1
|
Johnson OD, Paul S, Gutierrez JA, Russell WK, Ward MC. DNA damage-associated protein co-expression network in cardiomyocytes informs on tolerance to genetic variation and disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.14.607863. [PMID: 39185220 PMCID: PMC11343126 DOI: 10.1101/2024.08.14.607863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Cardiovascular disease (CVD) is associated with both genetic variants and environmental factors. One unifying consequence of the molecular risk factors in CVD is DNA damage, which must be repaired by DNA damage response proteins. However, the impact of DNA damage on global cardiomyocyte protein abundance, and its relationship to CVD risk remains unclear. We therefore treated induced pluripotent stem cell-derived cardiomyocytes with the DNA-damaging agent Doxorubicin (DOX) and a vehicle control, and identified 4,178 proteins that contribute to a network comprising 12 co-expressed modules and 403 hub proteins with high intramodular connectivity. Five modules correlate with DOX and represent distinct biological processes including RNA processing, chromatin regulation and metabolism. DOX-correlated hub proteins are depleted for proteins that vary in expression across individuals due to genetic variation but are enriched for proteins encoded by loss-of-function intolerant genes. While proteins associated with genetic risk for CVD, such as arrhythmia are enriched in specific DOX-correlated modules, DOX-correlated hub proteins are not enriched for known CVD risk proteins. Instead, they are enriched among proteins that physically interact with CVD risk proteins. Our data demonstrate that DNA damage in cardiomyocytes induces diverse effects on biological processes through protein co-expression modules that are relevant for CVD, and that the level of protein connectivity in DNA damage-associated modules influences the tolerance to genetic variation.
Collapse
Affiliation(s)
- Omar D. Johnson
- Biochemistry, Cellular and Molecular Biology Graduate Program, University of Texas Medical Branch, Galveston, Texas, USA
- MD-PhD Combined Degree Program, University of Texas Medical Branch, Galveston, Texas, USA
| | - Sayan Paul
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Jose A. Gutierrez
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, USA
| | - William K. Russell
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Michelle C. Ward
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
2
|
Martínez-Campelo L, Blanco-Verea A, López-Fernández T, Martínez-Monzonís A, Buño A, Mazón P, Zamora P, Norton N, Reddy JS, Velasco-Ruiz A, González-Neira A, Vulsteke C, Alonso-Gordoa T, Cruz R, Diz-de Almeida S, Carracedo A, González-Juanatey JR, López-Sendón J, Brion M. Meta-analysis of genome-wide association studies for cancer therapy-related cardiovascular dysfunction and functional mapping highlight an intergenic region close to TP63. Sci Rep 2024; 14:18413. [PMID: 39117733 PMCID: PMC11310459 DOI: 10.1038/s41598-024-69064-5] [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: 04/15/2024] [Accepted: 07/31/2024] [Indexed: 08/10/2024] Open
Abstract
Cancer therapy-related cardiac dysfunction (CTRCD), which commonly includes left ventricular dysfunction and heart failure, is the main adverse effect of anticancer therapy. In recent years several candidate genes studies and genome-wide association studies have identified common genetic variants associated with CTRCD, but evidence remains limited and few genetic variants are robust. A genome-wide meta-analysis of CTRCD was performed with 852 oncology patients receiving cancer therapy. DNA samples were genotyped and imputed to perform a GWAS meta-analysis for case-control (N = 852 (380 cases and 472 controls) and extreme phenotypes (N = 618 (78 cases and 472 controls) looking for genetic variants that predispose to CTRCD. The results were validated in a replicate cohort of 1,191 oncology patients (245 cases and 946 controls). Functional mapping of the replicated loci was then performed. The meta-analysis showed 9 and 17 loci suggestively associated (P-value < 1 × 10-5) with CTRCD in case-control and extreme phenotypes analyses, respectively. The 3q28 locus (rs rs7652759, P = 5.64 × 10-6) in the case-control analysis was the strongest signal, with up to 64 SNPs above the suggestive significance threshold. The rs7652759, an intergenic variant between TPRG1 and TP63 genes, was the only variant validated in the replication cohort (P-value = 0.01). Functional mapping of this significant locus revealed up to 5 new genes potentially involved in the CTRCD. We identified the intergenic region near TP63 as a novel CTRCD susceptibility locus. In the future, the genotyping of these markers could be considered in new CTRCD risk scores to improve preventive strategies in cardio-oncology.
