of Madrid in 1974, and his PhD in Biochemistry from the University of Valladolid in 1980. As a postdoctoral researcher, he first joined Dr. Jaime Renart's lab at the Institute of Enzymology and Molecular Pathology (now The Institute of Biomedical Research of Madrid, IIBm) of the Spanish National Research Council (CSIC), and then moved to Scotland, joining Dr. Allan Balmain's lab at the Beatson Institute for Cancer Research (Glasgow), sponsored by the European Molecular Biology Organization (EMBO) fellowship (1984–1986). In 1987, he returned to IIBm in Spain and has been there ever since, initially as Tenured Scientist, then as a Research Professor, running the Epithelial Carcinogenesis laboratory until his retirement in 2021, as professor “ad honorem”. Miguel has always been able to follow the right scientific pathway, which has always been characterised by a true love for science, hard work, sense of humour, honesty, and generosity.

Dr. Gary S. Goldberg
Dr. Ester Martín-Villar
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. Cells 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.

Published Papers (8 papers)

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

Research

Jump to: Review

17 pages, 3008 KiB  
Article
The E1a Adenoviral Gene Upregulates the Yamanaka Factors to Induce Partial Cellular Reprogramming
by Gracia Mendoza, Rebeca González-Pastor, Juan Miguel Sánchez, Altamira Arce-Cerezo, Miguel Quintanilla, Gema Moreno-Bueno, Anna Pujol, Carolina Belmar-López, Alba de Martino, Efrén Riu, Tristan A. Rodriguez and Pilar Martin-Duque
Cells 2023, 12(9), 1338; https://0-doi-org.brum.beds.ac.uk/10.3390/cells12091338 - 07 May 2023
Viewed by 2198
Abstract
The induction of pluripotency by enforced expression of different sets of genes in somatic cells has been achieved with reprogramming technologies first described by Yamanaka’s group. Methodologies for generating induced pluripotent stem cells are as varied as the combinations of genes used. It [...] Read more.
The induction of pluripotency by enforced expression of different sets of genes in somatic cells has been achieved with reprogramming technologies first described by Yamanaka’s group. Methodologies for generating induced pluripotent stem cells are as varied as the combinations of genes used. It has previously been reported that the adenoviral E1a gene can induce the expression of two of the Yamanaka factors (c-Myc and Oct-4) and epigenetic changes. Here, we demonstrate that the E1a-12S over-expression is sufficient to induce pluripotent-like characteristics closely to epiblast stem cells in mouse embryonic fibroblasts through the activation of the pluripotency gene regulatory network. These findings provide not only empirical evidence that the expression of one single factor is sufficient for partial reprogramming but also a potential mechanistic explanation for how viral infection could lead to neoplasia if they are surrounded by the appropriate environment or the right medium, as happens with the tumorogenic niche. Full article
Show Figures

Figure 1

17 pages, 3004 KiB  
Article
Transcriptomic Response to Acidosis Reveals Its Contribution to Bone Metastasis in Breast Cancer Cells
by Ana Sayuri Yamagata, Paula Paccielli Freire, Nícolas Jones Villarinho, Ramon Handerson Gomes Teles, Kelliton José Mendonça Francisco, Ruy Gastaldoni Jaeger and Vanessa Morais Freitas
Cells 2022, 11(3), 544; https://0-doi-org.brum.beds.ac.uk/10.3390/cells11030544 - 04 Feb 2022
Cited by 3 | Viewed by 1887
Abstract
Bone is the most common site of metastasis in breast cancer. Metastasis is promoted by acidosis, which is associated with osteoporosis. To investigate how acidosis could promote bone metastasis, we compared differentially expressed genes (DEGs) in MDA-MB-231 cancer cells in acidosis, bone metastasis, [...] Read more.
Bone is the most common site of metastasis in breast cancer. Metastasis is promoted by acidosis, which is associated with osteoporosis. To investigate how acidosis could promote bone metastasis, we compared differentially expressed genes (DEGs) in MDA-MB-231 cancer cells in acidosis, bone metastasis, and bone metastatic tumors. The DEGs were identified using Biojupies and GEO2R. The expression profiles were assessed with Morpheus. The overlapping DEGs between acidosis and bone metastasis were compared to the bulk of the DEGs in terms of the most important genes and enriched terms using CytoHubba and STRING. The expression of the genes in this overlap filtered by secreted proteins was assessed in the osteoporosis secretome. The analysis revealed that acidosis-associated transcriptomic changes were more similar to bone metastasis than bone metastatic tumors. Extracellular matrix (ECM) organization would be the main biological process shared between acidosis and bone metastasis. The secretome genes upregulated in acidosis, bone metastasis, and osteoporosis-associated mesenchymal stem cells are enriched for ECM organization and angiogenesis. Therefore, acidosis may be more important in the metastatic niche than in the primary tumor. Acidosis may contribute to bone metastasis by promoting ECM organization. Untreated osteoporosis could favor bone metastasis through the increased secretion of ECM organization proteins. Full article
Show Figures

