1
|
Abo-Ghneim FDF, Al-Koofee DAF, Mohammed HJ. Association of XRCC1 (rs1799782) and XPD (rs13181) gene polymorphisms with renal failure risk in a sample of Iraqi population: a case-control study. Mol Biol Rep 2025; 52:294. [PMID: 40056231 DOI: 10.1007/s11033-025-10408-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 03/04/2025] [Indexed: 03/10/2025]
Abstract
BACKGROUND End-stage chronic kidney disease (CKD) can lead to life-threatening complications and is caused primarily by CKD and cardiovascular issues. CKD is characterized by the inability of the kidneys to filter waste and excess fluids from the blood. This study investigated the associations of the genetic variants XRCC1 rs1799782 (C194T) and ERCC2/XPD rs25487 (Q399R) with CKD susceptibility in Iraqi patients and related biochemical changes. METHODS The research was performed from 25/01/2023 to 30/06/2023, we analyzed the genetic associations of two SNPs of DNA repair genes (XRCC1 and ERCC2/XPD) in a case‒control study involving 219 CKD patients diagnosed by a nephrologist and 246 healthy controls. Data and blood samples were collected, and the genotype distribution frequency was determined via the PCR-based high-resolution melting (PCR-HRM) technique. RESULTS This study included 465 participants, with 219 CKD patients and 246 healthy controls. XRCC1 and ERCC2/XPD gene polymorphisms were significantly associated with CKD susceptibility in Iraqi patients (p = 0.025 and p = 0.0001, respectively). Multivariate linear regression confirmed the associations of rs1799782 G/A and rs13181T/G with CKD, adjusting for sex, age, and BMI. Moderate and statistically significant linkage disequilibrium (0.43) between the two SNPs was observed, indicating nonrandom associations. CONCLUSION XRCC1 (rs1799782) and ERCC2/XPD (rs13181) polymorphisms are associated with an increased risk of CKD. The AG haplotype model is particularly related to increased CKD susceptibility in Iraqi patients, suggesting the importance of these DNA repair gene polymorphisms in CKD risk assessment.
Collapse
Affiliation(s)
- Fahad D F Abo-Ghneim
- Department of Medical Lab., Faculty of Healthy and Medical Techniques, Al-Furat Al-Awsat Technical University, Najaf, Iraq
| | - Dhafer A F Al-Koofee
- Department of Clinical Lab. Sc., Faculty of Pharmacy, University of Kufa, Najaf, 54002, Iraq.
| | | |
Collapse
|
2
|
Gong P, Guo Z, Wang S, Gao S, Cao Q. Histone Phosphorylation in DNA Damage Response. Int J Mol Sci 2025; 26:2405. [PMID: 40141048 PMCID: PMC11941871 DOI: 10.3390/ijms26062405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/01/2025] [Accepted: 03/05/2025] [Indexed: 03/28/2025] Open
Abstract
The DNA damage response (DDR) is crucial for maintaining genomic stability and preventing the accumulation of mutations that can lead to various diseases, including cancer. The DDR is a complex cellular regulatory network that involves DNA damage sensing, signal transduction, repair, and cell cycle arrest. Modifications in histone phosphorylation play important roles in these processes, facilitating DNA repair factor recruitment, damage signal transduction, chromatin remodeling, and cell cycle regulation. The precise regulation of histone phosphorylation is critical for the effective repair of DNA damage, genomic integrity maintenance, and the prevention of diseases such as cancer, where DNA repair mechanisms are often compromised. Thus, understanding histone phosphorylation in the DDR provides insights into DDR mechanisms and offers potential therapeutic targets for diseases associated with genomic instability, including cancers.
Collapse
Affiliation(s)
- Ping Gong
- Hunan Institute of Microbiology, Changsha 410009, China; (Z.G.); (S.W.); (S.G.)
| | - Zhaohui Guo
- Hunan Institute of Microbiology, Changsha 410009, China; (Z.G.); (S.W.); (S.G.)
| | - Shengping Wang
- Hunan Institute of Microbiology, Changsha 410009, China; (Z.G.); (S.W.); (S.G.)
| | - Shufeng Gao
- Hunan Institute of Microbiology, Changsha 410009, China; (Z.G.); (S.W.); (S.G.)
| | - Qinhong Cao
- College of Biological Sciences, China Agricultural University, No.2 Yuan-Ming-Yuan West Road, Beijing 100193, China
| |
Collapse
|
3
|
Smahel M, Johari SD, Smahelova J, Pfeiferova L, Nunvar J. Spatial immune heterogeneity in a mouse tumor model after immunotherapy. Cancer Sci 2025; 116:622-632. [PMID: 39624899 PMCID: PMC11875765 DOI: 10.1111/cas.16421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 11/15/2024] [Accepted: 11/19/2024] [Indexed: 03/05/2025] Open
Abstract
Cancer immunotherapy is increasingly used in clinical practice, but its success rate is reduced by tumor escape from the immune system. This may be due to the genetic instability of tumor cells, which allows them to adapt to the immune response and leads to intratumoral immune heterogeneity. The study investigated spatial immune heterogeneity in the tumor microenvironment and its possible drivers in a mouse model of tumors induced by human papillomaviruses (HPV) following immunotherapy. Gene expression was determined by RNA sequencing and mutations by whole exome sequencing. A comparison of different tumor areas revealed heterogeneity in immune cell infiltration, gene expression, and mutation composition. While the mean numbers of mutations with every impact on gene expression or protein function were comparable in treated and control tumors, mutations with high or moderate impact were increased after immunotherapy. The genes mutated in treated tumors were significantly enriched in genes associated with ECM metabolism, degradation, and interactions, HPV infection and carcinogenesis, and immune processes such as antigen processing and presentation, Toll-like receptor signaling, and cytokine production. Gene expression analysis of DNA damage and repair factors revealed that immunotherapy upregulated Apobec1 and Apobec3 genes and downregulated genes related to homologous recombination and translesion synthesis. In conclusion, this study describes the intratumoral immune heterogeneity, that could lead to tumor immune escape, and suggests the potential mechanisms involved.
Collapse
Affiliation(s)
- Michal Smahel
- Department of Genetics and Microbiology, Faculty of ScienceCharles University, BIOCEVVestecCzech Republic
| | - Shweta Dilip Johari
- Department of Genetics and Microbiology, Faculty of ScienceCharles University, BIOCEVVestecCzech Republic
| | - Jana Smahelova
- Department of Genetics and Microbiology, Faculty of ScienceCharles University, BIOCEVVestecCzech Republic
| | - Lucie Pfeiferova
- Laboratory of Genomics and BioinformaticsInstitute of Molecular Genetics, Czech Academy of SciencesPragueCzech Republic
| | - Jaroslav Nunvar
- Department of Genetics and Microbiology, Faculty of ScienceCharles University, BIOCEVVestecCzech Republic
| |
Collapse
|
4
|
Ampolini EA, Jimenez-Sainz J, Long DT. The Development of ATM Inhibitors in Cancer Therapy. Target Oncol 2025; 20:281-297. [PMID: 40024979 PMCID: PMC11933189 DOI: 10.1007/s11523-025-01136-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/18/2025] [Indexed: 03/04/2025]
Abstract
The ataxia-telangiectasia mutated (ATM) protein kinase plays a critical role in activating the cellular response to DNA double-strand breaks and promoting homology-directed repair. ATM is frequently mutated in cancer, contributing to an accumulation of DNA damage that drives genomic instability. To exploit cancer cells' inherent vulnerability to DNA damage, various small molecule inhibitors have been developed that target ATM. ATM inhibitors have shown great versatility in preclinical studies and increasing use in the clinic. Here, we review the development of ATM inhibitors and their role in cancer therapy. We describe their limitations and the advances that have led to increases in both the number and diversity of active clinical trials targeting ATM. We also discuss ATM's role in personalized medicine and the current challenges to more widespread use of ATM inhibitors in the clinic.
Collapse
Affiliation(s)
- Elizabeth A Ampolini
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Judit Jimenez-Sainz
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - David T Long
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, 29425, USA.
| |
Collapse
|
5
|
Dikoglu E, Pareja F. Molecular Basis of Breast Tumor Heterogeneity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1464:237-257. [PMID: 39821029 DOI: 10.1007/978-3-031-70875-6_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Breast cancer (BC) is a profoundly heterogenous disease, with diverse molecular, histological, and clinical variations. The intricate molecular landscape of BC is evident even at early stages, illustrated by the complexity of the evolution from precursor lesions to invasive carcinoma. The key for therapeutic decision-making is the dynamic assessment of BC receptor status and clinical subtyping. Hereditary BC adds an additional layer of complexity to the disease, given that different cancer susceptibility genes contribute to distinct phenotypes and genomic features. Furthermore, the various BC subtypes display distinct metabolic demands and immune microenvironments. Finally, genotypic-phenotypic correlations in special histologic subtypes of BC inform diagnostic and therapeutic approaches, highlighting the significance of thoroughly comprehending BC heterogeneity.