Collapse
Affiliation(s)
- L Martínez-Campelo
- Instituto de Investigación Sanitaria de Santiago, Xenética Cardiovascular, Santiago De Compostela, Spain.
| | - A Blanco-Verea
- Instituto de Investigación Sanitaria de Santiago, Xenética Cardiovascular, Santiago De Compostela, Spain
| | - T López-Fernández
- Hospital Universitario La Paz, Servicio de Cardiología, Madrid, Spain
| | - A Martínez-Monzonís
- Hospital Clínico Universitario de Santiago de Compostela, Servicio de Cardiología, CIBER de Enfermedades Cardiovasculares, Santiago De Compostela, Spain
| | - A Buño
- Hospital Universitario La Paz, Servicio de Análisis Clínicos, IdiPaz, Madrid, Spain
| | - P Mazón
- Hospital Clínico Universitario de Santiago de Compostela, Servicio de Cardiología, CIBER de Enfermedades Cardiovasculares, Santiago De Compostela, Spain
| | - P Zamora
- Hospital Universitario La Paz, Servicio de Oncología, Madrid, Spain
| | - N Norton
- Department of Cancer Biology, Mayo Clinic Florida, Jacksonville, USA
| | - J S Reddy
- Department of Quantitative Health Sciences, Mayo Clinic Florida, Jacksonville, USA
| | - A Velasco-Ruiz
- Spanish National Cancer Research Centre, Human Cancer Genetics Programme, Madrid, Spain
| | - A González-Neira
- Spanish National Cancer Research Centre, Human Cancer Genetics Programme, Madrid, Spain
| | - C Vulsteke
- Department of Medical Oncology, Integrated Cancer Center Ghent, Ghent, Belgium
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Wilrijk, Belgium
| | - T Alonso-Gordoa
- Department of Medical Oncology, University Hospital Ramón y Cajal, Madrid, Spain
| | - R Cruz
- Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas, Universidade de Santiago de Compostela, CIBER de Enfermedades Raras, Madrid, Spain
| | - S Diz-de Almeida
- Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas, Universidade de Santiago de Compostela, CIBER de Enfermedades Raras, Madrid, Spain
| | - A Carracedo
- Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas, Universidade de Santiago de Compostela, CIBER de Enfermedades Raras, Madrid, Spain
- Fundación Pública Galega de Medicina Xenómica, Grupo de Medicina Xenómica, Santiago De Compostela, Spain
| | - J R González-Juanatey
- Hospital Clínico Universitario de Santiago de Compostela, Servicio de Cardiología, CIBER de Enfermedades Cardiovasculares, Santiago De Compostela, Spain
| | - J López-Sendón
- Instituto de Investigación Hospital Universitario La Paz (IdiPaz), Madrid, Spain
| | - M Brion
- Instituto de Investigación Sanitaria de Santiago, Xenética Cardiovascular, Santiago De Compostela, Spain
- Hospital Clínico Universitario de Santiago de Compostela, Servicio de Cardiología, CIBER de Enfermedades Cardiovasculares, Santiago De Compostela, Spain
| |
Collapse
|
3
|
Wang H, Zhang Y, Du S. Integrated analysis of lactate-related genes identifies POLRMT as a novel marker promoting the proliferation, migration and energy metabolism of hepatocellular carcinoma via Wnt/β-Catenin signaling. Am J Cancer Res 2024; 14:1316-1337. [PMID: 38590398 PMCID: PMC10998737 DOI: 10.62347/zttg4319] [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: 10/14/2023] [Accepted: 03/13/2024] [Indexed: 04/10/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is a prevalent and deadly form of cancer globally with typically unfavorable outcomes. Increasing research suggests that lactate serves as an important carbon contributor to cellular metabolism and holds a crucial part in the progression, sustenance, and treatment response of tumors. However, the contribution of lactate-related genes (LRGs) in HCC is still unclear. In this study, we analyzed TCGA datasets and screened 21 differentially expressed LRGs related to long-term survivals in HCC patients. Pan-cancer assays revealed that 21 LRGs expression exhibited a dysregulated level in man types of tumors and associated with clinical prognosis of tumor patients. The analysis of 21 LRGs successfully classified HCC samples into two molecular subtypes, and these two subtypes showed significant differences in clinical information, gene expression, and immune characteristics. Subsequently, based on the aforementioned 21 LRGs, a novel prognostic signature (DTYMK, IRAK1, POLRMT, MPV17, UQCRH, PDSS1, SLC16A3, SPP1 and LDHD) was generated by LASSO-Cox regression analysis. Survival assays demonstrated that the signature performed well in predicting the overall survival of patients with HCC. The results of Gene Set Variation Analysis indicated that the high GSVA scores were associated with poor prognosis. Moreover, we also investigated the correlation between GSVA scores and various signaling pathways in HCC. Among the nine prognostic genes, our attention focused on POLRMT which was highly expressed in HCC specimens based on TCGA datasets and several HCC cell lines. In addition, functional assays indicated that POLRMT distinctly promoted the proliferation, migration and energy metabolism of HCC cells via regulating Wnt/β-Catenin signaling. Overall, through the establishment of a novel prognostic signature, we have provided potential clinical value for assessing the prognosis of HCC patients. Furthermore, our study has identified the high expression of POLRMT in HCC and demonstrated its crucial role in HCC cell proliferation. These findings hold great importance in advancing our understanding of the pathophysiology of HCC, identifying new therapeutic targets, and improving patient survival rates.