Figure 1

15 pages, 2774 KiB  
Article
Comparison of Colorectal Cancer Stem Cells and Oxaliplatin-Resistant Cells Unveils Functional Similarities
by Vanessa Rodríguez-Fanjul, Rosa Guerrero-López, Beatriz Fernández-Varas, Rosario Perona, Ana Sastre-Perona and Leandro Sastre
Cells 2022, 11(3), 511; https://0-doi-org.brum.beds.ac.uk/10.3390/cells11030511 - 01 Feb 2022
Cited by 7 | Viewed by 1979
Abstract
Colorectal cancer is the second most common cancer in women, the third in men, and an important cause of cancer-related mortality. Recurrence and the development of chemotherapy resistance are major hindrances for patients’ treatment. The presence of cancer stem cells with chemotherapy resistance [...] Read more.
Colorectal cancer is the second most common cancer in women, the third in men, and an important cause of cancer-related mortality. Recurrence and the development of chemotherapy resistance are major hindrances for patients’ treatment. The presence of cancer stem cells with chemotherapy resistance able to generate proliferating tumor cells contributes to tumor recurrence and resistance. In addition, tumor cells can develop chemoresistance through adaptation mechanisms. In this article, cancer stem cells were isolated from HT29 and SW620 colorectal cancer cell lines. Oxaliplatin resistance was induced by a single drug treatment simulating the usual guidelines of patient treatment. A comparison of these two populations showed similarities since cancer stem cells presented increased oxaliplatin resistance, and resistant cells contained an increased number of cancer stem cells. Cancer stem cells isolated from resistant cells showed increased oxaliplatin resistance. Cell invasion capacity and epithelial-mesenchymal transition were increased both in cancer stem cells and oxaliplatin-resistant cells. mRNA expression analysis showed that both cell types shared a significant proportion of commonly regulated genes. In summary, the data presented indicate that colorectal cancer stem cells and oxaliplatin-resistant cells are highly related cell populations that might have interesting implications in the development of tumor recurrence and resistance to chemotherapy. Full article
Show Figures

Figure 1

16 pages, 3648 KiB  
Article
Defucosylated Mouse–Dog Chimeric Anti-EGFR Antibody Exerts Antitumor Activities in Mouse Xenograft Models of Canine Tumors
by Guanjie Li, Tomokazu Ohishi, Mika K. Kaneko, Junko Takei, Takuya Mizuno, Manabu Kawada, Masaki Saito, Hiroyuki Suzuki and Yukinari Kato
Cells 2021, 10(12), 3599; https://0-doi-org.brum.beds.ac.uk/10.3390/cells10123599 - 20 Dec 2021
Cited by 7 | Viewed by 3155
Abstract
The epidermal growth factor receptor (EGFR) contributes to tumor malignancy via gene amplification and protein overexpression. Previously, we developed an anti-human EGFR (hEGFR) monoclonal antibody, namely EMab-134, which detects hEGFR and dog EGFR (dEGFR) with high sensitivity and specificity. In this study, we [...] Read more.
The epidermal growth factor receptor (EGFR) contributes to tumor malignancy via gene amplification and protein overexpression. Previously, we developed an anti-human EGFR (hEGFR) monoclonal antibody, namely EMab-134, which detects hEGFR and dog EGFR (dEGFR) with high sensitivity and specificity. In this study, we produced a defucosylated mouse–dog chimeric anti-EGFR monoclonal antibody, namely E134Bf. In vitro analysis revealed that E134Bf highly exerted antibody-dependent cellular cytotoxicity and complement-dependent cytotoxicity against a canine osteosarcoma cell line (D-17) and a canine fibroblastic cell line (A-72), both of which express endogenous dEGFR. Moreover, in vivo administration of E134Bf significantly suppressed the development of D-17 and A-72 compared with the control dog IgG in mouse xenografts. These results indicate that E134Bf exerts antitumor effects against dEGFR-expressing canine cancers and could be valuable as part of an antibody treatment regimen for dogs. Full article
Show Figures