Collapse
Affiliation(s)
- Esra Dikoglu
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Fresia Pareja
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
6
|
Guedes AR, Soares JP, Cunha R, Silva AM, Gaivão I. DNA damage of peripheral blood lymphocytes as a dual biomarker: Diagnostic and treatment response in woman breast cancer patients. Cancer Biomark 2025; 42:18758592241308748. [PMID: 40109217 DOI: 10.1177/18758592241308748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
BackgroundBreast cancer is the leading malignancy among women and the lack of ideal early biomarkers hampers diagnosis and treatment monitoring. Genomic instability, central to breast cancer development, makes DNA damage a potential biomarker for these purposes.ObjectiveThis study aims to evaluate the predictive value of DNA damage for diagnosis, and treatment monitoring in breast cancer, with CA 15-3, a conventional cancer biomarker, included for comparison to assess the added value of DNA damage measurement.MethodsDNA damage was measured in peripheral blood lymphocytes of 58 breast cancer patients, and 31 healthy controls, employing comet assay, both before and after treatment. Serum CA 15-3 levels were assessed at the same time points for comparison.ResultsDNA damage levels were significantly higher in cancer patients compared to healthy controls, with the most elevated levels observed in patients with advanced-stage disease, irrespective of age, sex, lifestyle, or genetic status. Post-treatment assessments showed a significant rise in DNA damage. In comparison, CA 15-3 showed less consistent relevance for diagnostic and monitoring.ConclusionsThis study underscores the greater potential of DNA damage as a consistent and reliable biomarker for breast cancer, with CA 15-3 providing complementary but less consistent data for clinical decision-making.
Collapse
Affiliation(s)
- Ana Rita Guedes
- Animal and Veterinary Research Center (CECAV) and Department of Genetics and Biotechnology, University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), UTAD, Vila Real, Portugal
- Department of Biology and Environment (DeBA) and Centre for Research and Technology of Agro-Environmental and Biological Sciences (CITAB), UTAD, Vila Real, Portugal
| | - Jorge Pinto Soares
- Research Centre of Sports, Health, and Human Development (CIDES), UTAD, Vila Real, Portugal
| | - Renato Cunha
- Local Health Unit of Trás-os-Montes and Alto Douro, Vila Real, Portugal
| | - Amélia Maria Silva
- Department of Biology and Environment (DeBA) and Centre for Research and Technology of Agro-Environmental and Biological Sciences (CITAB), UTAD, Vila Real, Portugal
| | - Isabel Gaivão
- Animal and Veterinary Research Center (CECAV) and Department of Genetics and Biotechnology, University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), UTAD, Vila Real, Portugal
| |
Collapse
|
7
|
Hikisz P, Wawrzyniak P, Adamus-Grabicka AA, Jacenik D, Budzisz E. Evaluation of In Vitro Biological Activity of Flavanone/Chromanone Derivatives: Molecular Analysis of Anticancer Mechanisms in Colorectal Cancer. Int J Mol Sci 2024; 25:12985. [PMID: 39684693 DOI: 10.3390/ijms252312985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 11/28/2024] [Accepted: 12/01/2024] [Indexed: 12/18/2024] Open
Abstract
The primary objective of this study was to evaluate the anticancer activity of six flavanone/chromanone derivatives: 3-benzylideneflavanones/3-benzylidenechroman-4-ones and their 3-spiro-1-pirazolines analogs. We employed five colon cancer cell lines with varying degrees of metastasis and genetic profiles as our research model. Our investigation focused primarily on assessing the pro-oxidant properties of the tested derivatives and their impact on overall antiproliferative activity. To comprehensively evaluate the cytotoxic properties of these compounds, we analyzed their genotoxic, pro-apoptotic, and autophagy-inducing effects. Our findings indicate that three of the six analyzed derivatives exhibited promising antiproliferative activity against cancer cells, with IC50 values ranging from 10 to 30 μM. Strong pro-oxidant properties were identified as a key mechanism underlying their cytotoxic activity. The generation of oxidative stress, which varied depending on the specific flavanone/chromanone derivative, resulted from increased intracellular reactive oxygen species (ROS) levels and decreased glutathione (GSH) concentrations. Furthermore, oxidative stress likely contributed to the induction of apoptosis/autophagy in cancer cells and the emergence of significant DNA damage.
Collapse
Affiliation(s)
- Pawel Hikisz
- Department of Oncobiology and Epigenetics, Institute of Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland
| | - Piotr Wawrzyniak
- Department of Molecular Biology of Cancer, Medical University of Lodz, 6/8 Mazowiecka Street, 92-215 Lodz, Poland
| | - Angelika A Adamus-Grabicka
- Department of Bioinorganic Chemistry, Faculty of Pharmacy, Medical University of Lodz, Muszynskiego 1, 90-151 Lodz, Poland
| | - Damian Jacenik
- Department of Cytobiochemistry, Faculty of Biology and Environmental Protection, University of Lodz, ul. Pomorska 141/143, 90-236 Lodz, Poland
| | - Elzbieta Budzisz
- Department of the Chemistry of Cosmetic Raw Materials, Medical University of Lodz, 90-151 Lodz, Poland
| |
Collapse
|
8
|
Spirrison AN, Lannigan DA. RSK1 and RSK2 as therapeutic targets: an up-to-date snapshot of emerging data. Expert Opin Ther Targets 2024; 28:1047-1059. [PMID: 39632509 PMCID: PMC11801519 DOI: 10.1080/14728222.2024.2433123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 11/19/2024] [Indexed: 12/07/2024]
Abstract
INTRODUCTION The four members of the p90 ribosomal S6 kinase (RSK) family are serine/threonine protein kinases, which are phosphorylated and activated by ERK1/2. RSK1/2/3 are further phosphorylated by PDK1. Receiving inputs from two major signaling pathways places RSK as a key signaling node in numerous pathologies. A plethora of RSK1/2 substrates have been identified, and in the majority of cases the causative roles these RSK substrates play in the pathology are unknown. AREAS COVERED The majority of studies have focused on RSK1/2 and their functions in a diverse group of cancers. However, RSK1/2 are known to have important functions in cardiovascular disease and neurobiological disorders. Based on the literature, we identified substrates that are common in these pathologies with the goal of identifying fundamental physiological responses to RSK1/2. EXPERT OPINION The core group of targets in pathologies driven by RSK1/2 are associated with the immune response. However, there is a paucity of the literature addressing RSK function in inflammation, which is critical to know as the pan RSK inhibitor, PMD-026, is entering phase II clinical trials for metastatic breast cancer. A RSK inhibitor has the potential to be used in numerous diverse diseases and disorders.
Collapse
Affiliation(s)
| | - Deborah A. Lannigan
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
- Department of Pathology, Microbiology & Immunology, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
9
|
Brooke G, Wendel S, Banerjee A, Wallace N. Opportunities to advance cervical cancer prevention and care. Tumour Virus Res 2024; 18:200292. [PMID: 39490532 PMCID: PMC11566706 DOI: 10.1016/j.tvr.2024.200292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/05/2024] Open
Abstract
Cervical cancer (CaCx) is a major public health issue, with over 600,000 women diagnosed annually. CaCx kills someone every 90 s, mostly in low- and middle-income countries. There are effective yet imperfect mechanisms to prevent CaCx. Since human papillomavirus (HPV) infections cause most CaCx, they can be prevented by vaccination. Screening methodologies can identify premalignant lesions and allow interventions before a CaCx develops. However, these tools are less feasible in resource-poor environments. Additionally, current screening modalities cannot triage lesions based on their relative risk of progression, which results in overtreatment. CaCx care relies heavily on genotoxic agents that cause severe side effects. This review discusses ways that recent technological advancements could be leveraged to improve CaCx care and prevention.
Collapse
Affiliation(s)
- Grant Brooke
- Division of Biology, Kansas State University, Manhattan, KS 66506, USA
| | - Sebastian Wendel
- Department of Kinesiology, Kansas State University, Manhattan, KS 66506, USA
| | - Abhineet Banerjee
- Division of Biology, Kansas State University, Manhattan, KS 66506, USA
| | - Nicholas Wallace
- Department of Kinesiology, Kansas State University, Manhattan, KS 66506, USA.
| |
Collapse
|
10
|
D'Amato A, Iacopetta D, Ceramella J, Troiano R, Mariconda A, Catalano A, Marra M, Saturnino C, Rosano C, Sinicropi MS, Longo P. Design, synthesis and biological evaluation of multitarget hybrid molecules containing NHC-Au(I) complexes and carbazole moieties. Eur J Med Chem 2024; 277:116757. [PMID: 39142149 DOI: 10.1016/j.ejmech.2024.116757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 07/15/2024] [Accepted: 08/01/2024] [Indexed: 08/16/2024]
Abstract
N-heterocyclic carbenes (NHCs) represent suitable ligands for rapid and efficient drug design, because they offer the advantage of being easily chemically modified and can bind several substituents, including transition metals as, for instance, gold derivatives. Gold-NHC complexes possess various biological activities and were demonstrated good candidates as anticancer drugs. Besides, carbazole derivatives are characterized by various pharmacological properties, such as anticancer, antibacterial, anti-inflammatory, and anti-psychotropic. Amongst the latter, N-thioalkyl carbazoles were proved to inhibit cancer cells damaging the nuclear DNA, through the inhibition of human topoisomerases. Herein, we report the design, synthesis and biological evaluation of nine new hybrid molecules in which NHC-Au(I) complexes and N-alkylthiolated carbazoles are linked together, in order to obtain novel biological multitarget agents. We demonstrated that the lead hybrid complexes possess anticancer, anti-inflammatory and antioxidant properties, with a high potential as useful tools for treating distinct aspects of several diseases, amongst them cancer.
