1
|
Vastrad B, Vastrad C. Screening and identification of key biomarkers associated with endometriosis using bioinformatics and next-generation sequencing data analysis. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2024; 25:116. [DOI: 10.1186/s43042-024-00572-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 08/23/2024] [Indexed: 01/04/2025] Open
Abstract
Abstract
Background
Endometriosis is a common cause of endometrial-type mucosa outside the uterine cavity with symptoms such as painful periods, chronic pelvic pain, pain with intercourse and infertility. However, the early diagnosis of endometriosis is still restricted. The purpose of this investigation is to identify and validate the key biomarkers of endometriosis.
Methods
Next-generation sequencing dataset GSE243039 was obtained from the Gene Expression Omnibus database, and differentially expressed genes (DEGs) between endometriosis and normal control samples were identified. After screening of DEGs, gene ontology (GO) and REACTOME pathway enrichment analyses were performed. Furthermore, a protein–protein interaction (PPI) network was constructed and modules were analyzed using the Human Integrated Protein–Protein Interaction rEference database and Cytoscape software, and hub genes were identified. Subsequently, a network between miRNAs and hub genes, and network between TFs and hub genes were constructed using the miRNet and NetworkAnalyst tool, and possible key miRNAs and TFs were predicted. Finally, receiver operating characteristic curve analysis was used to validate the hub genes.
Results
A total of 958 DEGs, including 479 upregulated genes and 479 downregulated genes, were screened between endometriosis and normal control samples. GO and REACTOME pathway enrichment analyses of the 958 DEGs showed that they were mainly involved in multicellular organismal process, developmental process, signaling by GPCR and muscle contraction. Further analysis of the PPI network and modules identified 10 hub genes, including vcam1, snca, prkcb, adrb2, foxq1, mdfi, actbl2, prkd1, dapk1 and actc1. Possible target miRNAs, including hsa-mir-3143 and hsa-mir-2110, and target TFs, including tcf3 (transcription factor 3) and clock (clock circadian regulator), were predicted by constructing a miRNA-hub gene regulatory network and TF-hub gene regulatory network.
Conclusions
This investigation used bioinformatics techniques to explore the potential and novel biomarkers. These biomarkers might provide new ideas and methods for the early diagnosis, treatment and monitoring of endometriosis.
Collapse
|
2
|
Kodiha M, Azad N, Chu S, Crampton N, Stochaj U. Oxidative stress and signaling through EGFR and PKA pathways converge on the nuclear transport factor RanBP1. Eur J Cell Biol 2024; 103:151376. [PMID: 38011756 DOI: 10.1016/j.ejcb.2023.151376] [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: 09/30/2023] [Revised: 11/01/2023] [Accepted: 11/17/2023] [Indexed: 11/29/2023] Open
Abstract
Nuclear protein trafficking requires the soluble transport factor RanBP1. The subcellular distribution of RanBP1 is dynamic, as the protein shuttles between the nucleus and cytoplasm. To date, the signaling pathways regulating RanBP1 subcellular localization are poorly understood. During interphase, RanBP1 resides mostly in the cytoplasm. We show here that oxidative stress concentrates RanBP1 in the nucleus, and our study defines the underlying mechanisms. Specifically, RanBP1's cysteine residues are not essential for its oxidant-induced relocation. Furthermore, our pharmacological approaches uncover that signaling mediated by epidermal growth factor receptor (EGFR) and protein kinase A (PKA) control RanBP1 localization during stress. In particular, pharmacological inhibitors of EGFR or PKA diminish the oxidant-dependent relocation of RanBP1. Mutant analysis identified serine 60 and tyrosine 103 as regulators of RanBP1 nuclear accumulation during oxidant exposure. Taken together, our results define RanBP1 as a target of oxidative stress and a downstream effector of EGFR and PKA signaling routes. This positions RanBP1 at the intersection of important cellular signaling circuits.