Collapse
Affiliation(s)
- Huifen Wang
- Department of Gastroenterology, China-Japan Friendship Hospital Beijing 100029, P. R. China
| | - Yanli Zhang
- Department of Gastroenterology, China-Japan Friendship Hospital Beijing 100029, P. R. China
| | - Shiyu Du
- Department of Gastroenterology, China-Japan Friendship Hospital Beijing 100029, P. R. China
| |
Collapse
|
4
|
Abstract
Anthracycline-induced cardiotoxicity (AIC) is a serious and common side effect of anthracycline therapy. Identification of genes and genetic variants associated with AIC risk has clinical potential as a cardiotoxicity predictive tool and to allow the development of personalized therapies. In this review, we provide an overview of the function of known AIC genes identified by association studies and categorize them based on their mechanistic implication in AIC. We also discuss the importance of functional validation of AIC-associated variants in human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) to advance the implementation of genetic predictive biomarkers. Finally, we review how patient-specific hiPSC-CMs can be used to identify novel patient-relevant functional targets and for the discovery of cardioprotectant drugs to prevent AIC. Implementation of functional validation and use of hiPSC-CMs for drug discovery will identify the next generation of highly effective and personalized cardioprotectants and accelerate the inclusion of approved AIC biomarkers into clinical practice.
Collapse
Affiliation(s)
- Romina B Cejas
- Department of Pharmacology and Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA;
| | - Kateryna Petrykey
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Yadav Sapkota
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Paul W Burridge
- Department of Pharmacology and Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA;
| |
Collapse
|
5
|
Piryaei Z, Salehi Z, Ebrahimie E, Ebrahimi M, Kavousi K. Meta-analysis of integrated ChIP-seq and transcriptome data revealed genomic regions affected by estrogen receptor alpha in breast cancer. BMC Med Genomics 2023; 16:219. [PMID: 37715225 PMCID: PMC10503144 DOI: 10.1186/s12920-023-01655-z] [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: 02/24/2023] [Accepted: 09/04/2023] [Indexed: 09/17/2023] Open
Abstract
BACKGROUND The largest group of patients with breast cancer are estrogen receptor-positive (ER+) type. The estrogen receptor acts as a transcription factor and triggers cell proliferation and differentiation. Hence, investigating ER-DNA interaction genomic regions can help identify genes directly regulated by ER and understand the mechanism of ER action in cancer progression. METHODS In the present study, we employed a workflow to do a meta-analysis of ChIP-seq data of ER+ cell lines stimulated with 10 nM and 100 nM of E2. All publicly available data sets were re-analyzed with the same platform. Then, the known and unknown batch effects were removed. Finally, the meta-analysis was performed to obtain meta-differentially bound sites in estrogen-treated MCF7 cell lines compared to vehicles (as control). Also, the meta-analysis results were compared with the results of T47D cell lines for more precision. Enrichment analyses were also employed to find the functional importance of common meta-differentially bound sites and associated genes among both cell lines. RESULTS Remarkably, POU5F1B, ZNF662, ZNF442, KIN, ZNF410, and SGSM2 transcription factors were recognized in the meta-analysis but not in individual studies. Enrichment of the meta-differentially bound sites resulted in the candidacy of pathways not previously reported in breast cancer. PCGF2, HNF1B, and ZBED6 transcription factors were also predicted through the enrichment analysis of associated genes. In addition, comparing the meta-analysis results of both ChIP-seq and RNA-seq data showed that many transcription factors affected by ER were up-regulated. CONCLUSION The meta-analysis of ChIP-seq data of estrogen-treated MCF7 cell line leads to the identification of new binding sites of ER that have not been previously reported. Also, enrichment of the meta-differentially bound sites and their associated genes revealed new terms and pathways involved in the development of breast cancer which should be examined in future in vitro and in vivo studies.