Collapse
Affiliation(s)
- A D'Amato
- Department of Chemistry and Biology "A. Zambelli", University of Salerno, Via Giovanni Paolo II 132, I-84084, Fisciano, SA, Italy
| | - D Iacopetta
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via Pietro Bucci, I-87036, Arcavacata di Rende, CS, Italy
| | - J Ceramella
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via Pietro Bucci, I-87036, Arcavacata di Rende, CS, Italy
| | - R Troiano
- Department of Chemistry and Biology "A. Zambelli", University of Salerno, Via Giovanni Paolo II 132, I-84084, Fisciano, SA, Italy
| | - A Mariconda
- Department of Science, University of Basilicata, Via dell'Ateneo Lucano 10, I-85100, Potenza, Italy.
| | - A Catalano
- Department of Pharmacy-Drug Sciences, University of Bari "Aldo Moro", Via Orabona 4, 70126, Bari, Italy.
| | - M Marra
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via Pietro Bucci, I-87036, Arcavacata di Rende, CS, Italy
| | - C Saturnino
- Department of Science, University of Basilicata, Via dell'Ateneo Lucano 10, I-85100, Potenza, Italy
| | - C Rosano
- U.O. Proteomica e Spettrometria di Massa, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132, Genova, Italy
| | - M S Sinicropi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via Pietro Bucci, I-87036, Arcavacata di Rende, CS, Italy
| | - P Longo
- Department of Chemistry and Biology "A. Zambelli", University of Salerno, Via Giovanni Paolo II 132, I-84084, Fisciano, SA, Italy
| |
Collapse
|
11
|
Pigg HC, Alley KR, Griffin CR, Moon CH, Kraske SJ, DeRose VJ. The unique Pt(II)-induced nucleolar stress response and its deviation from DNA damage response pathways. J Biol Chem 2024; 300:107858. [PMID: 39374783 PMCID: PMC11612370 DOI: 10.1016/j.jbc.2024.107858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 09/09/2024] [Accepted: 09/11/2024] [Indexed: 10/09/2024] Open
Abstract
The mechanisms of action for the platinum compounds cisplatin and oxaliplatin have yet to be fully elucidated, despite the worldwide use of these drugs. Recent studies suggest that the two compounds may be working through different mechanisms, with cisplatin inducing cell death via the DNA damage response (DDR) and oxaliplatin utilizing a nucleolar stress-based cell death pathway. While cisplatin-induced DDR has been subject to much research, the mechanisms for oxaliplatin's influence on the nucleolus are not well understood. Prior work has outlined structural parameters for Pt(II) derivatives capable of nucleolar stress induction. In this work, we gain insight into the nucleolar stress response induced by these Pt(II) derivatives by investigating potential correlations between this unique pathway and DDR. Key findings from this study indicate that Pt(II)-induced nucleolar stress occurs when DDR is inhibited and works independently of the ATM/ATR-dependent DDR pathway. We also determine that Pt(II)-induced stress may be linked to the G1 cell cycle phase, as cisplatin can induce nucleolar stress when cell cycle inhibition occurs at the G1/S checkpoint. Finally, we compare Pt(II)-induced nucleolar stress with other small-molecule nucleolar stress-inducing compounds Actinomycin D, BMH-21, and CX-5461 and find that Pt(II) compounds cause irreversible nucleolar stress, whereas the reversibility of nucleolar stress induced by small-molecules varies. Taken together, these findings contribute to a better understanding of Pt(II)-induced nucleolar stress, its deviation from ATM/ATR-dependent DDR, and the possible influence of cell cycle on the ability of Pt(II) compounds to cause nucleolar stress.
Collapse
Affiliation(s)
- Hannah C Pigg
- Department of Chemistry and Biochemistry, University of Oregon, Eugene, Oregon, USA
| | - Katelyn R Alley
- Department of Chemistry and Biochemistry, University of Oregon, Eugene, Oregon, USA
| | | | - Caleb H Moon
- Department of Chemistry and Biochemistry, University of Oregon, Eugene, Oregon, USA
| | - Sarah J Kraske
- Department of Chemistry and Biochemistry, University of Oregon, Eugene, Oregon, USA
| | - Victoria J DeRose
- Department of Chemistry and Biochemistry, University of Oregon, Eugene, Oregon, USA.
| |
Collapse
|
12
|
Moosavi F, Hassani B, Nazari S, Saso L, Firuzi O. Targeting DNA damage response in pancreatic ductal adenocarcinoma: A review of preclinical and clinical evidence. Biochim Biophys Acta Rev Cancer 2024; 1879:189185. [PMID: 39326802 DOI: 10.1016/j.bbcan.2024.189185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 09/18/2024] [Accepted: 09/18/2024] [Indexed: 09/28/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is associated with one of the most unfavorable prognoses across all malignancies. In this review, we investigate the role of inhibitors targeting crucial regulators of DNA damage response (DDR) pathways, either as single treatments or in combination with chemotherapeutic agents and targeted therapies in PDAC. The most prominent clinical benefit of PARP inhibitors' monotherapy is related to the principle of synthetic lethality in individuals harboring BRCA1/2 and other DDR gene mutations as predictive biomarkers. Moreover, induction of BRCAness with inhibitors of RTKs, including VEGFR and c-MET and their downstream signaling pathways, RAS/RAF/MEK/ERK and PI3K/AKT/mTOR in order to expand the application of PARP inhibitors in patients without DDR mutations, has also been addressed. Other DDR-targeting agents beyond PARP inhibitors, including inhibitors of ATM, ATR, CHEK1/2, and WEE1 have also demonstrated their potential in preclinical models of PDAC and may hold promise in future studies.
Collapse
Affiliation(s)
- Fatemeh Moosavi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Bahareh Hassani
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Somayeh Nazari
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Luciano Saso
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of Rome, P. le Aldo Moro 5, 00185 Rome, Italy
| | - Omidreza Firuzi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
13
|
Feng Y, Yang J, He Z, Liu X, Ma C. CRISPR-Cas-based biosensors for the detection of cancer biomarkers. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024; 16:6634-6653. [PMID: 39258950 DOI: 10.1039/d4ay01446d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Along with discovering cancer biomarkers, non-invasive detection methods have played a critical role in early cancer diagnosis and prognostic improvement. Some traditional detection methods have been used for detecting cancer biomarkers, but they are time-consuming and involve materials and human costs. With great flexibility, sensitivity and specificity, the clustered regularly interspaced short palindromic repeats (CRISPR)-associated system provides a wide range of application prospects in this field. Herein, we introduce the background of the CRISPR-Cas (CRISPR-associated) system and comprehensively summarize the diagnosis strategies of cancer mediated by the CRISPR-Cas system, including four kinds of biochemical-based markers: nucleic acid, enzyme, tumor-specific protein and exosome. Furthermore, we discuss the challenges in implementing the CRISPR-Cas system in clinical applications.
Collapse
Affiliation(s)
- Yuxin Feng
- School of Life Sciences, Central South University, Changsha 410013, China.
- Clinical Medicine Eight-year Program, Xiangya School of Medicine, Central South University, Changsha 410078, China
| | - Jinmeng Yang
- School of Life Sciences, Central South University, Changsha 410013, China.
- Clinical Medicine Eight-year Program, Xiangya School of Medicine, Central South University, Changsha 410078, China
| | - Ziping He
- School of Life Sciences, Central South University, Changsha 410013, China.
- Clinical Medicine Eight-year Program, Xiangya School of Medicine, Central South University, Changsha 410078, China
| | - Xinfa Liu
- School of Life Sciences, Central South University, Changsha 410013, China.
| | - Changbei Ma
- School of Life Sciences, Central South University, Changsha 410013, China.
| |
Collapse
|
14
|
Du Q, Yuen HY, Pan J, Sun C, Wu D, Liu J, Wu G, Zhao X, Wang S. Metronidazole-modified Au@BSA nanocomposites for dual sensitization of radiotherapy in solid tumors. J Mater Chem B 2024; 12:9686-9694. [PMID: 39193619 DOI: 10.1039/d4tb00910j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
The hypoxic microenvironment of solid tumors can lead to reduced therapeutic DNA damage to the tumor cells, thus diminishing tumor sensitivity to radiotherapy. Although hypoxic radiosensitizers can improve radiotherapy efficacy by enhancing the role of oxygen, their effects are limited by the uneven distribution of oxygen within solid tumor tissues. In this study, a novel radiosensitizer via leveraging gold complexes and metronidazole (MN) was synthesized to improve radiotherapeutic efficacy. The gold atoms incorporated in the radiosensitizer enabled efficient deposition of high-energy radiation; the hydrophobic metronidazole was reduced to hydrophilic aminoimidazole under hypoxia conditions and further promoted radiotherapy sensitization. The results of CCK-8 assays, Live/Dead assays, γ-H2AX immunofluorescence indicated that metronidazole-modified Au@BSA nanocomposites (NCs) exhibited excellent antitumor effects. The in vivo antitumor tests further showed an inhibition rate of 100%. These results demonstrated that the NCs successfully enhanced radiotherapy efficacy by the dual sensitization strategy. Overall, we believe this multimodal radiosensitizing nanocomplex can significantly inhibit tumor growth and metastasis, with their hypoxia-oriented characteristics ensuring a higher efficacy and safety.
