1
|
Amarilla‐Quintana S, Navarro P, Hernández I, Ramos A, Montero‐Calle A, Cabezas‐Sainz P, Barrero MJ, Megías D, Vilaplana‐Martí B, Epifano C, Gómez‐Dominguez D, Monzón S, Cuesta I, Sánchez L, Barderas R, García‐Donas J, Martín A, Pérez de Castro I. CRISPR targeting of FOXL2 c.402C>G mutation reduces malignant phenotype in granulosa tumor cells and identifies anti-tumoral compounds. Mol Oncol 2025; 19:1092-1116. [PMID: 39776254 PMCID: PMC11977662 DOI: 10.1002/1878-0261.13799] [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: 07/10/2024] [Revised: 11/20/2024] [Accepted: 12/24/2024] [Indexed: 01/11/2025] Open
Abstract
Forkhead box L2 (FOXL2) encodes a transcription factor essential for sex determination, and ovary development and maintenance. Mutations in this gene are implicated in syndromes involving premature ovarian failure and granulosa cell tumors (GCTs). This rare cancer accounts for less than 5% of diagnosed ovarian cancers and is causally associated with the FOXL2 c.402C>G, p.C134W mutation in 97% of the adult cases (AGCTs). In this study, we employed CRISPR technology to specifically eliminate the FOXL2 c.402C>G mutation in granulosa tumor cells. Our results show that this Cas9-mediated strategy selectively targets the mutation without affecting the wild-type allele. Granulosa cells lacking FOXL2 c.402C>G exhibit a reduced malignant phenotype, with significant changes in cell proliferation and invasion. Furthermore, these modified cells are more susceptible to dasatinib and ketoconazole. Transcriptomic and proteomic analyses reveal that CRISPR-modified granulosa tumor cells shift their expression profiles towards a wild-type-like phenotype. Additionally, this altered expression signature has led to the identification of new compounds with antiproliferative and pro-apoptotic effects on granulosa tumor cells. Our findings demonstrate the potential of CRISPR technology for the specific targeting and elimination of a mutation causing GCTs, highlighting its therapeutic promise for treating this rare ovarian cancer.
Collapse
Affiliation(s)
- Sandra Amarilla‐Quintana
- Instituto de Investigación de Enfermedades RarasInstituto de Salud Carlos IIIMadridSpain
- Programa de Doctorado en Ciencias Biomédicas y Salud Pública IMIENS‐UNED‐ISCIII, Escuela Internacional de Doctorado de la Universidad Nacional de Educación a Distancia (EIDUNED)MadridSpain
| | - Paloma Navarro
- HM Hospitales‐Centro Integral Oncológico HM Clara CampalMadridSpain
| | - Iván Hernández
- Instituto de Investigación de Enfermedades RarasInstituto de Salud Carlos IIIMadridSpain
| | - Alejandra Ramos
- Instituto de Investigación de Enfermedades RarasInstituto de Salud Carlos IIIMadridSpain
| | - Ana Montero‐Calle
- Chronic Disease Program (UFIEC), Instituto de Salud Carlos IIIMadridSpain
| | - Pablo Cabezas‐Sainz
- Department of Zoology, Genetics and Physical AnthropologyUniversidade de Santiago de CompostelaLugoSpain
| | - Maria J Barrero
- Instituto de Investigación de Enfermedades RarasInstituto de Salud Carlos IIIMadridSpain
| | - Diego Megías
- Confocal Microscopy UnitInstituto de Salud Carlos IIIMadridSpain
| | - Borja Vilaplana‐Martí
- Instituto de Investigación de Enfermedades RarasInstituto de Salud Carlos IIIMadridSpain
| | - Carolina Epifano
- Instituto de Investigación de Enfermedades RarasInstituto de Salud Carlos IIIMadridSpain
| | | | - Sara Monzón
- Bioinformatic UnitInstituto de Salud Carlos IIIMadridSpain
| | - Isabel Cuesta
- Bioinformatic UnitInstituto de Salud Carlos IIIMadridSpain
| | - Laura Sánchez
- Department of Zoology, Genetics and Physical AnthropologyUniversidade de Santiago de CompostelaLugoSpain
| | - Rodrigo Barderas
- Chronic Disease Program (UFIEC), Instituto de Salud Carlos IIIMadridSpain
| | | | - Alberto Martín
- Instituto de Investigación de Enfermedades RarasInstituto de Salud Carlos IIIMadridSpain
- Present address:
Centro de Investigación del CáncerUniversidad de SalamancaSalamancaSpain
| | | |
Collapse
|
2
|
Su Y, Ao X, Long Y, Zhang Z, Zhang M, Zhang Z, Wei M, Shan S, Lu S, Yu Y, Xu B. C1GALT1 high expression enhances the progression of glioblastoma through the EGFR-AKT/ERK cascade. Cell Signal 2024; 125:111513. [PMID: 39561885 DOI: 10.1016/j.cellsig.2024.111513] [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: 09/09/2024] [Revised: 11/08/2024] [Accepted: 11/14/2024] [Indexed: 11/21/2024]
Abstract
Core1 β1,3-galactosyltransferase (C1GALT1) is an essential glycotransferase controlling the elongation of GalNAc-type O-glycosylation and its altered expression contributes tumor progression in various cancers. However, the mechanism how C1GALT1 influences gliomas remains unclear. Here,our results from The Cancer Genome Atlas (TCGA) database, The Chinese Glioma Genome Atlas (CGGA) database and the Clinical Proteomic Tumor Analysis Consortium (CPTAC) database showed that the expression of C1GALT1 was increased in higher grade gliomas namely glioblastoma compared with low grade gliomas or non-tumor tissues and significantly associated with poor survival. Downregulation of C1GALT1 suppressed cell proliferation, invasion, and migration in glioma cell lines. Consistent with the result in vitro, C1GALT1 knockdown distinctly inhibited the weight and tumor growth in nude mice. Mechanistically, C1GALT1 knockdown decreased the level of terminal galactose O-glycosylation and phosphorylation on epidermal growth factor receptor (EGFR). Moreover, The AKT/ERK phosphorylation was attenuated in C1GALT1 knockdown cells. And C1GALT1 knockdown decreased the expression of cyclinD1, matrix metalloproteinase 9 (MMP9) through the AKT/ERK signaling pathway Furthermore, transcription factor SP1 which the expression was found to be associated the C1GALT1 expression could bind to the promoter of C1GALT1 gene and regulated its expression. In conclusion, our data show that C1GALT1 enhances the progression of glioma by regulated the O-glycosylation and phosphorylation of EGFR and the subsequent downstream AKT/ERK signaling pathway. Therefore, C1GALT1 represents a potential target for the diagnosis and treatment of glioma.