Collapse
Affiliation(s)
- Zeynab Piryaei
- Department of Bioinformatics, Kish International Campus University of Tehran, Kish, Iran
- Laboratory of Complex Biological Systems and Bioinformatics (CBB), Department of Bioinformatics, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Zahra Salehi
- Laboratory of Complex Biological Systems and Bioinformatics (CBB), Department of Bioinformatics, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
- Hematology-Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Esmaeil Ebrahimie
- Genomics Research Platform, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, VIC, Australia
| | - Mansour Ebrahimi
- School of Animal and Veterinary Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Kaveh Kavousi
- Laboratory of Complex Biological Systems and Bioinformatics (CBB), Department of Bioinformatics, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran.
| |
Collapse
|
6
|
Alexandraki A, Papageorgiou E, Zacharia M, Keramida K, Papakonstantinou A, Cipolla CM, Tsekoura D, Naka K, Mazzocco K, Mauri D, Tsiknakis M, Manikis GC, Marias K, Marcou Y, Kakouri E, Konstantinou I, Daniel M, Galazi M, Kampouroglou E, Ribnikar D, Brown C, Karanasiou G, Antoniades A, Fotiadis D, Filippatos G, Constantinidou A. New Insights in the Era of Clinical Biomarkers as Potential Predictors of Systemic Therapy-Induced Cardiotoxicity in Women with Breast Cancer: A Systematic Review. Cancers (Basel) 2023; 15:3290. [PMID: 37444400 PMCID: PMC10340234 DOI: 10.3390/cancers15133290] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/09/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023] Open
Abstract
Cardiotoxicity induced by breast cancer therapies is a potentially serious complication associated with the use of various breast cancer therapies. Prediction and better management of cardiotoxicity in patients receiving chemotherapy is of critical importance. However, the management of cancer therapy-related cardiac dysfunction (CTRCD) lacks clinical evidence and is based on limited clinical studies. AIM To provide an overview of existing and potentially novel biomarkers that possess a promising predictive value for the early and late onset of CTRCD in the clinical setting. METHODS A systematic review of published studies searching for promising biomarkers for the prediction of CTRCD in patients with breast cancer was undertaken according to PRISMA guidelines. A search strategy was performed using PubMed, Google Scholar, and Scopus for the period 2013-2023. All subjects were >18 years old, diagnosed with breast cancer, and received breast cancer therapies. RESULTS The most promising biomarkers that can be used for the development of an alternative risk cardiac stratification plan for the prediction and/or early detection of CTRCD in patients with breast cancer were identified. CONCLUSIONS We highlighted the new insights associated with the use of currently available biomarkers as a standard of care for the management of CTRCD and identified potentially novel clinical biomarkers that could be further investigated as promising predictors of CTRCD.