Collapse
Affiliation(s)
- Qijun Du
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, China.
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, China
| | - Ho-Yin Yuen
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong 999077, China.
| | - Jingke Pan
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, China.
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, China
| | - Chenwei Sun
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, China.
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, China
| | - Di Wu
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, China.
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, China
| | - Jie Liu
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, China.
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, China
| | - Guohua Wu
- Luoyang Key Laboratory of Clinical Multiomics and Translational Medicine, Henan Key Laboratory of Rare Diseases, Endocrinology and Metabolism Center, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang 471003, China.
| | - Xin Zhao
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong 999077, China.
| | - Shuqi Wang
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, China.
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, China
- Tianfu Jincheng Laboratory, City of Future Medicine, Chengdu 641400, China
- Clinical Research Center for Respiratory Disease, West China Hospital, Sichuan University, Chengdu 610065, China
| |
Collapse
|
15
|
Li J, Jia Z, Dong L, Cao H, Huang Y, Xu H, Xie Z, Jiang Y, Wang X, Liu J. DNA damage response in breast cancer and its significant role in guiding novel precise therapies. Biomark Res 2024; 12:111. [PMID: 39334297 PMCID: PMC11437670 DOI: 10.1186/s40364-024-00653-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
DNA damage response (DDR) deficiency has been one of the emerging targets in treating breast cancer in recent years. On the one hand, DDR coordinates cell cycle and signal transduction, whose dysfunction may lead to cell apoptosis, genomic instability, and tumor development. Conversely, DDR deficiency is an intrinsic feature of tumors that underlies their response to treatments that inflict DNA damage. In this review, we systematically explore various mechanisms of DDR, the rationale and research advances in DDR-targeted drugs in breast cancer, and discuss the challenges in its clinical applications. Notably, poly (ADP-ribose) polymerase (PARP) inhibitors have demonstrated favorable efficacy and safety in breast cancer with high homogenous recombination deficiency (HRD) status in a series of clinical trials. Moreover, several studies on novel DDR-related molecules are actively exploring to target tumors that become resistant to PARP inhibition. Before further clinical application of new regimens or drugs, novel and standardized biomarkers are needed to develop for accurately characterizing the benefit population and predicting efficacy. Despite the promising efficacy of DDR-related treatments, challenges of off-target toxicity and drug resistance need to be addressed. Strategies to overcome drug resistance await further exploration on DDR mechanisms, and combined targeted drugs or immunotherapy will hopefully provide more precise or combined strategies and expand potential responsive populations.
Collapse
Affiliation(s)
- Jiayi Li
- Department of Breast Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- School of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
| | - Ziqi Jia
- Department of Breast Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Lin Dong
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Heng Cao
- Department of Breast Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yansong Huang
- Department of Breast Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- School of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
| | - Hengyi Xu
- School of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Zhixuan Xie
- School of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
| | - Yiwen Jiang
- School of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
| | - Xiang Wang
- Department of Breast Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Jiaqi Liu
- Department of Breast Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
16
|
Aly SH, Elbadry AMM, Doghish AS, El-Nashar HAS. Unveiling the pharmacological potential of plant triterpenoids in breast cancer management: an updated review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:5571-5596. [PMID: 38563878 PMCID: PMC11329582 DOI: 10.1007/s00210-024-03054-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 03/18/2024] [Indexed: 04/04/2024]
Abstract
Breast cancer is the most prevalent type of cancer, the fifth leading cause of cancer-related deaths, and the second leading cause of cancer deaths among women globally. Recent research has provided increasing support for the significance of phytochemicals, both dietary and non-dietary, particularly triterpenoids, in the mitigation and management of breast cancer. Recent studies showed that triterpenoids are promising agents in the treatment and inhibition of breast cancer achieved through the implementation of several molecular modes of action on breast cancer cells. This review discusses recent innovations in plant triterpenoids and their underlying mechanisms of action in combating breast cancer within the timeframe spanning from 2017 to 2023. The present work is an overview of different plant triterpenoids with significant inhibition on proliferation, migration, apoptosis resistance, tumor angiogenesis, or metastasis in various breast cancer cells. The anticancer impact of triterpenoids may be attributed to their antiproliferative activity interfering with angiogenesis and differentiation, regulation of apoptosis, DNA polymerase inhibition, change in signal transductions, and impeding metastasis. The present review focuses on several targets, mechanisms, and pathways associated with pentacyclic triterpenoids, which are responsible for their anticancer effects. We could conclude that natural triterpenoids are considered promising agents to conquer breast cancer.
Collapse
Affiliation(s)
- Shaza H Aly
- Department of Pharmacognosy, Faculty of Pharmacy, Badr University in Cairo, Cairo, 11829, Egypt.
| | - Abdullah M M Elbadry
- Badr University in Cairo Research Center, Badr University in Cairo, Badr City, 11829, Cairo, Egypt
| | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, , 11829, Cairo, Egypt.
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Al-Azhar University, Nasr City, 11231, Cairo, Egypt.
| | - Heba A S El-Nashar
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt.
| |
Collapse
|
17
|
Liu Y, Yalavarthi S, Yang F, Abdul-Rashid Y, Tang S, Long Z, Qin Y, Wu K, Wang Z. Insights into treatment-specific prognostic somatic mutations in NSCLC from the AACR NSCLC GENIE BPC cohort analysis. BMC Pulm Med 2024; 24:309. [PMID: 38956553 PMCID: PMC11218090 DOI: 10.1186/s12890-024-03124-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 06/23/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND Treatment of non-small lung cancer (NSCLC) has evolved in recent years, benefiting from advances in immunotherapy and targeted therapy. However, limited biomarkers exist to assist clinicians and patients in selecting the most effective, personalized treatment strategies. Targeted next-generation sequencing-based genomic profiling has become routine in cancer treatment and generated crucial clinicogenomic data over the last decade. This has made the development of mutational biomarkers for drug response possible. METHODS To investigate the association between a patient's responses to a specific somatic mutation treatment, we analyzed the NSCLC GENIE BPC cohort, which includes 2,004 tumor samples from 1,846 patients. RESULTS We identified somatic mutation signatures associated with response to immunotherapy and chemotherapy, including carboplatin-, cisplatin-, pemetrexed- or docetaxel-based chemotherapy. The prediction power of the chemotherapy-associated signature was significantly affected by epidermal growth factor receptor (EGFR) mutation status. Therefore, we developed an EGFR wild-type-specific mutation signature for chemotherapy selection. CONCLUSION Our treatment-specific gene signatures will assist clinicians and patients in selecting from multiple treatment options.
Collapse
Affiliation(s)
- Yi Liu
- Department of Neurosurgery, the Third XiangYa Hospital of Central South University, Changsha, 410013, PR China
| | - Sindhu Yalavarthi
- Department of Nanoscience, The Joint School of Nanoscience and Nanoengineering, University of North Carolina Greensboro, Greensboro, NC, 27401, USA
| | - Fan Yang
- Department of Neurosurgery, the Third XiangYa Hospital of Central South University, Changsha, 410013, PR China
| | - Yusif Abdul-Rashid
- Department of Nanoscience, The Joint School of Nanoscience and Nanoengineering, University of North Carolina Greensboro, Greensboro, NC, 27401, USA
| | - Shenkun Tang
- Department of Neurosurgery, the Third XiangYa Hospital of Central South University, Changsha, 410013, PR China
| | - Zihe Long
- Department of Neurosurgery, the Third XiangYa Hospital of Central South University, Changsha, 410013, PR China
| | - Yongkai Qin
- Department of Neurosurgery, the Third XiangYa Hospital of Central South University, Changsha, 410013, PR China
| | - Kerui Wu
- Department of Nanoscience, The Joint School of Nanoscience and Nanoengineering, University of North Carolina Greensboro, Greensboro, NC, 27401, USA
| | - Zhifei Wang
- Department of Neurosurgery, the Third XiangYa Hospital of Central South University, Changsha, 410013, PR China.
| |
Collapse
|
18
|
Li L, Gao Y, Yu B, Zhang J, Ma G, Jin X. Role of LncRNA H19 in tumor progression and treatment. Mol Cell Probes 2024; 75:101961. [PMID: 38579914 DOI: 10.1016/j.mcp.2024.101961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 03/29/2024] [Accepted: 04/03/2024] [Indexed: 04/07/2024]
Abstract
As one of the earliest discovered lncRNA molecules, lncRNA H19 is usually expressed in large quantities during embryonic development and is involved in cell differentiation and tissue formation. In recent years, the role of lncRNA H19 in tumors has been gradually recognized. Increasing evidence suggests that its aberrant expression is closely related to cancer development. LncRNA H19 as an oncogene not only promotes the growth, proliferation, invasion and metastasis of many tumors, but also develops resistance to treatment, affecting patients' prognosis and survival. Therefore, in this review, we summarise the extensive research on the involvement of lncRNA H19 in tumor progression and discuss how lncRNA H19, as a key target gene, affects tumor sensitivity to radiotherapy, chemotherapy and immunotherapy by participating in multiple cellular processes and regulating multiple signaling pathways, which provides a promising prospect for further research into the treatment of cancer.