Collapse
Affiliation(s)
- Yanting Su
- School of Basic Medical Sciences, Xianning Medical Colloge, Hubei University of Science and Technology, Xianning 437100, PR China
| | - Xin Ao
- School of Pharmacy, Xianning Medical Colloge, Hubei University of Science and Technology, Xianning 437100, PR China
| | - Yunfeng Long
- School of Pharmacy, Xianning Medical Colloge, Hubei University of Science and Technology, Xianning 437100, PR China
| | - Zhengrong Zhang
- School of Pharmacy, Xianning Medical Colloge, Hubei University of Science and Technology, Xianning 437100, PR China
| | - Mingzhu Zhang
- School of Pharmacy, Xianning Medical Colloge, Hubei University of Science and Technology, Xianning 437100, PR China
| | - Zhenwang Zhang
- Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, PR China
| | - Mingjie Wei
- School of Pharmacy, Xianning Medical Colloge, Hubei University of Science and Technology, Xianning 437100, PR China
| | - Shigang Shan
- School of Basic Medical Sciences, Xianning Medical Colloge, Hubei University of Science and Technology, Xianning 437100, PR China
| | - Surui Lu
- School of Basic Medical Sciences, Xianning Medical Colloge, Hubei University of Science and Technology, Xianning 437100, PR China
| | - You Yu
- School of Basic Medical Sciences, Xianning Medical Colloge, Hubei University of Science and Technology, Xianning 437100, PR China.
| | - Bo Xu
- School of Basic Medical Sciences, Xianning Medical Colloge, Hubei University of Science and Technology, Xianning 437100, PR China.
| |
Collapse
|
3
|
Rejas-González R, Montero-Calle A, Pastora Salvador N, Crespo Carballés MJ, Ausín-González E, Sánchez-Naves J, Pardo Calderón S, Barderas R, Guzman-Aranguez A. Unraveling the nexus of oxidative stress, ocular diseases, and small extracellular vesicles to identify novel glaucoma biomarkers through in-depth proteomics. Redox Biol 2024; 77:103368. [PMID: 39326071 PMCID: PMC11462071 DOI: 10.1016/j.redox.2024.103368] [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: 08/01/2024] [Revised: 09/04/2024] [Accepted: 09/20/2024] [Indexed: 09/28/2024] Open
Abstract
Chronic ocular pathologies such as cataracts and glaucoma are emerging as an important problem for public health due to the changes in lifestyle and longevity. These age-related ocular diseases are largely mediated by oxidative stress. Small extracellular vesicles (sEVs) are involved in cell-to-cell communication and transport. There is an increasing interest about the function of small extracellular vesicles (sEVs) in the eye. However, the proteome content and characterization of sEVs released by ocular cells under pathological conditions are not yet well known. Here, we aimed to analyze the protein profile of sEVs and the intracellular protein content from two ocular cell lines (lens epithelial cells and retinal ganglion cells) exposed to oxidative stress to identify altered proteins that could serve as potential diagnostic biomarkers. The protein content was analyzed by quantitative mass spectrometry-based proteomics. Validation was performed by WB and ELISA using cell extracts and aqueous humor from cataract and glaucoma patients. After data analysis, 176 and 7 dysregulated proteins with an expression ratio≥1.5 were identified in lens epithelial cells' protein extract and sEVs, respectively, upon oxidative stress induction. In retinal ganglion cells, oxidative stress induction resulted in the dysregulation of 1033 proteins in cell extracts and 9 proteins in sEVs. In addition, by WB and ELISA, the dysregulation of proteins was mostly confirmed in aqueous humor samples from cataract or glaucoma patients in comparison to ICL individuals, with RAD23B showing high glaucoma diagnostic ability. Importantly, this work expands the knowledge of the proteome characterization of cataracts and glaucoma and provides new potential diagnostic glaucoma biomarkers.
Collapse
Affiliation(s)
- Raquel Rejas-González
- Chronic Disease Programme, UFIEC, Instituto de Salud Carlos III, 28220, Majadahonda, Madrid, Spain; Biochemistry and Molecular Biology Department, Facultad de Óptica y Optometría, Universidad Complutense de Madrid, 28037, Madrid, Spain
| | - Ana Montero-Calle
- Chronic Disease Programme, UFIEC, Instituto de Salud Carlos III, 28220, Majadahonda, Madrid, Spain
| | | | | | - Emma Ausín-González
- Opthalmology Service, Hospital Universitario Infanta Leonor, 28031, Madrid, Spain
| | | | - Sara Pardo Calderón
- Chronic Disease Programme, UFIEC, Instituto de Salud Carlos III, 28220, Majadahonda, Madrid, Spain; Biochemistry and Molecular Biology Department, Facultad de Óptica y Optometría, Universidad Complutense de Madrid, 28037, Madrid, Spain
| | - Rodrigo Barderas
- Chronic Disease Programme, UFIEC, Instituto de Salud Carlos III, 28220, Majadahonda, Madrid, Spain; CIBER of Frailty and Healthy Aging (CIBERFES), 28029, Madrid, Spain.
| | - Ana Guzman-Aranguez
- Biochemistry and Molecular Biology Department, Facultad de Óptica y Optometría, Universidad Complutense de Madrid, 28037, Madrid, Spain.