Collapse
Affiliation(s)
- Alexia Alexandraki
- A.G. Leventis Clinical Trials Unit, Bank of Cyprus Oncology Centre, 32 Acropoleos Avenue, Nicosia 2006, Cyprus; (E.P.); (M.Z.)
| | - Elisavet Papageorgiou
- A.G. Leventis Clinical Trials Unit, Bank of Cyprus Oncology Centre, 32 Acropoleos Avenue, Nicosia 2006, Cyprus; (E.P.); (M.Z.)
| | - Marina Zacharia
- A.G. Leventis Clinical Trials Unit, Bank of Cyprus Oncology Centre, 32 Acropoleos Avenue, Nicosia 2006, Cyprus; (E.P.); (M.Z.)
| | - Kalliopi Keramida
- 2nd Department of Cardiology, Attikon University Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece;
- Cardiology Department, General Anti-Cancer Oncological Hospital, Agios Savvas, 11522 Athens, Greece
| | - Andri Papakonstantinou
- Department of Oncology-Pathology, Karolinska Institute, 17176 Stockholm, Sweden;
- Department for Breast, Endocrine Tumours and Sarcoma, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Carlo M. Cipolla
- Cardioncology and Second Opinion Division, European Institute of Oncology (IEO), IRCCS, Via Ripamonti 435, 20141 Milan, Italy;
| | - Dorothea Tsekoura
- 2nd Department of Surgery, Aretaieio University Hospital, National and Kapodistrian University of Athens, 76 Vas. Sofias Av., 11528 Athens, Greece; (D.T.); (E.K.)
| | - Katerina Naka
- 2nd Cardiology Department, University of Ioannina Medical School, 45110 Ioannina, Greece;
| | - Ketti Mazzocco
- Applied Research Division for Cognitive and Psychological Science, European Institute of Oncology IRCCS, 20139 Milan, Italy;
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Davide Mauri
- Department of Medical Oncology, University of Ioannina, 45110 Ioannina, Greece;
| | - Manolis Tsiknakis
- Department of Electrical and Computer Engineering, Hellenic Mediterranean University, 71410 Heraklion, Greece; (M.T.); (K.M.)
- Computational BioMedicine Laboratory (CBML), Institute of Computer Science, Foundation for Research and Technology Hellas (FORTH), 70013 Heraklion, Greece;
| | - Georgios C. Manikis
- Computational BioMedicine Laboratory (CBML), Institute of Computer Science, Foundation for Research and Technology Hellas (FORTH), 70013 Heraklion, Greece;
| | - Kostas Marias
- Department of Electrical and Computer Engineering, Hellenic Mediterranean University, 71410 Heraklion, Greece; (M.T.); (K.M.)
- Computational BioMedicine Laboratory (CBML), Institute of Computer Science, Foundation for Research and Technology Hellas (FORTH), 70013 Heraklion, Greece;
| | - Yiola Marcou
- Department of Medical Oncology, Bank of Cyprus Oncology Centre, 32 Acropoleos Avenue, Nicosia 2006, Cyprus; (Y.M.); (E.K.); (I.K.); (M.G.)
| | - Eleni Kakouri
- Department of Medical Oncology, Bank of Cyprus Oncology Centre, 32 Acropoleos Avenue, Nicosia 2006, Cyprus; (Y.M.); (E.K.); (I.K.); (M.G.)
| | - Ifigenia Konstantinou
- Department of Medical Oncology, Bank of Cyprus Oncology Centre, 32 Acropoleos Avenue, Nicosia 2006, Cyprus; (Y.M.); (E.K.); (I.K.); (M.G.)
| | - Maria Daniel
- Department of Radiation Oncology, Bank of Cyprus Oncology Centre, 32 Acropoleos Avenue, Nicosia 2006, Cyprus;
| | - Myria Galazi
- Department of Medical Oncology, Bank of Cyprus Oncology Centre, 32 Acropoleos Avenue, Nicosia 2006, Cyprus; (Y.M.); (E.K.); (I.K.); (M.G.)
| | - Effrosyni Kampouroglou
- 2nd Department of Surgery, Aretaieio University Hospital, National and Kapodistrian University of Athens, 76 Vas. Sofias Av., 11528 Athens, Greece; (D.T.); (E.K.)