Collapse
Affiliation(s)
- Linjing Li
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou, 730000, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuting Gao
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China; College of Life Sciences, Northwest Normal University, Gansu Province, Lanzhou, 730070, China
| | - Boyi Yu
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou, 730000, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jiahao Zhang
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China; School of Public Health, Lanzhou University, Gansu Province, Lanzhou, 730000, China
| | - Guorong Ma
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, 730000, China
| | - Xiaodong Jin
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou, 730000, China; University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
19
|
Prabhu KS, Kuttikrishnan S, Ahmad N, Habeeba U, Mariyam Z, Suleman M, Bhat AA, Uddin S. H2AX: A key player in DNA damage response and a promising target for cancer therapy. Biomed Pharmacother 2024; 175:116663. [PMID: 38688170 DOI: 10.1016/j.biopha.2024.116663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 04/22/2024] [Accepted: 04/24/2024] [Indexed: 05/02/2024] Open
Abstract
Cancer is caused by a complex interaction of factors that interrupt the normal growth and division of cells. At the center of this process is the intricate relationship between DNA damage and the cellular mechanisms responsible for maintaining genomic stability. When DNA damage is not repaired, it can cause genetic mutations that contribute to the initiation and progression of cancer. On the other hand, the DNA damage response system, which involves the phosphorylation of the histone variant H2AX (γH2AX), is crucial in preserving genomic integrity by signaling and facilitating the repair of DNA double-strand breaks. This review provides an explanation of the molecular dynamics of H2AX in the context of DNA damage response. It emphasizes the crucial role of H2AX in recruiting and localizing repair machinery at sites of chromatin damage. The review explains how H2AX phosphorylation, facilitated by the master kinases ATM and ATR, acts as a signal for DNA damage, triggering downstream pathways that govern cell cycle checkpoints, apoptosis, and the cellular fate decision between repair and cell death. The phosphorylation of H2AX is a critical regulatory point, ensuring cell survival by promoting repair or steering cells towards apoptosis in cases of catastrophic genomic damage. Moreover, we explore the therapeutic potential of targeting H2AX in cancer treatment, leveraging its dual function as a biomarker of DNA integrity and a therapeutic target. By delineating the pathways that lead to H2AX phosphorylation and its roles in apoptosis and cell cycle control, we highlight the significance of H2AX as both a prognostic tool and a focal point for therapeutic intervention, offering insights into its utility in enhancing the efficacy of cancer treatments.
Collapse
Affiliation(s)
- Kirti S Prabhu
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar.
| | - Shilpa Kuttikrishnan
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar
| | - Nuha Ahmad
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar
| | - Ummu Habeeba
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar
| | - Zahwa Mariyam
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar
| | - Muhammad Suleman
- Laboratory of Animal Research Center, Qatar University, Doha 2713, Qatar
| | - Ajaz A Bhat
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha, Qatar
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; Laboratory of Animal Research Center, Qatar University, Doha 2713, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; Department of Biosciences, Integral University, Lucknow, Uttar Pradesh, India.
| |
Collapse
|
20
|
Ballout F, Lu H, Bhat N, Chen L, Peng D, Chen Z, Chen S, Sun X, Giordano S, Corso S, Zaika A, McDonald O, Livingstone AS, El-Rifai W. Targeting SMAD3 Improves Response to Oxaliplatin in Esophageal Adenocarcinoma Models by Impeding DNA Repair. Clin Cancer Res 2024; 30:2193-2205. [PMID: 38592373 PMCID: PMC11096039 DOI: 10.1158/1078-0432.ccr-24-0027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 02/14/2024] [Accepted: 03/15/2024] [Indexed: 04/10/2024]
Abstract
PURPOSE TGFβ signaling is implicated in the progression of most cancers, including esophageal adenocarcinoma (EAC). Emerging evidence indicates that TGFβ signaling is a key factor in the development of resistance toward cancer therapy. EXPERIMENTAL DESIGN In this study, we developed patient-derived organoids and patient-derived xenograft models of EAC and performed bioinformatics analysis combined with functional genetics to investigate the role of SMAD family member 3 (SMAD3) in EAC resistance to oxaliplatin. RESULTS Chemotherapy nonresponding patients showed enrichment of SMAD3 gene expression when compared with responders. In a randomized patient-derived xenograft experiment, SMAD3 inhibition in combination with oxaliplatin effectively diminished tumor burden by impeding DNA repair. SMAD3 interacted directly with protein phosphatase 2A (PP2A), a key regulator of the DNA damage repair protein ataxia telangiectasia mutated (ATM). SMAD3 inhibition diminished ATM phosphorylation by enhancing the binding of PP2A to ATM, causing excessive levels of DNA damage. CONCLUSIONS Our results identify SMAD3 as a promising therapeutic target for future combination strategies for the treatment of patients with EAC.
Collapse
Affiliation(s)
- Farah Ballout
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, Florida, 33136, USA
| | - Heng Lu
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, Florida, 33136, USA
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida, 33136, USA
| | - Nadeem Bhat
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, Florida, 33136, USA
| | - Lei Chen
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, Florida, 33136, USA
| | - Dunfa Peng
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, Florida, 33136, USA
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida, 33136, USA
| | - Zheng Chen
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, Florida, 33136, USA
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida, 33136, USA
| | - Steven Chen
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida, 33136, USA
- Department of Public Health Sciences, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Xiaodian Sun
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida, 33136, USA
- Department of Public Health Sciences, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Silvia Giordano
- Department of Oncology, University of Torino, Candiolo, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, 10060, Torino, Italy
| | - Simona Corso
- Department of Oncology, University of Torino, Candiolo, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, 10060, Torino, Italy
| | - Alexander Zaika
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, Florida, 33136, USA
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida, 33136, USA
- Department of Veterans Affairs, Miami Healthcare System, Miami, Florida, USA
| | - Oliver McDonald
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida, 33136, USA
- Department of Pathology, Miller School of Medicine, University of Miami, Miami, Florida, 33136, USA
| | - Alan S. Livingstone
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, Florida, 33136, USA
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida, 33136, USA
| | - Wael El-Rifai
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, Florida, 33136, USA
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida, 33136, USA
- Department of Veterans Affairs, Miami Healthcare System, Miami, Florida, USA
| |
Collapse
|
21
|
Jung WJ, Park SJ, Cha S, Kim K. Factors affecting the cleavage efficiency of the CRISPR-Cas9 system. Anim Cells Syst (Seoul) 2024; 28:75-83. [PMID: 38440123 PMCID: PMC10911232 DOI: 10.1080/19768354.2024.2322054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 02/17/2024] [Indexed: 03/06/2024] Open
Abstract
The CRISPR-Cas system stands out as a promising genome editing tool due to its cost-effectiveness and time efficiency compared to other methods. This system has tremendous potential for treating various diseases, including genetic disorders and cancer, and promotes therapeutic research for a wide range of genetic diseases. Additionally, the CRISPR-Cas system simplifies the generation of animal models, offering a more accessible alternative to traditional methods. The CRISPR-Cas9 system can be used to cleave target DNA strands that need to be corrected, causing double-strand breaks (DSBs). DNA with DSBs can then be recovered by the DNA repair pathway that the CRISPR-Cas9 system uses to edit target gene sequences. High cleavage efficiency of the CRISPR-Cas9 system is thus imperative for effective gene editing. Herein, we explore several factors affecting the cleavage efficiency of the CRISPR-Cas9 system. These factors include the GC content of the protospacer-adjacent motif (PAM) proximal and distal regions, single-guide RNA (sgRNA) properties, and chromatin state. These considerations contribute to the efficiency of genome editing.