Collapse
Affiliation(s)
- Mohamed Kodiha
- Department of Physiology McGill University, Montreal H3G 1Y6, Canada
| | - Nabila Azad
- Department of Physiology McGill University, Montreal H3G 1Y6, Canada
| | - Siwei Chu
- Department of Physiology McGill University, Montreal H3G 1Y6, Canada
| | - Noah Crampton
- Department of Physiology McGill University, Montreal H3G 1Y6, Canada
| | - Ursula Stochaj
- Department of Physiology McGill University, Montreal H3G 1Y6, Canada.
| |
Collapse
|
3
|
Cai X, Li Y, Zheng J, Liu L, Jiao Z, Lin J, Jiang S, Lin X, Sun Y. Modeling of senescence-related chemoresistance in ovarian cancer using data analysis and patient-derived organoids. Front Oncol 2024; 13:1291559. [PMID: 38370348 PMCID: PMC10869451 DOI: 10.3389/fonc.2023.1291559] [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: 09/09/2023] [Accepted: 12/26/2023] [Indexed: 02/20/2024] Open
Abstract
Background Ovarian cancer (OC) is a malignant tumor associated with poor prognosis owing to its susceptibility to chemoresistance. Cellular senescence, an irreversible biological state, is intricately linked to chemoresistance in cancer treatment. We developed a senescence-related gene signature for prognostic prediction and evaluated personalized treatment in patients with OC. Methods We acquired the clinical and RNA-seq data of OC patients from The Cancer Genome Atlas and identified a senescence-related prognostic gene set through differential and cox regression analysis in distinct chemotherapy response groups. A prognostic senescence-related signature was developed and validated by OC patient-derived-organoids (PDOs). We leveraged gene set enrichment analysis (GSEA) and ESTIMATE to unravel the potential functions and immune landscape of the model. Moreover, we explored the correlation between risk scores and potential chemotherapeutic agents. After confirming the congruence between organoids and tumor tissues through immunohistochemistry, we measured the IC50 of cisplatin in PDOs using the ATP activity assay, categorized by resistance and sensitivity to the drug. We also investigated the expression patterns of model genes across different groups. Results We got 2740 differentially expressed genes between two chemotherapy response groups including 43 senescence-related genes. Model prognostic genes were yielded through univariate cox analysis, and multifactorial cox analysis. Our work culminated in a senescence-related prognostic model based on the expression of SGK1 and VEGFA. Simultaneously, we successfully constructed and propagated three OC PDOs for drug screening. PCR and WB from PDOs affirmed consistent expression trends as those of our model genes derived from comprehensive data analysis. Specifically, SGK1 exhibited heightened expression in cisplatin-resistant OC organoids, while VEGFA manifested elevated expression in the sensitive group (P<0.05). Intriguingly, GSEA results unveiled the enrichment of model genes in the PPAR signaling pathway, pivotal regulator in chemoresistance and tumorigenesis. This revelation prompted the identification of potential beneficial drugs for patients with a high-risk score, including gemcitabine, dabrafenib, epirubicin, oxaliplatin, olaparib, teniposide, ribociclib, topotecan, venetoclax. Conclusion Through the formulation of a senescence-related signature comprising SGK1 and VEGFA, we established a promising tool for prognosticating chemotherapy reactions, predicting outcomes, and steering therapeutic strategies. Patients with high VEGFA and low SGK1 expression levels exhibit heightened sensitivity to chemotherapy.
Collapse
Affiliation(s)
- Xintong Cai
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Yanhong Li
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Jianfeng Zheng
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Li Liu
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Zicong Jiao
- Department of Translational Medicine, Scientific Research System, Geneplus -Beijing Institute, Beijing, China
| | - Jie Lin
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Shan Jiang
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Xuefen Lin
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Yang Sun
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| |
Collapse
|
4
|
Lin X, Ma Q, Chen L, Guo W, Huang Z, Huang T, Cai YD. Identifying genes associated with resistance to KRAS G12C inhibitors via machine learning methods. Biochim Biophys Acta Gen Subj 2023; 1867:130484. [PMID: 37805078 DOI: 10.1016/j.bbagen.2023.130484] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 10/02/2023] [Accepted: 10/04/2023] [Indexed: 10/09/2023]
Abstract
BACKGROUND Targeted therapy has revolutionized cancer treatment, greatly improving patient outcomes and quality of life. Lung cancer, specifically non-small cell lung cancer, is frequently driven by the G12C mutation at the KRAS locus. The development of KRAS inhibitors has been a breakthrough in the field of cancer research, given the crucial role of KRAS mutations in driving tumor growth and progression. However, over half of patients with cancer bypass inhibition show limited response to treatment. The mechanisms underlying tumor cell resistance to this treatment remain poorly understood. METHODS To address above gap in knowledge, we conducted a study aimed to elucidate the differences between tumor cells that respond positively to KRAS (G12C) inhibitor therapy and those that do not. Specifically, we analyzed single-cell gene expression profiles from KRAS G12C-mutant tumor cell models (H358, H2122, and SW1573) treated with KRAS G12C (ARS-1620) inhibitor, which contained 4297 cells that continued to proliferate under treatment and 3315 cells that became quiescent. Each cell was represented by the expression levels on 8687 genes. We then designed an innovative machine learning based framework, incorporating seven feature ranking algorithms and four classification algorithms to identify essential genes and establish quantitative rules. RESULTS Our analysis identified some top-ranked genes, including H2AFZ, CKS1B, TUBA1B, RRM2, and BIRC5, that are known to be associated with the progression of multiple cancers. CONCLUSION Above genes were relevant to tumor cell resistance to targeted therapy. This study provides important insights into the molecular mechanisms underlying tumor cell resistance to KRAS inhibitor treatment.