Figure 1

15 pages, 1865 KiB  
Article
Preclinical Evaluation of Podoplanin-Targeted Alpha-Radioimmunotherapy with the Novel Antibody NZ-16 for Malignant Mesothelioma
by Hitomi Sudo, Atsushi B. Tsuji, Aya Sugyo, Mika K. Kaneko, Yukinari Kato, Kotaro Nagatsu, Hisashi Suzuki and Tatsuya Higashi
Cells 2021, 10(10), 2503; https://0-doi-org.brum.beds.ac.uk/10.3390/cells10102503 - 22 Sep 2021
Cited by 11 | Viewed by 2836
Abstract
The prognosis of advanced mesothelioma is poor. Podoplanin (PDPN) is highly expressed in most malignant mesothelioma. This study aimed to evaluate the potential alpha-radioimmunotherapy (RIT) with a newly developed anti-PDPN antibody, NZ-16, compared with a previous antibody, NZ-12. Methods: The in vitro properties [...] Read more.
The prognosis of advanced mesothelioma is poor. Podoplanin (PDPN) is highly expressed in most malignant mesothelioma. This study aimed to evaluate the potential alpha-radioimmunotherapy (RIT) with a newly developed anti-PDPN antibody, NZ-16, compared with a previous antibody, NZ-12. Methods: The in vitro properties of radiolabeled antibodies were evaluated by cell binding and competitive inhibition assays using PDPN-expressing H226 mesothelioma cells. The biodistribution of 111In-labeled antibodies was studied in tumor-bearing mice. The absorbed doses were estimated based on biodistribution data. Tumor volumes and body weights of mice treated with 90Y- and 225Ac-labeled NZ-16 were measured for 56 days. Histologic analysis was conducted. Results: The radiolabeled NZ-16 specifically bound to H226 cells with higher affinity than NZ-12. The biodistribution studies showed higher tumor uptake of radiolabeled NZ-16 compared with NZ-12, providing higher absorbed doses to tumors. RIT with 225Ac- and 90Y-labeled NZ-16 had a significantly higher antitumor effect than RIT with 90Y-labeled NZ-12. 225Ac-labeled NZ-16 induced a larger amount of necrotic change and showed a tendency to suppress tumor volumes and prolonged survival than 90Y-labeled NZ-16. There is no obvious adverse effect. Conclusions: Alpha-RIT with the newly developed NZ-16 is a promising therapeutic option for malignant mesothelioma. Full article
Show Figures

Graphical abstract

Review

Jump to: Research

17 pages, 1413 KiB  
Review
Epithelial Mesenchymal Transition (EMT) and Associated Invasive Adhesions in Solid and Haematological Tumours
by David Greaves and Yolanda Calle
Cells 2022, 11(4), 649; https://0-doi-org.brum.beds.ac.uk/10.3390/cells11040649 - 13 Feb 2022
Cited by 25 | Viewed by 3941
Abstract
In solid tumours, cancer cells that undergo epithelial mesenchymal transition (EMT) express characteristic gene expression signatures that promote invasive migration as well as the development of stemness, immunosuppression and drug/radiotherapy resistance, contributing to the formation of currently untreatable metastatic tumours. The cancer traits [...] Read more.
In solid tumours, cancer cells that undergo epithelial mesenchymal transition (EMT) express characteristic gene expression signatures that promote invasive migration as well as the development of stemness, immunosuppression and drug/radiotherapy resistance, contributing to the formation of currently untreatable metastatic tumours. The cancer traits associated with EMT can be controlled by the signalling nodes at characteristic adhesion sites (focal contacts, invadopodia and microtentacles) where the regulation of cell migration, cell cycle progression and pro-survival signalling converge. In haematological tumours, ample evidence accumulated during the last decade indicates that the development of an EMT-like phenotype is indicative of poor disease prognosis. However, this EMT phenotype has not been directly linked to the assembly of specific forms of adhesions. In the current review we discuss the role of EMT in haematological malignancies and examine its possible link with the progression towards more invasive and aggressive forms of these tumours. We also review the known types of adhesions formed by haematological malignancies and speculate on their possible connection with the EMT phenotype. We postulate that understanding the architecture and regulation of EMT-related adhesions will lead to the discovery of new therapeutic interventions to overcome disease progression and resistance to therapies. Full article
Show Figures