| |
Collapse
|
4
|
Montero-Calle A, Garranzo-Asensio M, Poves C, Sanz R, Dziakova J, Peláez-García A, de Los Ríos V, Martinez-Useros J, Fernández-Aceñero MJ, Barderas R. In-Depth Proteomic Analysis of Paraffin-Embedded Tissue Samples from Colorectal Cancer Patients Revealed TXNDC17 and SLC8A1 as Key Proteins Associated with the Disease. J Proteome Res 2024; 23:4802-4820. [PMID: 39441737 DOI: 10.1021/acs.jproteome.3c00749] [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: 10/25/2024]
Abstract
A deeper understanding of colorectal cancer (CRC) biology would help to identify specific early diagnostic markers. Here, we conducted quantitative proteomics on FFPE healthy, adenoma, and adenocarcinoma tissue samples from six stage I sporadic CRC patients to identify dysregulated proteins during early CRC development. Two independent quantitative 10-plex TMT experiments were separately performed. After protein extraction, trypsin digestion, and labeling, proteins were identified and quantified by using a Q Exactive mass spectrometer. A total of 2681 proteins were identified and quantified after data analysis and bioinformatics with MaxQuant and the R program. Among them, 284 and 280 proteins showed significant upregulation and downregulation (expression ratio ≥1.5 or ≤0.67, p-value ≤0.05), respectively, in adenoma and/or adenocarcinoma compared to healthy tissue. Ten dysregulated proteins were selected to study their role in CRC by WB, IHC, TMA, and ELISA using tissue and plasma samples from CRC patients, individuals with premalignant colorectal lesions (adenomas), and healthy individuals. In vitro loss-of-function cell-based assays and in vivo experiments using three CRC cell lines with different metastatic properties assessed the important roles of SLC8A1 and TXNDC17 in CRC and liver metastasis. Additionally, SLC8A1 and TXNDC17 protein levels in plasma possessed the diagnostic ability of early CRC stages.
Collapse
Affiliation(s)
- Ana Montero-Calle
- Chronic Disease Programme (UFIEC), Instituto de Salud Carlos III, Majadahonda, E-28220 Madrid, Spain
- Proteomics Core UCCTs, Instituto de Salud Carlos III, Majadahonda, E-28220 Madrid, Spain
| | - María Garranzo-Asensio
- Chronic Disease Programme (UFIEC), Instituto de Salud Carlos III, Majadahonda, E-28220 Madrid, Spain
| | - Carmen Poves
- Gastroenterology Unit, Hospital Universitario Clínico San Carlos, E-28040 Madrid, Spain
| | - Rodrigo Sanz
- Surgical Digestive Department, Hospital Universitario Clínico San Carlos, E-28040 Madrid, Spain
| | - Jana Dziakova
- Surgical Digestive Department, Hospital Universitario Clínico San Carlos, E-28040 Madrid, Spain
| | - Alberto Peláez-García
- Molecular Pathology and Therapeutic Targets Group, Hospital La Paz Institute for Health Research (IdiPAZ), E-28046 Madrid, Spain
| | | | - Javier Martinez-Useros
- Translational Oncology Division, OncoHealth Institute, Health Research Institute-Fundación Jiménez Díaz, Fundación Jiménez Díaz University Hospital/Universidad Autónoma de Madrid (IIS-FJD/UAM), E-28040 Madrid, Spain
- Area of Physiology, Department of Basic Health Sciences, Faculty of Health Sciences, Rey Juan Carlos University, Alcorcón, E-28922 Madrid, Spain
| | | | - Rodrigo Barderas
- Chronic Disease Programme (UFIEC), Instituto de Salud Carlos III, Majadahonda, E-28220 Madrid, Spain
- Proteomics Core UCCTs, Instituto de Salud Carlos III, Majadahonda, E-28220 Madrid, Spain
- CIBER of Frailty and Healthy Aging (CIBERFES), E-28029 Madrid, Spain
| |
Collapse
|
5
|
Liu Y, Chen J, Xiong J, Hu JQ, Yang LY, Sun YX, Wei Y, Zhao Y, Li X, Zheng QH, Qi WC, Liang FR. Potential cardiac-derived exosomal miRNAs involved in cardiac healing and remodeling after myocardial ischemia-reperfusion injury. Sci Rep 2024; 14:24275. [PMID: 39414956 PMCID: PMC11484883 DOI: 10.1038/s41598-024-75517-8] [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: 01/09/2024] [Accepted: 10/07/2024] [Indexed: 10/18/2024] Open
Abstract
Migratory cells exist in the heart, such as immune cells, fibroblasts, endothelial cells, etc. During myocardium injury, such as ischemia-reperfusion (MIRI), cells migrate to the site of injury to perform repair functions. However, excessive aggregation of these cells may exacerbate damage to the structure and function of the heart, such as acute myocarditis and myocardial fibrosis. Myocardial injury releases exosomes, which are a type of vesicle with signal transduction function and the miRNA carried by exosomes can control cell migration function. Therefore, regulating this migratory cell population through cardiac-derived exosomal miRNA is crucial for protecting and maintaining cardiac function. Through whole transcriptome RNA sequencing, exosomal miRNA sequencing and single-cell dataset analysis, we (1) determined the potential molecular regulatory role of the lncRNA‒miRNA‒mRNA axis in MIRI, (2) screened four important exosomal miRNAs that could be released by cardiac tissue, and (3) screened seven genes related to cell locomotion that are regulated by four miRNAs, among which Tradd and Ephb6 may be specific for promoting migration of different cells of myocardial tissue in myocardial infarct. We generated a core miRNA‒mRNA network based on the functions of the target genes, which may be not only a target for cardiac repair but also a potential diagnostic marker for interactions between the heart and other tissues or organs. In conclusion, we elucidated the potential mechanism of MIRI in cardiac remodeling from the perspective of cell migration, and inhibition of cellular overmigration based on this network may provide new therapeutic targets for MIRI and to prevent MIRI from developing into other diseases.
Collapse
Affiliation(s)
- Yu Liu
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Jiao Chen
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Jian Xiong
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Jin-Qun Hu
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Li-Yuan Yang
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Yu-Xin Sun
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Ying Wei
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Yi Zhao
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Xiao Li
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Qian-Hua Zheng
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Wen-Chuan Qi
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China.
| | - Fan-Rong Liang
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China.