Collapse
Affiliation(s)
- Xiandong Lin
- Laboratory of Radiation Oncology and Radiobiology, Clinical Oncology School of Fujian Medical University and Fujian Cancer Hospital, Fuzhou 350014, China.
| | - QingLan Ma
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Lei Chen
- College of Information Engineering, Shanghai Maritime University, Shanghai 201306, China
| | - Wei Guo
- Key Laboratory of Stem Cell Biology, Shanghai Jiao Tong University School of Medicine (SJTUSM) & Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai 200030, China
| | - Zhiyi Huang
- College of Chemistry, Fuzhou University, Fuzhou 350000, China
| | - Tao Huang
- Bio-Med Big Data Center, CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China; CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Yu-Dong Cai
- School of Life Sciences, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
5
|
Bian X, Xue H, Jing D, Wang Y, Zhou G, Zhu F. Role of Serum/Glucocorticoid-Regulated Kinase 1 (SGK1) in Immune and Inflammatory Diseases. Inflammation 2023; 46:1612-1625. [PMID: 37353719 DOI: 10.1007/s10753-023-01857-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 06/09/2023] [Accepted: 06/12/2023] [Indexed: 06/25/2023]
Abstract
Serum/glucocorticoid-regulated kinase 1 (SGK1), a member of the serine/threonine protein kinase gene family, is primarily regulated by serum and glucocorticoids. SGK1 is involved in the development of tumors and fibrotic diseases. However, relatively little research has been conducted on their role in immune and inflammatory diseases. SGK1 may act as a pivotal immune regulatory gene by modulating immune cells (e.g., T cells, macrophages, dendritic cells, and neutrophils) and functions and is involved in the pathogenesis of some immune and inflammatory diseases, such as inflammatory bowel disease, multiple sclerosis, allergic diseases, sepsis, and major depressive disorder. This review aims to provide an overview of the latest research focusing on the immune and inflammatory regulatory roles of SGK1 and provide new insights into diagnostic and therapeutic approaches for immune and inflammatory diseases.
Collapse
Affiliation(s)
- Xixi Bian
- Clinical Medical College of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Honglu Xue
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Dehuai Jing
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Yan Wang
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Guangxi Zhou
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China.
| | - Fengqin Zhu
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China.
