New Sight in Cancer Genetics

A special issue of Biology (ISSN 2079-7737). This special issue belongs to the section "Cancer Biology".

Deadline for manuscript submissions: 31 May 2024 | Viewed by 4963

Special Issue Editors

1. Cytogenetics and Genomics Laboratory, Institute of Cellular and Molecular Biology, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
2. Institute for Clinical and Biomedical Research (iCBR), Center of Investigation on Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine (FMUC), University of Coimbra, 3004-531 Coimbra, Portugal
Interests: genomic; omics in cancer; head and neck cancer; methylation and genetic mechanisms; oncobiology; copy number alterations; prognosis biomarkers
1. Cytogenetics and Genomics Laboratory, Institute of Cellular and Molecular Biology, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
2. Institute for Clinical and Biomedical Research (iCBR), Center of Investigation on Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine (FMUC), University of Coimbra, 3004-531 Coimbra, Portugal
Interests: cytogenetics; human genetics, cancer genetics, oncobiology
Department of Medicine, Cancer Research Center (IBMCC-CSIC/USAL), CIBERONC and Institute for Biomedical Research of Salamanca (IBSAL), University of Salamanca (USAL), Salamanca, Spain
Interests: leukemia; lymphoma; early cancer diagnosis; immunophenotyping; genetics; diagnosis; prognosis; minimal residual disease; immune monitoring; CART; EuroFlow
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

In general, it has remained challenging to treat cancer despite the innovative and revolutionary research taking place worldwide. The remarkable advances over the last decade in multi-omics technologies, especially next-generation sequencing (NGS) and genome analysis, have led to significant improvements in our knowledge of the molecular mechanisms of cancer. Together with bioinformatic tools and Artificial Intelligence, it is possible to use computer programming to improve cancer diagnosis, drug development, and precision medicine.

This Special Issue aims to present robust molecular signatures and biomarkers of unmet clinical problems in cancer field, mostly focusing on predicting treatment response, making early diagnosis and assessing disease prognosis through the use of different omics approaches, such as genomic, epigenomic, transcriptomis, proteomic, metabolomic methods. We welcome original research articles and reviews to this Special Issue. Research areas may include (but are not limited to) the following topics: new developments in cancer research in order to identify biomarkers and molecular signatures with high possibility of clinical application and therapeutic development.

We look forward to receiving your contributions.

Prof. Dr. Ilda Patrícia Ribeiro
Prof. Dr. Isabel Marques Carreira
Prof. Dr. Alberto Orfao
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. Biology 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 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

  • genomic and epigenetic characterization
  • diagnosis and prognosis biomarkers
  • omics profiles
  • genetic and genomic signature
  • molecular heterogeneity
  • molecular targeted therapy
  • oncogenic signaling pathways
  • copy number alterations and mutational landscape
  • chromosome abnormalities

Published Papers (3 papers)

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

Research

Jump to: Review

23 pages, 7697 KiB  
Article
An Exploration of Small Molecules That Bind Human Single-Stranded DNA Binding Protein 1
by Zachariah P. Schuurs, Alexander P. Martyn, Carl P. Soltau, Sam Beard, Esha T. Shah, Mark N. Adams, Laura V. Croft, Kenneth J. O’Byrne, Derek J. Richard and Neha S. Gandhi
Biology 2023, 12(11), 1405; https://doi.org/10.3390/biology12111405 - 06 Nov 2023
Cited by 1 | Viewed by 1497
Abstract
Human single-stranded DNA binding protein 1 (hSSB1) is critical to preserving genome stability, interacting with single-stranded DNA (ssDNA) through an oligonucleotide/oligosaccharide binding-fold. The depletion of hSSB1 in cell-line models leads to aberrant DNA repair and increased sensitivity to irradiation. hSSB1 is over-expressed in [...] Read more.
Human single-stranded DNA binding protein 1 (hSSB1) is critical to preserving genome stability, interacting with single-stranded DNA (ssDNA) through an oligonucleotide/oligosaccharide binding-fold. The depletion of hSSB1 in cell-line models leads to aberrant DNA repair and increased sensitivity to irradiation. hSSB1 is over-expressed in several types of cancers, suggesting that hSSB1 could be a novel therapeutic target in malignant disease. hSSB1 binding studies have focused on DNA; however, despite the availability of 3D structures, small molecules targeting hSSB1 have not been explored. Quinoline derivatives targeting hSSB1 were designed through a virtual fragment-based screening process, synthesizing them using AlphaLISA and EMSA to determine their affinity for hSSB1. In parallel, we further screened a structurally diverse compound library against hSSB1 using the same biochemical assays. Three compounds with nanomolar affinity for hSSB1 were identified, exhibiting cytotoxicity in an osteosarcoma cell line. To our knowledge, this is the first study to identify small molecules that modulate hSSB1 activity. Molecular dynamics simulations indicated that three of the compounds that were tested bound to the ssDNA-binding site of hSSB1, providing a framework for the further elucidation of inhibition mechanisms. These data suggest that small molecules can disrupt the interaction between hSSB1 and ssDNA, and may also affect the ability of cells to repair DNA damage. This test study of small molecules holds the potential to provide insights into fundamental biochemical questions regarding the OB-fold. Full article
(This article belongs to the Special Issue New Sight in Cancer Genetics)
Show Figures