- Sichuan Clinical Medicine Research Center of Acupuncture-Moxibustion, Chengdu, 610075, China.
| |
Collapse
|
6
|
Rejas-González R, Montero-Calle A, Valverde A, Salvador NP, Carballés MJC, Ausín-González E, Sánchez-Naves J, Campuzano S, Barderas R, Guzman-Aranguez A. Proteomics Analyses of Small Extracellular Vesicles of Aqueous Humor: Identification and Validation of GAS6 and SPP1 as Glaucoma Markers. Int J Mol Sci 2024; 25:6995. [PMID: 39000104 PMCID: PMC11241616 DOI: 10.3390/ijms25136995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 06/21/2024] [Accepted: 06/21/2024] [Indexed: 07/16/2024] Open
Abstract
Cataracts and glaucoma account for a high percentage of vision loss and blindness worldwide. Small extracellular vesicles (sEVs) are released into different body fluids, including the eye's aqueous humor. Information about their proteome content and characterization in ocular pathologies is not yet well established. In this study, aqueous humor sEVs from healthy individuals, cataracts, and glaucoma patients were studied, and their specific protein profiles were characterized. Moreover, the potential of identified proteins as diagnostic glaucoma biomarkers was evaluated. The protein content of sEVs from patients' aqueous humor with cataracts and glaucoma compared to healthy individuals was analyzed by quantitative proteomics. Validation was performed by western blot (WB) and ELISA. A total of 828 peptides and 192 proteins were identified and quantified. After data analysis with the R program, 8 significantly dysregulated proteins from aqueous humor sEVs in cataracts and 16 in glaucoma showed an expression ratio ≥ 1.5. By WB and ELISA using directly aqueous humor samples, the dysregulation of 9 proteins was mostly confirmed. Importantly, GAS6 and SPP1 showed high diagnostic ability of glaucoma, which in combination allowed for discriminating glaucoma patients from control individuals with an area under the curve of 76.1% and a sensitivity of 65.6% and a specificity of 87.7%.
Collapse
Affiliation(s)
- Raquel Rejas-González
- Chronic Disease Programme, UFIEC, Instituto de Salud Carlos III, 28220 Majadahonda, Madrid, Spain; (R.R.-G.); (A.M.-C.)
- Biochemistry and Molecular Biology Department, Facultad de Óptica y Optometría, Universidad Complutense de Madrid, 28037 Madrid, Spain
| | - Ana Montero-Calle
- Chronic Disease Programme, UFIEC, Instituto de Salud Carlos III, 28220 Majadahonda, Madrid, Spain; (R.R.-G.); (A.M.-C.)
| | - Alejandro Valverde
- Analytical Chemistry Department, Facultad de CC. Químicas, Universidad Complutense de Madrid, Pza. de las Ciencias 2, 28040 Madrid, Spain; (A.V.); (S.C.)
| | - Natalia Pastora Salvador
- Opthalmology Service, Hospital Universitario Infanta Leonor, 28031 Madrid, Spain; (N.P.S.); (M.J.C.C.); (E.A.-G.)
| | - María José Crespo Carballés
- Opthalmology Service, Hospital Universitario Infanta Leonor, 28031 Madrid, Spain; (N.P.S.); (M.J.C.C.); (E.A.-G.)
| | - Emma Ausín-González
- Opthalmology Service, Hospital Universitario Infanta Leonor, 28031 Madrid, Spain; (N.P.S.); (M.J.C.C.); (E.A.-G.)
| | | | - Susana Campuzano
- Analytical Chemistry Department, Facultad de CC. Químicas, Universidad Complutense de Madrid, Pza. de las Ciencias 2, 28040 Madrid, Spain; (A.V.); (S.C.)
| | - Rodrigo Barderas
- Chronic Disease Programme, UFIEC, Instituto de Salud Carlos III, 28220 Majadahonda, Madrid, Spain; (R.R.-G.); (A.M.-C.)
- CIBER of Frailty and Healthy Aging (CIBERFES), 28029 Madrid, Spain
| | - Ana Guzman-Aranguez
- Biochemistry and Molecular Biology Department, Facultad de Óptica y Optometría, Universidad Complutense de Madrid, 28037 Madrid, Spain
| |
Collapse
|
7
|
Huang L, Li Z, Xu Z, Yu R, Ding C, Sun T, Kong L, Xia Z. C1GALT1 induces the carcinogenesis of thyroid cancer through regulation by miR-141-3p and GLUT1. Heliyon 2024; 10:e31778. [PMID: 38845937 PMCID: PMC11153184 DOI: 10.1016/j.heliyon.2024.e31778] [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: 02/28/2024] [Revised: 05/20/2024] [Accepted: 05/21/2024] [Indexed: 06/09/2024] Open
Abstract
Core 1 β 1,3-galactosyltransferase 1 (C1GALT1) acts as an important glycosyltransferase in the occurrence and development of tumor glycosylation. However, the regulatory mechanisms of C1GALT1 in thyroid cancer (TC) is still unclear. In this study, we discovered that the expression level of C1GALT1 was significantly increased in thyroid adenocarcinoma tissues and cell lines (p < 0.01). Meanwhile, gene silencing of C1GALT1 inhibited the proliferation (CCK-8 assay), migration (wound healing), and invasion (Transwell) of TC cells (p < 0.05). Further investigation indicated that miR-141-3p had a negative correlation with C1GALT1 and suppressed cancer carcinogenesis in TC cells. Moreover, we first found that glucose transporter 1 (GLUT1) was a downstream element of C1GALT1 and was positively correlated with C1GALT1 levels in TC. The GLUT1 could reverse the inhibitory effects of siRNA C1GALT1 on cell development (p < 0.05). These data suggest that the miR-141-3p/C1GALT1/GLUT1 axis plays an essential role during TC progression and may be a probable biomarker or therapeutic target for thyroid cancer patients.