| |
Collapse
|
6
|
Buonaiuto R, Neola G, Cecere SC, Caltavituro A, Cefaliello A, Pietroluongo E, De Placido P, Giuliano M, Arpino G, De Angelis C. Glucocorticoid Receptor and Ovarian Cancer: From Biology to Therapeutic Intervention. Biomolecules 2023; 13:biom13040653. [PMID: 37189400 DOI: 10.3390/biom13040653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 03/27/2023] [Accepted: 03/31/2023] [Indexed: 04/08/2023] Open
Abstract
Ovarian cancer (OC) is the leading cause of death from gynecological malignancies worldwide. Fortunately, recent advances in OC biology and the discovery of novel therapeutic targets have led to the development of novel therapeutic agents that may improve the outcome of OC patients. The glucocorticoid receptor (GR) is a ligand-dependent transcriptional factor known for its role in body stress reactions, energy homeostasis and immune regulation. Notably, evidence suggests that GR may play a relevant role in tumor progression and may affect treatment response. In cell culture models, administration of low levels of glucocorticoids (GCs) suppresses OC growth and metastasis. Conversely, high GR expression has been associated with poor prognostic features and long-term outcomes in patients with OC. Moreover, both preclinical and clinical data have shown that GR activation impairs the effectiveness of chemotherapy by inducing the apoptotic pathways and cell differentiation. In this narrative review, we summarize data related to the function and role of GR in OC. To this aim, we reorganized the controversial and fragmented data regarding GR activity in OC and herein describe its potential use as a prognostic and predictive biomarker. Moreover, we explored the interplay between GR and BRCA expression and reviewed the latest therapeutic strategies such as non-selective GR antagonists and selective GR modulators to enhance chemotherapy sensitivity, and to finally provide new treatment options in OC patients.
Collapse
Affiliation(s)
- Roberto Buonaiuto
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Giuseppe Neola
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Sabrina Chiara Cecere
- Oncologia Clinica Sperimentale Uro-Ginecologica, Istituto Nazionale Tumori IRCCS Fondazione G Pascale, 80131 Naples, Italy
| | - Aldo Caltavituro
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Amedeo Cefaliello
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Erica Pietroluongo
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Pietro De Placido
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Mario Giuliano
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Grazia Arpino
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Carmine De Angelis
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| |
Collapse
|
7
|
D’Antona L, Amato R, Brescia C, Rocca V, Colao E, Iuliano R, Blazer-Yost BL, Perrotti N. Kinase Inhibitors in Genetic Diseases. Int J Mol Sci 2023; 24:ijms24065276. [PMID: 36982349 PMCID: PMC10048847 DOI: 10.3390/ijms24065276] [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: 01/13/2023] [Revised: 03/02/2023] [Accepted: 03/06/2023] [Indexed: 03/12/2023] Open
Abstract
Over the years, several studies have shown that kinase-regulated signaling pathways are involved in the development of rare genetic diseases. The study of the mechanisms underlying the onset of these diseases has opened a possible way for the development of targeted therapies using particular kinase inhibitors. Some of these are currently used to treat other diseases, such as cancer. This review aims to describe the possibilities of using kinase inhibitors in genetic pathologies such as tuberous sclerosis, RASopathies, and ciliopathies, describing the various pathways involved and the possible targets already identified or currently under study.
Collapse
Affiliation(s)
- Lucia D’Antona
- Department of Health Sciences, University “Magna Graecia” at Catanzaro, 88100 Catanzaro, Italy
- Medical Genetics Unit, University Hospital “Mater Domini” at Catanzaro, 88100 Catanzaro, Italy
| | - Rosario Amato
- Department of Health Sciences, University “Magna Graecia” at Catanzaro, 88100 Catanzaro, Italy
- Medical Genetics Unit, University Hospital “Mater Domini” at Catanzaro, 88100 Catanzaro, Italy
| | - Carolina Brescia
- Department of Health Sciences, University “Magna Graecia” at Catanzaro, 88100 Catanzaro, Italy
| | - Valentina Rocca
- Medical Genetics Unit, University Hospital “Mater Domini” at Catanzaro, 88100 Catanzaro, Italy
- Department of Experimental and Clinical Medicine, University “Magna Graecia” at Catanzaro, 88100 Catanzaro, Italy
| | - Emma Colao
- Medical Genetics Unit, University Hospital “Mater Domini” at Catanzaro, 88100 Catanzaro, Italy
| | - Rodolfo Iuliano
- Department of Health Sciences, University “Magna Graecia” at Catanzaro, 88100 Catanzaro, Italy
- Medical Genetics Unit, University Hospital “Mater Domini” at Catanzaro, 88100 Catanzaro, Italy
| | - Bonnie L. Blazer-Yost
- Department of Biology, Indiana University Purdue University, Indianapolis, IN 46202, USA
| | - Nicola Perrotti
- Department of Health Sciences, University “Magna Graecia” at Catanzaro, 88100 Catanzaro, Italy
- Medical Genetics Unit, University Hospital “Mater Domini” at Catanzaro, 88100 Catanzaro, Italy
- Correspondence:
| |
Collapse
|
8
|
Targeting Breast Cancer: An Overlook on Current Strategies. Int J Mol Sci 2023; 24:ijms24043643. [PMID: 36835056 PMCID: PMC9959993 DOI: 10.3390/ijms24043643] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/08/2023] [Accepted: 02/09/2023] [Indexed: 02/16/2023] Open
Abstract
Breast cancer (BC) is one of the most widely diagnosed cancers and a leading cause of cancer death among women worldwide. Globally, BC is the second most frequent cancer and first most frequent gynecological one, affecting women with a relatively low case-mortality rate. Surgery, radiotherapy, and chemotherapy are the main treatments for BC, even though the latter are often not aways successful because of the common side effects and the damage caused to healthy tissues and organs. Aggressive and metastatic BCs are difficult to treat, thus new studies are needed in order to find new therapies and strategies for managing these diseases. In this review, we intend to give an overview of studies in this field, presenting the data from the literature concerning the classification of BCs and the drugs used in therapy for the treatment of BCs, along with drugs in clinical studies.