| | - Domen Ribnikar
- Division of Medical Oncology, Institute of Oncology Ljubljana, Faculty of Medicine, University of Ljubljana, Zaloska Cesta 2, 1000 Ljubljana, Slovenia;
| | - Cameron Brown
- Translational Medicine, Stremble Ventures Ltd., 59 Christaki Kranou, Limassol 4042, Cyprus;
| | - Georgia Karanasiou
- Biomedical Research Institute, Foundation for Research and Technology, Hellas, 45500 Ioannina, Greece;
| | - Athos Antoniades
- Research and Development, Stremble Ventures Ltd., 59 Christaki Kranou, Limassol 4042, Cyprus;
| | - Dimitrios Fotiadis
- Unit of Medical Technology and Intelligent Information Systems, Department of Materials Science and Engineering, University of Ioannina, 45110 Ioannina, Greece;
| | - Gerasimos Filippatos
- Cardio-Oncology Clinic, Heart Failure Unit, Department of Cardiology, National and Kapodistrian University of Athens Medical School, Athens University Hospital Attikon, 11527 Athens, Greece;
| | - Anastasia Constantinidou
- Department of Medical Oncology, Bank of Cyprus Oncology Centre, 32 Acropoleos Avenue, Nicosia 2006, Cyprus; (Y.M.); (E.K.); (I.K.); (M.G.)
- School of Medicine, University of Cyprus, Panepistimiou 1, Aglantzia, Nicosia 2408, Cyprus
| |
Collapse
|
7
|
Treder N, Szuszczewicz N, Roszkowska A, Olędzka I, Bączek T, Bień E, Krawczyk MA, Plenis A. Magnetic Solid-Phase Microextraction Protocol Based on Didodecyldimethylammonium Bromide-Functionalized Nanoparticles for the Quantification of Epirubicin in Biological Matrices. Pharmaceutics 2023; 15:pharmaceutics15041227. [PMID: 37111712 PMCID: PMC10145736 DOI: 10.3390/pharmaceutics15041227] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 03/30/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Due to epirubicin's (EPI) narrow therapeutic index and risk of cardiotoxicity, it is critical to monitor concentrations of this drug when being used to treat cancer patients. In this study, a simple and fast magnetic solid-phase microextraction (MSPME) protocol for the determination of EPI in plasma and urine samples is developed and tested. Experiments were performed using prepared Fe3O4-based nanoparticles coated with silica and a double-chain surfactant-namely, didodecyldimethylammonium bromide (DDAB)-as a magnetic sorbent. All the prepared samples were analyzed via liquid chromatography coupled with fluorescence detection (LC-FL). The validation parameters indicated good linearity in the range of 0.001-1 µg/mL with a correlation coefficient > 0.9996 for plasma samples, and in the range of 0.001-10 µg/mL with a correlation coefficient > 0.9997 for urine samples. The limit of detection (LOD) and limit of quantification (LOQ) for both matrices were estimated at 0.0005 µg/mL and 0.001 µg/mL, respectively. The analyte recovery after sample pretreatment was 80 ± 5% for the plasma samples and 90 ± 3% for the urine samples. The developed method's applicability for monitoring EPI concentrations was evaluated by employing it to analyze real plasma and urine samples collected from a pediatric cancer patient. The obtained results confirmed the proposed MSPME-based method's usefulness, and enabled the determination of the EPI concentration-time profile in the studied patient. The miniaturization of the sampling procedure, along with the significant reduction in pre-treatment steps, make the proposed protocol a promising alternative to routine approaches to monitoring EPI levels in clinical laboratories.
Collapse
Affiliation(s)
- Natalia Treder
- Department of Analytical Chemistry, Medical University of Gdansk, 80-416 Gdansk, Poland
- Department of Pharmaceutical Chemistry, Medical University of Gdansk, 80-416 Gdansk, Poland
| | - Natalia Szuszczewicz
- Department of Analytical Chemistry, Medical University of Gdansk, 80-416 Gdansk, Poland
| | - Anna Roszkowska
- Department of Pharmaceutical Chemistry, Medical University of Gdansk, 80-416 Gdansk, Poland
| | - Ilona Olędzka
- Department of Pharmaceutical Chemistry, Medical University of Gdansk, 80-416 Gdansk, Poland
| | - Tomasz Bączek
- Department of Pharmaceutical Chemistry, Medical University of Gdansk, 80-416 Gdansk, Poland
| | - Ewa Bień
- Department of Pediatrics, Hematology and Oncology, Medical University Gdansk, 80-211 Gdansk, Poland
| | - Małgorzata Anna Krawczyk
- Department of Pediatrics, Hematology and Oncology, Medical University Gdansk, 80-211 Gdansk, Poland
| | - Alina Plenis
- Department of Analytical Chemistry, Medical University of Gdansk, 80-416 Gdansk, Poland
| |
Collapse
|
8
|
Association Studies in Clinical Pharmacogenetics. Pharmaceutics 2022; 15:pharmaceutics15010113. [PMID: 36678742 PMCID: PMC9867244 DOI: 10.3390/pharmaceutics15010113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 12/16/2022] [Indexed: 12/30/2022] Open
Abstract
In recent times, the progress of Clinical Pharmacogenetics has been remarkable [...].