Graphical abstract

13 pages, 1753 KiB  
Article
3D Chromatin Architecture Re-Wiring at the CDH3/CDH1 Loci Contributes to E-Cadherin to P-Cadherin Expression Switch in Gastric Cancer
by Celina São José, Carla Pereira, Marta Ferreira, Ana André, Hugo Osório, Irene Gullo, Fátima Carneiro and Carla Oliveira
Biology 2023, 12(6), 803; https://doi.org/10.3390/biology12060803 - 31 May 2023
Viewed by 1344
Abstract
Cadherins are cell–cell adhesion molecules, fundamental for cell architecture and polarity. E-cadherin to P-cadherin switch can rescue adherens junctions in epithelial tumours. Herein, we disclose a mechanism for E-cadherin to P-cadherin switch in gastric cancers. CDH1 and CDH3 mRNA expression was obtained from [...] Read more.
Cadherins are cell–cell adhesion molecules, fundamental for cell architecture and polarity. E-cadherin to P-cadherin switch can rescue adherens junctions in epithelial tumours. Herein, we disclose a mechanism for E-cadherin to P-cadherin switch in gastric cancers. CDH1 and CDH3 mRNA expression was obtained from 42 gastric tumours’ RNA-seq data. CRISPR-Cas9 was used to knock out CDH1 and a putative regulatory element. CDH1-depleted and parental cells were submitted to proteomics and enrichment GO terms analysis; ATAC-seq/4C-seq with a CDH1 promoter viewpoint to assess chromatin accessibility and conformation; and RT-PCR/flow cytometry to assess CDH1/E-cadherin and CDH3/P-cadherin expression. In 42% of gastric tumours analysed, CDH1 to CDH3 switch was observed. CDH1 knockout triggered CDH1/E-cadherin complete loss and CDH3/P-cadherin expression increase at plasma membrane. This switch, likely rescuing adherens junctions, increased cell migration/proliferation, commonly observed in aggressive tumours. E- to P-cadherin switch accompanied increased CDH1 promoter interactions with CDH3–eQTL, absent in normal stomach and parental cells. CDH3–eQTL deletion promotes CDH3/CDH1 reduced expression. These data provide evidence that loss of CDH1/E-cadherin expression alters the CDH3 locus chromatin conformation, allowing a CDH1 promoter interaction with a CDH3-eQTL, and promoting CDH3/P-cadherin expression. These data highlight a novel mechanism triggering E- to P-cadherin switch in gastric cancer. Full article
(This article belongs to the Special Issue New Sight in Cancer Genetics)
Show Figures

Figure 1

Review

Jump to: Research

15 pages, 816 KiB  
Review
Current Applications and Challenges of Next-Generation Sequencing in Plasma Circulating Tumour DNA of Ovarian Cancer
by Ricardo Roque, Ilda Patrícia Ribeiro, Margarida Figueiredo-Dias, Charlie Gourley and Isabel Marques Carreira
Biology 2024, 13(2), 88; https://doi.org/10.3390/biology13020088 - 31 Jan 2024
Viewed by 1462
Abstract
Circulating tumour DNA (ctDNA) facilitates longitudinal study of the tumour genome, which, unlike tumour tissue biopsies, globally reflects intratumor and intermetastatis heterogeneity. Despite its costs, next-generation sequencing (NGS) has revolutionised the study of ctDNA, ensuring a more comprehensive and multimodal approach, increasing data [...] Read more.
Circulating tumour DNA (ctDNA) facilitates longitudinal study of the tumour genome, which, unlike tumour tissue biopsies, globally reflects intratumor and intermetastatis heterogeneity. Despite its costs, next-generation sequencing (NGS) has revolutionised the study of ctDNA, ensuring a more comprehensive and multimodal approach, increasing data collection, and introducing new variables that can be correlated with clinical outcomes. Current NGS strategies can comprise a tumour-informed set of genes or the entire genome and detect a tumour fraction as low as 10−5. Despite some conflicting studies, there is evidence that ctDNA levels can predict the worse outcomes of ovarian cancer (OC) in both early and advanced disease. Changes in those levels can also be informative regarding treatment efficacy and tumour recurrence, capable of outperforming CA-125, currently the only universally utilised plasma biomarker in high-grade serous OC (HGSOC). Qualitative evaluation of sequencing shows that increasing copy number alterations and gene variants during treatment may correlate with a worse prognosis in HGSOC. However, following tumour clonality and emerging variants during treatment poses a more unique opportunity to define treatment response, select patients based on their emerging resistance mechanisms, like BRCA secondary mutations, and discover potential targetable variants. Sequencing of tumour biopsies and ctDNA is not always concordant, likely as a result of clonal heterogeneity, which is better captured in the plasma samples than it is in a large number of biopsies. These incoherences may reflect tumour clonality and reveal the acquired alterations that cause treatment resistance. Cell-free DNA methylation profiles can be used to distinguish OC from healthy individuals, and NGS methylation panels have been shown to have excellent diagnostic capabilities. Also, methylation signatures showed promise in explaining treatment responses, including BRCA dysfunction. ctDNA is evolving as a promising new biomarker to track tumour evolution and clonality through the treatment of early and advanced ovarian cancer, with potential applicability in prognostic prediction and treatment selection. While its role in HGSOC paves the way to clinical applicability, its potential interest in other histological subtypes of OC remains unknown. Full article
(This article belongs to the Special Issue New Sight in Cancer Genetics)
Show Figures

Figure 1

Back to TopTop