Figure 1

24 pages, 1094 KiB  
Review
Roles of Podoplanin in Malignant Progression of Tumor
by Hiroyuki Suzuki, Mika K. Kaneko and Yukinari Kato
Cells 2022, 11(3), 575; https://0-doi-org.brum.beds.ac.uk/10.3390/cells11030575 - 07 Feb 2022
Cited by 36 | Viewed by 8146
Abstract
Podoplanin (PDPN) is a cell-surface mucin-like glycoprotein that plays a critical role in tumor development and normal development of the lung, kidney, and lymphatic vascular systems. PDPN is overexpressed in several tumors and is involved in their malignancy. PDPN induces platelet aggregation through [...] Read more.
Podoplanin (PDPN) is a cell-surface mucin-like glycoprotein that plays a critical role in tumor development and normal development of the lung, kidney, and lymphatic vascular systems. PDPN is overexpressed in several tumors and is involved in their malignancy. PDPN induces platelet aggregation through binding to platelet receptor C-type lectin-like receptor 2. Furthermore, PDPN modulates signal transductions that regulate cell proliferation, differentiation, migration, invasion, epithelial-to-mesenchymal transition, and stemness, all of which are crucial for the malignant progression of tumor. In the tumor microenvironment (TME), PDPN expression is upregulated in the tumor stroma, including cancer-associated fibroblasts (CAFs) and immune cells. CAFs play significant roles in the extracellular matrix remodeling and the development of immunosuppressive TME. Additionally, PDPN functions as a co-inhibitory molecule on T cells, indicating its involvement with immune evasion. In this review, we describe the mechanistic basis and diverse roles of PDPN in the malignant progression of tumors and discuss the possibility of the clinical application of PDPN-targeted cancer therapy, including cancer-specific monoclonal antibodies, and chimeric antigen receptor T technologies. Full article
Show Figures

Figure 1

32 pages, 1613 KiB  
Review
Mechanisms of Cisplatin Resistance in HPV Negative Head and Neck Squamous Cell Carcinomas
by Ana Belén Griso, Lucía Acero-Riaguas, Beatriz Castelo, José Luis Cebrián-Carretero and Ana Sastre-Perona
Cells 2022, 11(3), 561; https://0-doi-org.brum.beds.ac.uk/10.3390/cells11030561 - 05 Feb 2022
Cited by 18 | Viewed by 4087
Abstract
Head and neck squamous cell carcinomas (HNSCCs) are the eighth most common cancers worldwide. While promising new therapies are emerging, cisplatin-based chemotherapy remains the gold standard for advanced HNSCCs, although most of the patients relapse due to the development of resistance. This review [...] Read more.
Head and neck squamous cell carcinomas (HNSCCs) are the eighth most common cancers worldwide. While promising new therapies are emerging, cisplatin-based chemotherapy remains the gold standard for advanced HNSCCs, although most of the patients relapse due to the development of resistance. This review aims to condense the different mechanisms involved in the development of cisplatin resistance in HNSCCs and highlight future perspectives intended to overcome its related complications. Classical resistance mechanisms include drug import and export, DNA repair and oxidative stress control. Emerging research identified the prevalence of these mechanisms in populations of cancer stem cells (CSC), which are the cells mainly contributing to cisplatin resistance. The use of old and new CSC markers has enabled the identification of the characteristics within HNSCC CSCs predisposing them to treatment resistance, such as cell quiescence, increased self-renewal capacity, low reactive oxygen species levels or the acquisition of epithelial to mesenchymal transcriptional programs. In the present review, we will discuss how cell intrinsic and extrinsic cues alter the phenotype of CSCs and how they influence resistance to cisplatin treatment. In addition, we will assess how the stromal composition and the tumor microenvironment affect drug resistance and the acquisition of CSCs’ characteristics through a complex interplay between extracellular matrix content as well as immune and non-immune cell characteristics. Finally, we will describe how alterations in epigenetic modifiers or other signaling pathways can alter tumor behavior and cell plasticity to induce chemotherapy resistance. The data generated in recent years open up a wide range of promising strategies to optimize cisplatin therapy, with the potential to personalize HNSCC patient treatment strategies. Full article
Show Figures

Figure 1

Back to TopTop