Collapse
|
9
|
Current Status and Trends of Research on Anthracycline-Induced Cardiotoxicity from 2002 to 2021: A Twenty-Year Bibliometric and Visualization Analysis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:6260243. [PMID: 35993025 PMCID: PMC9388240 DOI: 10.1155/2022/6260243] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/21/2022] [Accepted: 07/24/2022] [Indexed: 12/30/2022]
Abstract
Anthracyclines constitute the cornerstone of numerous chemotherapy regimens for various cancers. However, the clinical application of anthracyclines is significantly limited to their dose-dependent cardiotoxicity. A comprehensive understanding of the current status of anthracycline-induced cardiotoxicity is necessary for in-depth research and optimal clinical protocols. Bibliometric analysis is widely applied in depicting development trends and tracking frontiers of a specific field. The present study is aimed at revealing the status and trends of anthracycline-induced cardiotoxicity during the past two decades by employing bibliometric software including R-bibliometric, VOSviewer, and CiteSpace. A total of 3504 publications concerning anthracycline-induced cardiotoxicity from 2002 to 2021 were collected from the Web of Science Core Collection database. Results showed significant growth in annual yields from 90 records in 2002 to 304 papers in 2021. The United States was the most productive country with the strongest collaboration worldwide in the field. Charles University in the Czech Republic was the institution that contributed the most papers, while 7 of the top 10 productive institutions were from the United States. The United States Department of Health and Human Services and the National Institutes of Health are the two agencies that provide financial support for more than 50% of sponsored publications. The research categories of included publications mainly belong to Oncology and Cardiac Cardiovascular Systems. The Journal of Clinical Oncology had a comprehensive impact on this research field with the highest IF value and many publications. Simunek Tomas from Charles University contributed the most publications, while Lipshultz Steven E. from the State University of New York possessed the highest H-index. In addition, the future research frontiers of anthracycline-induced cardiotoxicity might include early detection, pharmacogenomics, molecular mechanism, and cardiooncology. The present bibliometric analysis may provide a valuable reference for researchers and practitioners in future research directions.
Collapse
|
10
|
Badré A, Pan C. LINA: A Linearizing Neural Network Architecture for Accurate First-Order and Second-Order Interpretations. IEEE ACCESS : PRACTICAL INNOVATIONS, OPEN SOLUTIONS 2022; 10:36166-36176. [PMID: 35462722 PMCID: PMC9032252 DOI: 10.1109/access.2022.3163257] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
While neural networks can provide high predictive performance, it was a challenge to identify the salient features and important feature interactions used for their predictions. This represented a key hurdle for deploying neural networks in many biomedical applications that require interpretability, including predictive genomics. In this paper, linearizing neural network architecture (LINA) was developed here to provide both the first-order and the second-order interpretations on both the instance-wise and the model-wise levels. LINA combines the representational capacity of a deep inner attention neural network with a linearized intermediate representation for model interpretation. In comparison with DeepLIFT, LIME, Grad*Input and L2X, the first-order interpretation of LINA had better Spearman correlation with the ground-truth importance rankings of features in synthetic datasets. In comparison with NID and GEH, the second-order interpretation results from LINA achieved better precision for identification of the ground-truth feature interactions in synthetic datasets. These algorithms were further benchmarked using predictive genomics as a real-world application. LINA identified larger numbers of important single nucleotide polymorphisms (SNPs) and salient SNP interactions than the other algorithms at given false discovery rates. The results showed accurate and versatile model interpretation using LINA.
Collapse
Affiliation(s)
- Adrien Badré
- School of Computer Science, The University of Oklahoma, Norman, OK 73019, USA
| | - Chongle Pan
- School of Computer Science, The University of Oklahoma, Norman, OK 73019, USA
| |
Collapse
|