Collapse
Affiliation(s)
- Won Jun Jung
- Department of Physiology, Korea University College of Medicine, Seoul, Republic of Korea
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Soo-Ji Park
- Department of Physiology, Korea University College of Medicine, Seoul, Republic of Korea
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Seongkwang Cha
- Department of Physiology, Korea University College of Medicine, Seoul, Republic of Korea
- Neuroscience Research Institute, Korea University College of Medicine, Seoul, Republic of Korea
| | - Kyoungmi Kim
- Department of Physiology, Korea University College of Medicine, Seoul, Republic of Korea
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
22
|
Teng QX, Lei ZN, Wang JQ, Yang Y, Wu ZX, Acharekar ND, Zhang W, Yoganathan S, Pan Y, Wurpel J, Chen ZS, Fang S. Overexpression of ABCC1 and ABCG2 confers resistance to talazoparib, a poly (ADP-Ribose) polymerase inhibitor. Drug Resist Updat 2024; 73:101028. [PMID: 38340425 DOI: 10.1016/j.drup.2023.101028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 11/24/2023] [Accepted: 11/25/2023] [Indexed: 02/12/2024]
Abstract
AIMS The overexpression of ABC transporters on cancer cell membranes is one of the most common causes of multidrug resistance (MDR). This study investigates the impact of ABCC1 and ABCG2 on the resistance to talazoparib (BMN-673), a potent poly (ADP-ribose) polymerase (PARP) inhibitor, in ovarian cancer treatment. METHODS The cell viability test was used to indicate the effect of talazoparib in different cell lines. Computational molecular docking analysis was conducted to simulate the interaction between talazoparib and ABCC1 or ABCG2. The mechanism of talazoparib resistance was investigated by constructing talazoparib-resistant subline A2780/T4 from A2780 through drug selection with gradually increasing talazoparib concentration. RESULTS Talazoparib cytotoxicity decreased in drug-selected or gene-transfected cell lines overexpressing ABCC1 or ABCG2 but can be restored by ABCC1 or ABCG2 inhibitors. Talazoparib competitively inhibited substrate drug efflux activity of ABCC1 or ABCG2. Upregulated ABCC1 and ABCG2 protein expression on the plasma membrane of A2780/T4 cells enhances resistance to other substrate drugs, which could be overcome by the knockout of either gene. In vivo experiments confirmed the retention of drug-resistant characteristics in tumor xenograft mouse models. CONCLUSIONS The therapeutic efficacy of talazoparib in cancer may be compromised by its susceptibility to MDR, which is attributed to its interactions with the ABCC1 or ABCG2 transporters. The overexpression of these transporters can potentially diminish the therapeutic impact of talazoparib in cancer treatment.
Collapse
Affiliation(s)
- Qiu-Xu Teng
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Zi-Ning Lei
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA; Department of Oncology, The Seventh Affiliated Hospital Sun Yat-sen University, Shenzhen, Guangdong 518107, PR China
| | - Jing-Quan Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Yuqi Yang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Zhuo-Xun Wu
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Nikita Dilip Acharekar
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Wei Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA; Institute of Plastic Surgery, Weifang Medical University, Weifang, Shandong 261041, PR China
| | - Sabesan Yoganathan
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Yihang Pan
- Department of Oncology, The Seventh Affiliated Hospital Sun Yat-sen University, Shenzhen, Guangdong 518107, PR China
| | - John Wurpel
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA.
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA.
| | - Shuo Fang
- Department of Oncology, The Seventh Affiliated Hospital Sun Yat-sen University, Shenzhen, Guangdong 518107, PR China.
| |
Collapse
|
23
|
Myers SH, Poppi L, Rinaldi F, Veronesi M, Ciamarone A, Previtali V, Bagnolini G, Schipani F, Ortega Martínez JA, Girotto S, Di Stefano G, Farabegoli F, Walsh N, De Franco F, Roberti M, Cavalli A. An 19F NMR fragment-based approach for the discovery and development of BRCA2-RAD51 inhibitors to pursuit synthetic lethality in combination with PARP inhibition in pancreatic cancer. Eur J Med Chem 2024; 265:116114. [PMID: 38194775 DOI: 10.1016/j.ejmech.2023.116114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/29/2023] [Accepted: 12/30/2023] [Indexed: 01/11/2024]
Abstract
The BRCA2-RAD51 interaction remains an intriguing target for cancer drug discovery due to its vital role in DNA damage repair mechanisms, which cancer cells become particularly reliant on. Moreover, RAD51 has many synthetically lethal partners, including PARP1-2, which can be exploited to induce synthetic lethality in cancer. In this study, we established a 19F-NMR-fragment based approach to identify RAD51 binders, leading to two initial hits. A subsequent SAR program identified 46 as a low micromolar inhibitor of the BRCA2-RAD51 interaction. 46 was tested in different pancreatic cancer cell lines, to evaluate its ability to inhibit the homologous recombination DNA repair pathway, mediated by BRCA2-RAD51 and trigger synthetic lethality in combination with the PARP inhibitor talazoparib, through the induction of apoptosis. Moreover, we further analyzed the 46/talazoparib combination in 3D pancreatic cancer models. Overall, 46 showed its potential as a tool to evaluate the RAD51/PARP1-2 synthetic lethality mechanism, along with providing a prospect for further inhibitors development.
Collapse
Affiliation(s)
- Samuel H Myers
- Computational and Chemical Biology, Istituto Italiano di Tecnologia, 16163, Genoa, Italy
| | - Laura Poppi
- Department of Pharmacy and Biotechnology, University of Bologna, 40126, Bologna, Italy
| | - Francesco Rinaldi
- Computational and Chemical Biology, Istituto Italiano di Tecnologia, 16163, Genoa, Italy; Department of Pharmacy and Biotechnology, University of Bologna, 40126, Bologna, Italy
| | - Marina Veronesi
- Structural Biophysics Facility, Istituto Italiano di Tecnologia, 16163, Genoa, Italy; D3 PharmaChemistry, Istituto Italiano di Tecnologia, 16163, Genoa, Italy
| | - Andrea Ciamarone
- Computational and Chemical Biology, Istituto Italiano di Tecnologia, 16163, Genoa, Italy
| | - Viola Previtali
- Computational and Chemical Biology, Istituto Italiano di Tecnologia, 16163, Genoa, Italy
| | - Greta Bagnolini
- Department of Pharmacy and Biotechnology, University of Bologna, 40126, Bologna, Italy
| | - Fabrizio Schipani
- Computational and Chemical Biology, Istituto Italiano di Tecnologia, 16163, Genoa, Italy
| | | | - Stefania Girotto
- Computational and Chemical Biology, Istituto Italiano di Tecnologia, 16163, Genoa, Italy; Structural Biophysics Facility, Istituto Italiano di Tecnologia, 16163, Genoa, Italy
| | - Giuseppina Di Stefano
- Department of Surgical and Medical Sciences, University of Bologna, 40126, Bologna, Italy
| | - Fulvia Farabegoli
- Department of Pharmacy and Biotechnology, University of Bologna, 40126, Bologna, Italy
| | - Naomi Walsh
- School of Biotechnology, Dublin City University, D09 NR58, Dublin, Ireland
| | | | - Marinella Roberti
- Department of Pharmacy and Biotechnology, University of Bologna, 40126, Bologna, Italy.
| | - Andrea Cavalli
- Computational and Chemical Biology, Istituto Italiano di Tecnologia, 16163, Genoa, Italy; Swiss Federal Institute of Technology Lausanne (EPFL), Switzerland
| |
Collapse
|
24
|
Sianipar NF, Hadisaputri YE, Assidqi K, Salam S, Yusuf M, Destiarani W, Purnamaningsih R, So IG, Takara K, Asikin Y. In silico and in vitro Characterizations of Rodent Tuber (Typhonium flagelliforme) Mutant Plant Isolates against FXR Receptor on MCF-7 Cells. J Oleo Sci 2024; 73:1349-1360. [PMID: 39358218 DOI: 10.5650/jos.ess24020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/04/2024] Open
Abstract
Typhonium flagelliforme (T. flagelliforme) is an Indonesian rodent tuber plant traditionally used to treat cancer diseases. Although gamma-ray irradiation has been used to increase the content in the chemical compounds of the T. flagelliforme plants with anticancer activity ten times effective, the specific effect of the isolated compounds from the mutant plants has never been reported yet. The potential cytotoxic agents were characterized via nuclear magnetic resonance spectroscopy, infrared spectroscopy, and mass spectrometry as stigmasterol and 7α-hydroxyl stigmasterol; and their anticancer activity was investigated. The in silico biochemical profile of the two compounds were analyzed by molecular docking and molecular dynamics simulation to confirm its interaction with the agonist binding site of Farsenoid X receptor (FXR). Stigmasterol and 7α-hydroxyl stigmasterol can act as a competitive regulator with a high-affinity for the FXR. The results also showed that stigmasterol and 7α-hydroxyl stigmasterol were the most potential and active fraction of the T. flagelliforme mutant plant against the MCF-7 human breast cancer cell line, with IC 50 value 9.13 µM and 12.97 µM, compared with cisplastin as a control about 13.20 µM. These results demonstrate the potential of stigmasterol and 7α-hydroxyl stigmasterol in T. flagelliforme mutant plants to act towards cancer diseases.