Collapse
Affiliation(s)
- Li Huang
- Department of Pathology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan, China
| | - Zhen Li
- Department of Pathology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan, China
| | - Ziguang Xu
- Department of Pathology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan, China
| | - Ruili Yu
- Department of Pathology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan, China
| | - Chao Ding
- Department of Thyroid, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan, China
| | - Tingyi Sun
- Department of Pathology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan, China
| | - Lingfei Kong
- Department of Pathology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan, China
| | - Zhengchao Xia
- Department of Pharmacy, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan, China
| |
Collapse
|
8
|
Chen Y, Ji Y, Shen L, Li Y, Ren Y, Shi H, Li Y, Wu Y. High core 1β1,3-galactosyltransferase 1 expression is associated with poor prognosis and promotes cellular radioresistance in lung adenocarcinoma. J Cancer Res Clin Oncol 2024; 150:214. [PMID: 38662050 PMCID: PMC11045595 DOI: 10.1007/s00432-024-05745-y] [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: 02/09/2024] [Accepted: 04/07/2024] [Indexed: 04/26/2024]
Abstract
PURPOSE Core 1β1,3-galactosyltransferase 1 (C1GALT1) exhibits elevated expression in multiple cancers. The present study aimed to elucidate the clinical significance of C1GALT1 aberrant expression and its impact on radiosensitivity in lung adenocarcinoma (LUAD). METHODS The C1GALT1 expression and its clinical relevance were investigated through public databases and LUAD tissue microarray analyses. A549 and H1299 cells with either C1GALT1 knockdown or overexpression were further assessed through colony formation, gamma-H2A histone family member X immunofluorescence, 5-ethynyl-2'-deoxyuridine incorporation, and flow cytometry assays. Bioinformatics analysis was used to explore single cell sequencing data, revealing the influence of C1GALT1 on cancer-associated cellular states. Vimentin, N-cadherin, and E-cadherin protein levels were measured through western blotting. RESULTS The expression of C1GALT1 was significantly higher in LUAD tissues than in adjacent non-tumor tissues both at mRNA and protein level. High expression of C1GALT1 was correlated with lymph node metastasis, advanced T stage, and poor survival, and was an independent risk factor for overall survival. Radiation notably upregulated C1GALT1 expression in A549 and H1299 cells, while radiosensitivity was increased following C1GALT1 knockdown and decreased following overexpression. Experiment results showed that overexpression of C1GALT1 conferred radioresistance, promoting DNA repair, cell proliferation, and G2/M phase arrest, while inhibiting apoptosis and decreasing E-cadherin expression, alongside upregulating vimentin and N-cadherin in A549 and H1299 cells. Conversely, C1GALT1 knockdown had opposing effects. CONCLUSION Elevated C1GALT1 expression in LUAD is associated with an unfavorable prognosis and contributes to increased radioresistance potentially by affecting DNA repair, cell proliferation, cell cycle regulation, and epithelial-mesenchymal transition (EMT).
Collapse
Affiliation(s)
- Yong Chen
- Department of Medical Oncology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225009, Jiangsu, People's Republic of China
| | - Yanyan Ji
- Department of Medical Oncology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225009, Jiangsu, People's Republic of China
| | - Lin Shen
- Department of Medical Oncology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225009, Jiangsu, People's Republic of China
| | - Ying Li
- Department of Medical Oncology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225009, Jiangsu, People's Republic of China
| | - Yue Ren
- Department of Medical Oncology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225009, Jiangsu, People's Republic of China
| | - Hongcan Shi
- Department of Cardiothoracic Surgery, Medical College of Yangzhou University, Yangzhou University, Yangzhou, 225009, Jiangsu, People's Republic of China
| | - Yue Li
- Department of Medical Oncology, Clinical College of Dalian Medical University, Yangzhou, 225009, Jiangsu, People's Republic of China
| | - Yunjiang Wu
- Department of Thoracic Surgery, Affiliated Hospital of Yangzhou University, Yangzhou University, No. 368 Hanjiang Road, Yangzhou, 225009, Jiangsu, People's Republic of China.
| |
Collapse
|
9
|
Li L, Wang B, Zhao S, Xiong Q, Cheng A. The role of ANXA1 in the tumor microenvironment. Int Immunopharmacol 2024; 131:111854. [PMID: 38479155 DOI: 10.1016/j.intimp.2024.111854] [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: 01/25/2024] [Revised: 03/02/2024] [Accepted: 03/10/2024] [Indexed: 04/10/2024]
Abstract
Annexin A1 (ANXA1) is widely expressed in a variety of body tissues and cells and is also involved in tumor development through multiple pathways. The invasion, metastasis, and immune escape of tumor cells depend on the interaction between tumor cells and their surrounding environment. Research shows that ANXA1 can act on a variety of cells in the tumor microenvironment (TME), and subsequently affect the proliferation, invasion and metastasis of tumors. This article describes the role of ANXA1 in the various components of the tumor microenvironment and its mechanism of action, as well as the existing clinical treatment measures related to ANXA1. These findings provide insight for the further design of strategies targeting ANXA1 for the diagnosis and treatment of malignant tumors.
Collapse
Affiliation(s)
- Lanxin Li
- Hunan Engineering Research Center for Early Diagnosis and Treatment of Liver Cancer, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, China
| | - Baiqi Wang
- The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Shuang Zhao
- Hunan Engineering Research Center for Early Diagnosis and Treatment of Liver Cancer, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, China
| | - Qinglin Xiong
- Hunan Engineering Research Center for Early Diagnosis and Treatment of Liver Cancer, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, China
| | - Ailan Cheng
- Hunan Engineering Research Center for Early Diagnosis and Treatment of Liver Cancer, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, China.
| |
Collapse
|
10
|
Tian H, Yu JL, Chu X, Guan Q, Liu J, Liu Y. Unraveling the role of C1GALT1 in abnormal glycosylation and colorectal cancer progression. Front Oncol 2024; 14:1389713. [PMID: 38699634 PMCID: PMC11063370 DOI: 10.3389/fonc.2024.1389713] [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: 02/22/2024] [Accepted: 03/25/2024] [Indexed: 05/05/2024] Open
Abstract
C1GALT1 plays a pivotal role in colorectal cancer (CRC) development and progression through its involvement in various molecular mechanisms. This enzyme is central to the O-glycosylation process, producing tumor-associated carbohydrate antigens (TACA) like Tn and sTn, which are linked to cancer metastasis and poor prognosis. The interaction between C1GALT1 and core 3 synthase is crucial for the synthesis of core 3 O-glycans, essential for gastrointestinal health and mucosal barrier integrity. Aberrations in this pathway can lead to CRC development. Furthermore, C1GALT1's function is significantly influenced by its molecular chaperone, Cosmc, which is necessary for the proper folding of T-synthase. Dysregulation in this complex interaction contributes to abnormal O-glycan regulation, facilitating cancer progression. Moreover, C1GALT1 affects downstream signaling pathways and cellular behaviors, such as the epithelial-mesenchymal transition (EMT), by modifying O-glycans on key receptors like FGFR2, enhancing cancer cell invasiveness and metastatic potential. Additionally, the enzyme's relationship with MUC1, a mucin protein with abnormal glycosylation in CRC, highlights its role in cancer cell immune evasion and metastasis. Given these insights, targeting C1GALT1 presents a promising therapeutic strategy for CRC, necessitating further research to develop targeted inhibitors or activators. Future efforts should also explore C1GALT1's potential as a biomarker for early diagnosis, prognosis, and treatment response monitoring in CRC, alongside investigating combination therapies to improve patient outcomes.