Collapse
|
9
|
Audia S, Brescia C, Dattilo V, D’Antona L, Calvano P, Iuliano R, Trapasso F, Perrotti N, Amato R. RANBP1 (RAN Binding Protein 1): The Missing Genetic Piece in Cancer Pathophysiology and Other Complex Diseases. Cancers (Basel) 2023; 15:cancers15020486. [PMID: 36672435 PMCID: PMC9857238 DOI: 10.3390/cancers15020486] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/29/2022] [Accepted: 01/11/2023] [Indexed: 01/15/2023] Open
Abstract
RANBP1 encoded by RANBP1 or HTF9A (Hpall Tiny Fragments Locus 9A), plays regulatory functions of the RAN-network, belonging to the RAS superfamily of small GTPases. Through this function, RANBP1 regulates the RANGAP1 activity and, thus, the fluctuations between GTP-RAN and GDP-RAN. In the light of this, RANBP1 take actions in maintaining the nucleus-cytoplasmic gradient, thus making nuclear import-export functional. RANBP1 has been implicated in the inter-nuclear transport of proteins, nucleic acids and microRNAs, fully contributing to cellular epigenomic signature. Recently, a RANBP1 diriment role in spindle checkpoint formation and nucleation has emerged, thus constituting an essential element in the control of mitotic stability. Over time, RANBP1 has been demonstrated to be variously involved in human cancers both for the role in controlling nuclear transport and RAN activity and for its ability to determine the efficiency of the mitotic process. RANBP1 also appears to be implicated in chemo-hormone and radio-resistance. A key role of this small-GTPases related protein has also been demonstrated in alterations of axonal flow and neuronal plasticity, as well as in viral and bacterial metabolism and in embryological maturation. In conclusion, RANBP1 appears not only to be an interesting factor in several pathological conditions but also a putative target of clinical interest.
Collapse
Affiliation(s)
- Salvatore Audia
- Dipartimento di Scienze della Salute, Campus Salvatore Venuta, Università degli Studi “Magna Graecia” di Catanzaro, Viale Europa, 88100 Catanzaro, Italy
| | - Carolina Brescia
- Dipartimento di Scienze della Salute, Campus Salvatore Venuta, Università degli Studi “Magna Graecia” di Catanzaro, Viale Europa, 88100 Catanzaro, Italy
| | - Vincenzo Dattilo
- Dipartimento di Medicina Sperimentale e Clinica, Campus Salvatore Venuta, Università degli Studi “Magna Graecia” di Catanzaro, Viale Europa, 88100 Catanzaro, Italy
| | - Lucia D’Antona
- Dipartimento di Scienze della Salute, Campus Salvatore Venuta, Università degli Studi “Magna Graecia” di Catanzaro, Viale Europa, 88100 Catanzaro, Italy
| | - Pierluigi Calvano
- Dipartimento di Scienze della Salute, Campus Salvatore Venuta, Università degli Studi “Magna Graecia” di Catanzaro, Viale Europa, 88100 Catanzaro, Italy
| | - Rodolfo Iuliano
- Dipartimento di Scienze della Salute, Campus Salvatore Venuta, Università degli Studi “Magna Graecia” di Catanzaro, Viale Europa, 88100 Catanzaro, Italy
| | - Francesco Trapasso
- Dipartimento di Medicina Sperimentale e Clinica, Campus Salvatore Venuta, Università degli Studi “Magna Graecia” di Catanzaro, Viale Europa, 88100 Catanzaro, Italy
| | - Nicola Perrotti
- Dipartimento di Scienze della Salute, Campus Salvatore Venuta, Università degli Studi “Magna Graecia” di Catanzaro, Viale Europa, 88100 Catanzaro, Italy
| | - Rosario Amato
- Dipartimento di Scienze della Salute, Campus Salvatore Venuta, Università degli Studi “Magna Graecia” di Catanzaro, Viale Europa, 88100 Catanzaro, Italy
- Correspondence: ; Tel.: +39-0961-3694084
| |
Collapse
|
10
|