Collapse
Affiliation(s)
- Nesti Fronika Sianipar
- Biotechnology Department, Faculty of Engineering, Bina Nusantara University
- Food Biotechnology Research Center, Bina Nusantara University
| | - Yuni Elsa Hadisaputri
- Department of Biological Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran
- Biological Activity Division, Central Laboratory, Universitas Padjadjaran
| | - Khoirunnisa Assidqi
- Biotechnology Department, Faculty of Engineering, Bina Nusantara University
- Food Biotechnology Research Center, Bina Nusantara University
| | | | - Muhammad Yusuf
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran
| | - Wanda Destiarani
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran
| | | | - Idris Gautama So
- Management Department, Binus Business School, Undergraduate Program, Bina Nusantara University
| | - Kensaku Takara
- Department of Bioscience and Biotechnology, Faculty of Agriculture, University of the Ryukyus
- The United Graduate School of Agricultural Sciences, Kagoshima University
| | - Yonathan Asikin
- Department of Bioscience and Biotechnology, Faculty of Agriculture, University of the Ryukyus
- The United Graduate School of Agricultural Sciences, Kagoshima University
| |
Collapse
|
25
|
Qu Y, Qin S, Yang Z, Li Z, Liang Q, Long T, Wang W, Zeng D, Zhao Q, Dai Z, Ni Q, Zhao F, Kim W, Hou J. Targeting the DNA repair pathway for breast cancer therapy: Beyond the molecular subtypes. Biomed Pharmacother 2023; 169:115877. [PMID: 37951025 DOI: 10.1016/j.biopha.2023.115877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 11/05/2023] [Accepted: 11/07/2023] [Indexed: 11/13/2023] Open
Abstract
DNA repair is a vital mechanism in cells that protects against DNA damage caused by internal and external factors. It involves a network of signaling pathways that monitor and transmit damage signals, activating various cellular activities to repair DNA damage and maintain genomic integrity. Dysfunctions in this repair pathway are strongly associated with the development and progression of cancer. However, they also present an opportunity for targeted therapy in breast cancer. Extensive research has focused on developing inhibitors that play a crucial role in the signaling pathway of DNA repair, particularly due to the remarkable success of PARP1 inhibitors (PARPis) in treating breast cancer patients with BRCA1/2 mutations. In this review, we summarize the current research progress and clinical implementation of BRCA and BRCAness in targeted treatments for the DNA repair pathway. Additionally, we present advancements in diverse inhibitors of DNA repair, both as individual and combined approaches, for treating breast cancer. We also discuss the clinical application of DNA repair-targeted therapy for breast cancer, including the rationale, indications, and summarized clinical data for patients with different breast cancer subtypes. We assess their influence on cancer progression, survival rates, and major adverse reactions. Last, we anticipate forthcoming advancements in targeted therapy for cancer treatment and emphasize prospective areas of development.
Collapse
Affiliation(s)
- Yuting Qu
- Zunyi Medical University, No.6 Xuefu West Road, Zunyi, Guizhou Province, 563006, China; Department of Breast Surgery, Guizhou Provincial People's Hospital, NO.83 Zhongshan East Road, Guiyang, Guizhou Province 550002, China
| | - Sisi Qin
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan, 31151 Chungcheongnam-do, Republic of Korea
| | - Zhihui Yang
- Zunyi Medical University, No.6 Xuefu West Road, Zunyi, Guizhou Province, 563006, China; Department of Breast Surgery, Guizhou Provincial People's Hospital, NO.83 Zhongshan East Road, Guiyang, Guizhou Province 550002, China
| | - Zhuolin Li
- GuiZhou University Medical College, Guiyang, Guizhou Province 550025, China; Department of Breast Surgery, Guizhou Provincial People's Hospital, NO.83 Zhongshan East Road, Guiyang, Guizhou Province 550002, China
| | - Qinhao Liang
- Zunyi Medical University, No.6 Xuefu West Road, Zunyi, Guizhou Province, 563006, China; Department of Breast Surgery, Guizhou Provincial People's Hospital, NO.83 Zhongshan East Road, Guiyang, Guizhou Province 550002, China
| | - Ting Long
- Guizhou Medical University, NO.9 Beijing Road, Guiyang, Guizhou Province 550004, China; Department of Breast Surgery, Guizhou Provincial People's Hospital, NO.83 Zhongshan East Road, Guiyang, Guizhou Province 550002, China
| | - Weiyun Wang
- Guizhou University of Traditional Chinese Medicine, NO.50 Shi Dong Road, Guiyang, Guizhou Province 550002, China; Department of Breast Surgery, Guizhou Provincial People's Hospital, NO.83 Zhongshan East Road, Guiyang, Guizhou Province 550002, China
| | - Dan Zeng
- Guizhou Medical University, NO.9 Beijing Road, Guiyang, Guizhou Province 550004, China; Department of Breast Surgery, Guizhou Provincial People's Hospital, NO.83 Zhongshan East Road, Guiyang, Guizhou Province 550002, China
| | - Qing Zhao
- Guizhou Medical University, NO.9 Beijing Road, Guiyang, Guizhou Province 550004, China; Department of Breast Surgery, Guizhou Provincial People's Hospital, NO.83 Zhongshan East Road, Guiyang, Guizhou Province 550002, China
| | - Zehua Dai
- Department of Breast Surgery, Guizhou Provincial People's Hospital, NO.83 Zhongshan East Road, Guiyang, Guizhou Province 550002, China
| | - Qing Ni
- Department of Breast Surgery, Guizhou Provincial People's Hospital, NO.83 Zhongshan East Road, Guiyang, Guizhou Province 550002, China
| | - Fei Zhao
- College of Biology, Hunan University, Changsha 410082, China
| | - Wootae Kim
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan, 31151 Chungcheongnam-do, Republic of Korea.
| | - Jing Hou
- Department of Breast Surgery, Guizhou Provincial People's Hospital, NO.83 Zhongshan East Road, Guiyang, Guizhou Province 550002, China.
| |
Collapse
|
26
|
Heidari H, Lawrence DA. Climate Stressors and Physiological Dysregulations: Mechanistic Connections to Pathologies. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 21:28. [PMID: 38248493 PMCID: PMC10815632 DOI: 10.3390/ijerph21010028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 12/18/2023] [Accepted: 12/21/2023] [Indexed: 01/23/2024]
Abstract
This review delves into the complex relationship between environmental factors, their mechanistic cellular and molecular effects, and their significant impact on human health. Climate change is fueled by industrialization and the emission of greenhouse gases and leads to a range of effects, such as the redistribution of disease vectors, higher risks of disease transmission, and shifts in disease patterns. Rising temperatures pose risks to both food supplies and respiratory health. The hypothesis addressed is that environmental stressors including a spectrum of chemical and pathogen exposures as well as physical and psychological influences collectively impact genetics, metabolism, and cellular functions affecting physical and mental health. The objective is to report the mechanistic associations linking environment and health. As environmental stressors intensify, a surge in health conditions, spanning from allergies to neurodegenerative diseases, becomes evident; however, linkage to genetic-altered proteomics is more hidden. Investigations positing that environmental stressors cause mitochondrial dysfunction, metabolic syndrome, and oxidative stress, which affect missense variants and neuro- and immuno-disorders, are reported. These disruptions to homeostasis with dyslipidemia and misfolded and aggregated proteins increase susceptibility to cancers, infections, and autoimmune diseases. Proposed interventions, such as vitamin B supplements and antioxidants, target oxidative stress and may aid mitochondrial respiration and immune balance. The mechanistic interconnections of environmental stressors and disruptions in health need to be unraveled to develop strategies to protect public health.
Collapse
Affiliation(s)
- Hajar Heidari
- Department of Biomedical Sciences, University at Albany School of Public Health, Rensselaer, NY 12144, USA;
| | - David A. Lawrence
- Department of Biomedical Sciences, University at Albany School of Public Health, Rensselaer, NY 12144, USA;
- Department of Environmental Health Sciences, University at Albany School of Public Health, Rensselaer, NY 12144, USA
- Wadsworth Center, New York State Department of Health, Albany, NY 12208, USA
| |
Collapse
|
27
|
Huang L, Long Q, Su Q, Zhu X, Long X. Aflatoxin B1-DNA adducts modify the effects of post-operative adjuvant transarterial chemoembolization improving hepatocellular carcinoma prognosis. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2023; 4:780-792. [PMID: 37711588 PMCID: PMC10497403 DOI: 10.37349/etat.2023.00167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 07/10/2023] [Indexed: 09/16/2023] Open
Abstract
Aim DNA damage involves in the carcinogenesis of some cancer and may act as a target for therapeutic intervention of cancers. However, it is unclear whether aflatoxin B1 (AFB1)-DNA adducts (ADAs), an important kind of DNA damage caused by AFB1, affect the efficiency of post-operative adjuvant transarterial chemoembolization (po-TACE) treatment improving hepatocellular carcinoma (HCC) survival. Methods A hospital-based retrospective study, including 318 patients with Barcelona Clinic Liver Cancer (BCLC)-C stage HCC from high AFB1 exposure areas, to investigate the potential effects of ADAs in the tissues with HCC on po-TACE treatment. The amount of ADAs in the cancerous tissues was tested by competitive enzyme-linked immunosorbent assay (c-ELISA). Results Among these patients with HCC, the average amount of ADAs was 3.00 µmol/mol ± 1.51 µmol/mol DNA in their tissues with cancer. For these patients, increasing amount of ADAs was significantly associated with poorer overall survival (OS) and tumor reoccurrence-free survival (RFS), with corresponding death risk (DR) of 3.69 (2.78-4.91) and tumor recurrence risk (TRR) of 2.95 (2.24-3.88). The po-TACE therapy can efficiently improve their prognosis [DR = 0.59 (0.46-0.76), TRR = 0.63 (0.49-0.82)]. Interestingly, this improving role was more noticeable among these patients with high ADAs [DR = 0.36 (0.24-0.53), TRR = 0.40 (0.28-0.59)], but not among those with low ADAs (P > 0.05). Conclusions These results suggest that increasing ADAs in the cancerous tissues may be beneficial for po-TACE in ameliorating the survival of patients with HCC.