Collapse
Affiliation(s)
- Hong Tian
- Department of Oncology, Fourth People’s Hospital in Shenyang, China Medical University, Shenyang, China
| | - Jia-Li Yu
- Department of Gastroenterology, The First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, China
| | - Xiaoli Chu
- Department of Oncology, Fourth People’s Hospital in Shenyang, China Medical University, Shenyang, China
| | - Qi Guan
- Department of Oncology, Fourth People’s Hospital in Shenyang, China Medical University, Shenyang, China
| | - Juan Liu
- Department of Oncology, Fourth People’s Hospital in Shenyang, China Medical University, Shenyang, China
| | - Ying Liu
- Department of Oncology, Fourth People’s Hospital in Shenyang, China Medical University, Shenyang, China
| |
Collapse
|
11
|
Bao X, Liang Y, Chang H, Cai T, Feng B, Gordon K, Zhu Y, Shi H, He Y, Xie L. Targeting proprotein convertase subtilisin/kexin type 9 (PCSK9): from bench to bedside. Signal Transduct Target Ther 2024; 9:13. [PMID: 38185721 PMCID: PMC10772138 DOI: 10.1038/s41392-023-01690-3] [Citation(s) in RCA: 54] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 09/27/2023] [Accepted: 10/27/2023] [Indexed: 01/09/2024] Open
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) has evolved as a pivotal enzyme in lipid metabolism and a revolutionary therapeutic target for hypercholesterolemia and its related cardiovascular diseases (CVD). This comprehensive review delineates the intricate roles and wide-ranging implications of PCSK9, extending beyond CVD to emphasize its significance in diverse physiological and pathological states, including liver diseases, infectious diseases, autoimmune disorders, and notably, cancer. Our exploration offers insights into the interaction between PCSK9 and low-density lipoprotein receptors (LDLRs), elucidating its substantial impact on cholesterol homeostasis and cardiovascular health. It also details the evolution of PCSK9-targeted therapies, translating foundational bench discoveries into bedside applications for optimized patient care. The advent and clinical approval of innovative PCSK9 inhibitory therapies (PCSK9-iTs), including three monoclonal antibodies (Evolocumab, Alirocumab, and Tafolecimab) and one small interfering RNA (siRNA, Inclisiran), have marked a significant breakthrough in cardiovascular medicine. These therapies have demonstrated unparalleled efficacy in mitigating hypercholesterolemia, reducing cardiovascular risks, and have showcased profound value in clinical applications, offering novel therapeutic avenues and a promising future in personalized medicine for cardiovascular disorders. Furthermore, emerging research, inclusive of our findings, unveils PCSK9's potential role as a pivotal indicator for cancer prognosis and its prospective application as a transformative target for cancer treatment. This review also highlights PCSK9's aberrant expression in various cancer forms, its association with cancer prognosis, and its crucial roles in carcinogenesis and cancer immunity. In conclusion, this synthesized review integrates existing knowledge and novel insights on PCSK9, providing a holistic perspective on its transformative impact in reshaping therapeutic paradigms across various disorders. It emphasizes the clinical value and effect of PCSK9-iT, underscoring its potential in advancing the landscape of biomedical research and its capabilities in heralding new eras in personalized medicine.
Collapse
Affiliation(s)
- Xuhui Bao
- Institute of Therapeutic Cancer Vaccines, Fudan University Pudong Medical Center, Shanghai, China.
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China.
- Department of Oncology, Fudan University Pudong Medical Center, Shanghai, China.
- Center for Clinical Research, Fudan University Pudong Medical Center, Shanghai, China.
- Clinical Research Center for Cell-based Immunotherapy, Fudan University, Shanghai, China.
- Department of Pathology, Duke University Medical Center, Durham, NC, USA.
| | - Yongjun Liang
- Center for Medical Research and Innovation, Fudan University Pudong Medical Center, Shanghai, China
| | - Hanman Chang
- Institute for Food Safety and Health, Illinois Institute of Technology, Chicago, IL, USA
| | - Tianji Cai
- Department of Sociology, University of Macau, Taipa, Macau, China
| | - Baijie Feng
- Department of Oncology, Fudan University Pudong Medical Center, Shanghai, China
| | - Konstantin Gordon
- Medical Institute, Peoples' Friendship University of Russia, Moscow, Russia
- A. Tsyb Medical Radiological Research Center, Obninsk, Russia
| | - Yuekun Zhu
- Department of Colorectal Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Hailian Shi
- Shanghai Key Laboratory of Compound Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Zhangjiang Hi-tech Park, Shanghai, China
| | - Yundong He
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China.