Collapse
Affiliation(s)
- Liyan Huang
- Clinicopathological Diagnosis & Research Centre, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi Zhuang Autonomous Region, China
- Departement of Pathology, Graduate School of Youjiang Medical University for Nationalities, Baise 533000, Guangxi Zhuang Autonomous Region, China
| | - Qinqin Long
- Clinicopathological Diagnosis & Research Centre, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi Zhuang Autonomous Region, China
| | - Qunying Su
- Clinicopathological Diagnosis & Research Centre, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi Zhuang Autonomous Region, China
| | - Xiaoying Zhu
- Clinicopathological Diagnosis & Research Centre, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi Zhuang Autonomous Region, China
| | - Xidai Long
- Clinicopathological Diagnosis & Research Centre, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
28
|
Barroso T, Melo-Alvim C, Ribeiro LA, Casimiro S, Costa L. Targeting Inhibitor of Apoptosis Proteins to Overcome Chemotherapy Resistance-A Marriage between Targeted Therapy and Cytotoxic Chemotherapy. Int J Mol Sci 2023; 24:13385. [PMID: 37686191 PMCID: PMC10487656 DOI: 10.3390/ijms241713385] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/20/2023] [Accepted: 08/23/2023] [Indexed: 09/10/2023] Open
Abstract
Precision oncology is the ultimate goal of cancer treatment, i.e., to treat cancer and only cancer, leaving all the remaining cells and tissues as intact as possible. Classical chemotherapy and radiotherapy, however, are still effective in many patients with cancer by effectively inducing apoptosis of cancer cells. Cancer cells might resist apoptosis via the anti-apoptotic effects of the inhibitor of apoptosis proteins. Recently, the inhibitors of those proteins have been developed with the goal of enhancing the cytotoxic effects of chemotherapy and radiotherapy, and one of them, xevinapant, has already demonstrated effectiveness in a phase II clinical trial. This class of drugs represents an example of synergism between classical cytotoxic chemo- and radiotherapy and new targeted therapy.
Collapse
Affiliation(s)
- Tiago Barroso
- Medical Oncology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, 1649-035 Lisbon, Portugal; (C.M.-A.); (L.A.R.); (L.C.)
| | - Cecília Melo-Alvim
- Medical Oncology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, 1649-035 Lisbon, Portugal; (C.M.-A.); (L.A.R.); (L.C.)
| | - Leonor Abreu Ribeiro
- Medical Oncology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, 1649-035 Lisbon, Portugal; (C.M.-A.); (L.A.R.); (L.C.)
| | - Sandra Casimiro
- Luís Costa Lab, Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisbon, Portugal;
| | - Luís Costa
- Medical Oncology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, 1649-035 Lisbon, Portugal; (C.M.-A.); (L.A.R.); (L.C.)
- Luís Costa Lab, Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisbon, Portugal;
| |
Collapse
|
29
|
Allen CH, Skillings R, Ahmed D, Sanchez SC, Altwasser K, Hilan G, Willmore WG, Chauhan V, Cassol E, Murugkar S. Investigating ionizing radiation-induced changes in breast cancer cells using stimulated Raman scattering microscopy. JOURNAL OF BIOMEDICAL OPTICS 2023; 28:076501. [PMID: 37441447 PMCID: PMC10335321 DOI: 10.1117/1.jbo.28.7.076501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 06/21/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023]
Abstract
Significance Altered lipid metabolism of cancer cells has been implicated in increased radiation resistance. A better understanding of this phenomenon may lead to improved radiation treatment planning. Stimulated Raman scattering (SRS) microscopy enables label-free and quantitative imaging of cellular lipids but has never been applied in this domain. Aim We sought to investigate the radiobiological response in human breast cancer MCF7 cells using SRS microscopy, focusing on how radiation affects lipid droplet (LD) distribution and cellular morphology. Approach MCF7 breast cancer cells were exposed to either 0 or 30 Gy (X-ray) ionizing radiation and imaged using a spectrally focused SRS microscope every 24 hrs over a 72-hr time period. Images were analyzed to quantify changes in LD area per cell, lipid and protein content per cell, and cellular morphology. Cell viability and confluency were measured using a live cell imaging system while radiation-induced lipid peroxidation was assessed using BODIPY C11 staining and flow cytometry. Results The LD area per cell and total lipid and protein intensities per cell were found to increase significantly for irradiated cells compared to control cells from 48 to 72 hrs post irradiation. Increased cell size, vacuole formation, and multinucleation were observed as well. No significant cell death was observed due to irradiation, but lipid peroxidation was found to be greater in the irradiated cells than control cells at 72 hrs. Conclusions This pilot study demonstrates the potential of SRS imaging for investigating ionizing radiation-induced changes in cancer cells without the use of fluorescent labels.
Collapse
Affiliation(s)
- Christian Harry Allen
- Carleton University, Department of Physics, Ottawa, Ontario, Canada
- Carleton University, Ottawa-Carleton Institute for Biomedical Engineering, Ottawa, Ontario, Canada
| | - Robyn Skillings
- Carleton University, Department of Health Sciences, Ottawa, Ontario, Canada
| | - Duale Ahmed
- Carleton University, Department of Health Sciences, Ottawa, Ontario, Canada
| | - Sarita Cuadros Sanchez
- Health Canada, Consumer and Clinical Radiation Protection Bureau, Ottawa, Ontario, Canada
| | - Kaitlyn Altwasser
- Health Canada, Consumer and Clinical Radiation Protection Bureau, Ottawa, Ontario, Canada
| | - George Hilan
- Carleton University, Institute of Biochemistry, Departments of Biology and Chemistry, Ottawa, Canada
| | - William G. Willmore
- Carleton University, Institute of Biochemistry, Departments of Biology and Chemistry, Ottawa, Canada
| | - Vinita Chauhan
- Health Canada, Consumer and Clinical Radiation Protection Bureau, Ottawa, Ontario, Canada
| | - Edana Cassol
- Carleton University, Department of Health Sciences, Ottawa, Ontario, Canada
| | - Sangeeta Murugkar
- Carleton University, Department of Physics, Ottawa, Ontario, Canada
- Carleton University, Ottawa-Carleton Institute for Biomedical Engineering, Ottawa, Ontario, Canada
| |
Collapse
|
30
|
Qin S, Kitty I, Hao Y, Zhao F, Kim W. Maintaining Genome Integrity: Protein Kinases and Phosphatases Orchestrate the Balancing Act of DNA Double-Strand Breaks Repair in Cancer. Int J Mol Sci 2023; 24:10212. [PMID: 37373360 DOI: 10.3390/ijms241210212] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/13/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
DNA double-strand breaks (DSBs) are the most lethal DNA damages which lead to severe genome instability. Phosphorylation is one of the most important protein post-translation modifications involved in DSBs repair regulation. Kinases and phosphatases play coordinating roles in DSB repair by phosphorylating and dephosphorylating various proteins. Recent research has shed light on the importance of maintaining a balance between kinase and phosphatase activities in DSB repair. The interplay between kinases and phosphatases plays an important role in regulating DNA-repair processes, and alterations in their activity can lead to genomic instability and disease. Therefore, study on the function of kinases and phosphatases in DSBs repair is essential for understanding their roles in cancer development and therapeutics. In this review, we summarize the current knowledge of kinases and phosphatases in DSBs repair regulation and highlight the advancements in the development of cancer therapies targeting kinases or phosphatases in DSBs repair pathways. In conclusion, understanding the balance of kinase and phosphatase activities in DSBs repair provides opportunities for the development of novel cancer therapeutics.
Collapse
Affiliation(s)
- Sisi Qin
- Department of Pathology, University of Chicago, Chicago, IL 60637, USA
| | - Ichiwa Kitty
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan 31151, Chungcheongnam-do, Republic of Korea
| | - Yalan Hao
- Analytical Instrumentation Center, Hunan University, Changsha 410082, China
| | - Fei Zhao
- College of Biology, Hunan University, Changsha 410082, China
| | - Wootae Kim
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan 31151, Chungcheongnam-do, Republic of Korea
| |
Collapse
|