| | - Liyi Xie
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
12
|
Montero-Calle A, Jiménez de Ocaña S, Benavente-Naranjo R, Rejas-González R, Bartolomé RA, Martínez-Useros J, Sanz R, Dziaková J, Fernández-Aceñero MJ, Mendiola M, Casal JI, Peláez-García A, Barderas R. Functional Proteomics Characterization of the Role of SPRYD7 in Colorectal Cancer Progression and Metastasis. Cells 2023; 12:2548. [PMID: 37947626 PMCID: PMC10648221 DOI: 10.3390/cells12212548] [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: 07/04/2023] [Revised: 10/12/2023] [Accepted: 10/24/2023] [Indexed: 11/12/2023] Open
Abstract
SPRY domain-containing protein 7 (SPRYD7) is a barely known protein identified via spatial proteomics as being upregulated in highly metastatic-to-liver KM12SM colorectal cancer (CRC) cells in comparison to its isogenic poorly metastatic KM12C CRC cells. Here, we aimed to analyze SPRYD7's role in CRC via functional proteomics. Through immunohistochemistry, the overexpression of SPRYD7 was observed to be associated with the poor survival of CRC patients and with an aggressive and metastatic phenotype. Stable SPRYD7 overexpression was performed in KM12C and SW480 poorly metastatic CRC cells and in their isogenic highly metastatic-to-liver-KM12SM-and-to-lymph-nodes SW620 CRC cells, respectively. Upon upregulation of SPRYD7, in vitro and in vivo functional assays confirmed a key role of SPRYD7 in the invasion and migration of CRC cells and in liver homing and tumor growth. Additionally, transient siRNA SPRYD7 silencing allowed us to confirm in vitro functional results. Furthermore, SPRYD7 was observed as an inductor of angiogenesis. In addition, the dysregulated SPRYD7-associated proteome and SPRYD7 interactors were elucidated via 10-plex TMT quantitative proteins, immunoproteomics, and bioinformatics. After WB validation, the biological pathways associated with the stable overexpression of SPRYD7 were visualized. In conclusion, it was demonstrated here that SPRYD7 is a novel protein associated with CRC progression and metastasis. Thus, SPRYD7 and its interactors might be of relevance in identifying novel therapeutic targets for advanced CRC.
Collapse
Affiliation(s)
- Ana Montero-Calle
- Chronic Disease Programme (UFIEC), Instituto de Salud Carlos III, 28220 Madrid, Spain; (S.J.d.O.); (R.B.-N.); (R.R.-G.)
| | - Sofía Jiménez de Ocaña
- Chronic Disease Programme (UFIEC), Instituto de Salud Carlos III, 28220 Madrid, Spain; (S.J.d.O.); (R.B.-N.); (R.R.-G.)
| | - Ruth Benavente-Naranjo
- Chronic Disease Programme (UFIEC), Instituto de Salud Carlos III, 28220 Madrid, Spain; (S.J.d.O.); (R.B.-N.); (R.R.-G.)
| | - Raquel Rejas-González
- Chronic Disease Programme (UFIEC), Instituto de Salud Carlos III, 28220 Madrid, Spain; (S.J.d.O.); (R.B.-N.); (R.R.-G.)
| | - Rubén A. Bartolomé
- Centro de Investigaciones Biológicas Margarita Salas, CSIC, 28040 Madrid, Spain;
| | - Javier Martínez-Useros
- Translational Oncology Division, OncoHealth Institute, Health Research Institute-University Hospital Fundación Jiménez Díaz-Universidad Autónoma de Madrid, 28040 Madrid, Spain;
| | - Rodrigo Sanz
- Surgical Digestive Department, Hospital Universitario Clínico San Carlos, 28040 Madrid, Spain; (R.S.); (J.D.)
| | - Jana Dziaková
- Surgical Digestive Department, Hospital Universitario Clínico San Carlos, 28040 Madrid, Spain; (R.S.); (J.D.)
| | | | - Marta Mendiola
- Molecular Pathology and Therapeutic Targets Group, La Paz University Hospital (IdiPAZ), 28046 Madrid, Spain; (M.M.); (A.P.-G.)
| | - José Ignacio Casal
- Centro de Investigaciones Biológicas Margarita Salas, CSIC, 28040 Madrid, Spain;
| | - Alberto Peláez-García
- Molecular Pathology and Therapeutic Targets Group, La Paz University Hospital (IdiPAZ), 28046 Madrid, Spain; (M.M.); (A.P.-G.)
| | - Rodrigo Barderas
- Chronic Disease Programme (UFIEC), Instituto de Salud Carlos III, 28220 Madrid, Spain; (S.J.d.O.); (R.B.-N.); (R.R.-G.)
| |
Collapse
|
13
|
Montero-Calle A, Garranzo-Asensio M, Rejas-González R, Feliu J, Mendiola M, Peláez-García A, Barderas R. Benefits of FAIMS to Improve the Proteome Coverage of Deteriorated and/or Cross-Linked TMT 10-Plex FFPE Tissue and Plasma-Derived Exosomes Samples. Proteomes 2023; 11:35. [PMID: 37987315 PMCID: PMC10661291 DOI: 10.3390/proteomes11040035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/20/2023] [Accepted: 10/20/2023] [Indexed: 11/22/2023] Open
Abstract
The proteome characterization of complex, deteriorated, or cross-linked protein mixtures as paired clinical FFPE or exosome samples isolated from low plasma volumes (250 µL) might be a challenge. In this work, we aimed at investigating the benefits of FAIMS technology coupled to the Orbitrap Exploris 480 mass spectrometer for the TMT quantitative proteomics analyses of these complex samples in comparison to the analysis of protein extracts from cells, frozen tissue, and exosomes isolated from large volume plasma samples (3 mL). TMT experiments were performed using a two-hour gradient LC-MS/MS with or without FAIMS and two compensation voltages (CV = -45 and CV = -60). In the TMT experiments of cells, frozen tissue, or exosomes isolated from large plasma volumes (3 mL) with FAIMS, a limited increase in the number of identified and quantified proteins accompanied by a decrease in the number of peptides identified and quantified was observed. However, we demonstrated here a noticeable improvement (>100%) in the number of peptide and protein identifications and quantifications for the plasma exosomes isolated from low plasma volumes (250 µL) and FFPE tissue samples in TMT experiments with FAIMS in comparison to the LC-MS/MS analysis without FAIMS. Our results highlight the potential of mass spectrometry analyses with FAIMS to increase the depth into the proteome of complex samples derived from deteriorated, cross-linked samples and/or those where the material was scarce, such as FFPE and plasma-derived exosomes from low plasma volumes (250 µL), which might aid in the characterization of their proteome and proteoforms and in the identification of dysregulated proteins that could be used as biomarkers.
Collapse
Affiliation(s)
- Ana Montero-Calle
- Chronic Disease Programme (UFIEC), Instituto de Salud Carlos III, 28220 Majadahonda, Spain; (M.G.-A.); (R.R.-G.)
| | - María Garranzo-Asensio
- Chronic Disease Programme (UFIEC), Instituto de Salud Carlos III, 28220 Majadahonda, Spain; (M.G.-A.); (R.R.-G.)
| | - Raquel Rejas-González
- Chronic Disease Programme (UFIEC), Instituto de Salud Carlos III, 28220 Majadahonda, Spain; (M.G.-A.); (R.R.-G.)
| | - Jaime Feliu
- Translational Oncology Group, La Paz University Hospital (IdiPAZ), 28046 Madrid, Spain;
- Center for Biomedical Research in the Cancer Network (CIBERONC), Instituto de Salud Carlos III, 28046 Madrid, Spain;
| | - Marta Mendiola
- Center for Biomedical Research in the Cancer Network (CIBERONC), Instituto de Salud Carlos III, 28046 Madrid, Spain;
- Molecular Pathology and Therapeutic Targets Group, La Paz University Hospital (IdiPAZ), 28046 Madrid, Spain;
| | - Alberto Peláez-García
- Molecular Pathology and Therapeutic Targets Group, La Paz University Hospital (IdiPAZ), 28046 Madrid, Spain;
| | - Rodrigo Barderas
- Chronic Disease Programme (UFIEC), Instituto de Salud Carlos III, 28220 Majadahonda, Spain; (M.G.-A.); (R.R.-G.)
| |
Collapse
|
14
|
Pun FW, Ozerov IV, Zhavoronkov A. AI-powered therapeutic target discovery. Trends Pharmacol Sci 2023; 44:561-572. [PMID: 37479540 DOI: 10.1016/j.tips.2023.06.010] [Citation(s) in RCA: 95] [Impact Index Per Article: 47.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/20/2023] [Accepted: 06/23/2023] [Indexed: 07/23/2023]
Abstract
Disease modeling and target identification are the most crucial initial steps in drug discovery, and influence the probability of success at every step of drug development. Traditional target identification is a time-consuming process that takes years to decades and usually starts in an academic setting. Given its advantages of analyzing large datasets and intricate biological networks, artificial intelligence (AI) is playing a growing role in modern drug target identification. We review recent advances in target discovery, focusing on breakthroughs in AI-driven therapeutic target exploration. We also discuss the importance of striking a balance between novelty and confidence in target selection. An increasing number of AI-identified targets are being validated through experiments and several AI-derived drugs are entering clinical trials; we highlight current limitations and potential pathways for moving forward.
Collapse
Affiliation(s)
- Frank W Pun
- Insilico Medicine Hong Kong Ltd., Hong Kong Science and Technology Park, New Territories, Hong Kong
| | - Ivan V Ozerov
- Insilico Medicine Hong Kong Ltd., Hong Kong Science and Technology Park, New Territories, Hong Kong
| | - Alex Zhavoronkov
- Insilico Medicine Hong Kong Ltd., Hong Kong Science and Technology Park, New Territories, Hong Kong; Insilico Medicine MENA, 6F IRENA Building, Abu Dhabi, United Arab Emirates; Buck Institute for Research on Aging, Novato, CA, USA.
| |
Collapse
|
15
|
Ozcelikay G, Gamella M, Solís-Fernández G, Barderas R, Pingarrón JM, Campuzano S, Ozkan SA. Electrochemical bioplatform for the determination of the most common and carcinogenic human papillomavirus DNA. J Pharm Biomed Anal 2023; 231:115411. [PMID: 37094410 DOI: 10.1016/j.jpba.2023.115411] [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: 03/20/2023] [Revised: 04/18/2023] [Accepted: 04/18/2023] [Indexed: 04/26/2023]
Abstract
Nucleic acid-based analytical bioplatforms have gained importance as diagnostic tests for genomics and as early detection tools for diseases such as cancer. In this context, we report the development of an amperometric bioplatform for the determination of a specific human papillomavirus type 16 (HPV16) sequence. The bioplatform utilizes an immune-nucleic acid hybrid-sandwich assay. A biotinylated RNA capture probe (RNAbCp), complementary to the selected HPV16 target DNA sequence, was immobilised on the surface of streptavidin coated magnetic microbeads (Strep-MBs). The RNA/DNA heteroduplex resulting from the hybridization of the RNAbCP and the HPV16 target sequence was recognised by a commercial antibody that specifically bound to the heteroduplex (AbDNA-RNA). A horseradish-peroxide labeled secondary antibody (antiIgG-HRP) was used for the detection of AbDNA-RNA. Relying on amperometric detection of the resulting HRP-labeled magnetic bioconjugates captured on screen-printed electrodes (SPCEs) in the presence of H2O2 and hydroquinone (HQ), the biotool achieved a low limit of detection (0.5 pM) for the synthetic HPV16 target DNA. In addition, the developed bioplatform was able to discriminate between HPV16 positive and negative human cancer cells using only 25 ng of amplified DNA in a test time of 45 min.
Collapse
Affiliation(s)
- Göksu Ozcelikay
- Ankara University, Faculty of Pharmacy, Department of Analytical Chemistry, 06560 Ankara, Turkey; Department of Analytical Chemistry, Faculty of Chemistry, Complutense University of Madrid, 28040 Madrid, Spain
| | - Maria Gamella
- Department of Analytical Chemistry, Faculty of Chemistry, Complutense University of Madrid, 28040 Madrid, Spain
| | | | - Rodrigo Barderas
- Chronic Disease Programme, UFIEC, Institute of Health Carlos III, Majadahonda, 28220 Madrid, Spain
| | - José M Pingarrón
- Department of Analytical Chemistry, Faculty of Chemistry, Complutense University of Madrid, 28040 Madrid, Spain
| | - Susana Campuzano
- Department of Analytical Chemistry, Faculty of Chemistry, Complutense University of Madrid, 28040 Madrid, Spain.
| | - Sibel A Ozkan
- Ankara University, Faculty of Pharmacy, Department of Analytical Chemistry, 06560 Ankara, Turkey.
| |
Collapse
|