Hao Y, Li Z, Chang M, Zhang X. Effects of Salidroside Combined with Paclitaxel on Proliferation, Migration, and Epithelial Mesenchyme of Colorectal Cancer Cells. Drug Des Devel Ther 2022; 16:4079-4089. [PMID: 36465266 PMCID: PMC9716943 DOI: 10.2147/dddt.s384151] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 11/16/2022] [Indexed: 02/04/2024] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is a multifactorial disease and one of the most common malignancies worldwide. Salidroside (Sal) is a plant with a wide range of pharmacological effects and plays an important role in the treatment of many diseases, and is considered a new hope for the treatment of tumors. The purpose of this study was to investigate the effect of the combination of Sal and paclitaxel (Pac) on colorectal cancer cells and its mechanism of action. METHODS The effects of different mass concentrations of Sal, Pac, and the combination intervened in the cells for 48 h were examined using the CCK8 method. The inhibition rate was obtained, and the optimal concentration of the respective drug group was screened. The proliferative capacity of the respective group was obtained. Subsequently, the results of apoptosis, cloning, migration, invasion, and angiogenesis were observed through cell morphological analysis (shape observation and Hoechst staining), colony formation assay, cell scratching assay, Transwell, angiogenesis assay, and protein immunoblotting (Western blotting) to detect the expression of epithelial-mesenchymal transition (EMT)-associated proteins and PI3K pathway-associated proteins. RESULTS Different concentrations of Sal, Pac, and the combined application had significant effects in inhibiting cells in a concentration-dependent manner. Compared with the control group, the Sal group, the Pac group, and the combination group significantly inhibited the clonal number, migration, invasion, and tube-forming ability of colorectal cancer cells. Besides, the combined application had a better effect than the Sal and Pac groups. The apoptosis level was up-regulated in all drug groups, and the up-regulation was more significant in the combination group. The expression of E-cad protein was up-regulated, the expression of N-cad and Vim protein was down-regulated, and the expression of PI3K and AKT phosphorylation was down-regulated in the respective group, and the difference was more significant in the combination group compared with the group of individual drugs. CONCLUSION The combined application of Sal and Pac significantly can decrease the survival rate of colorectal cancer cells, and the mechanism may be correlated with the blocking of the PI3K/AKT pathway, thus inhibiting EMT.
Collapse
Affiliation(s)
- Yanjiao Hao
- Department of Life Science Research Center, College of Basic Medicine, Hebei North University, Zhangjiakou, Hebei, 075000, People’s Republic of China
| | - Zhiyu Li
- Department of Life Science Research Center, College of Basic Medicine, Hebei North University, Zhangjiakou, Hebei, 075000, People’s Republic of China
| | - Mingzhi Chang
- Department of Life Science Research Center, College of Basic Medicine, Hebei North University, Zhangjiakou, Hebei, 075000, People’s Republic of China
| | - Xiaoli Zhang
- Department of Life Science Research Center, College of Basic Medicine, Hebei North University, Zhangjiakou, Hebei, 075000, People’s Republic of China
| |
Collapse
|
11
|
Lucia Ruiz Benitez M, Severo Sabedra Sousa F, Peter Furtado I, Carlos Rodrigues Junior J, Victoria Mascarenhas Borba M, Vieira Segatto N, Tabarelli G, Klein Couto G, Júlia Damé Fonseca Paschoal M, Silveira Pacheco B, E. D. Rodrigues O, Collares T, Kömmling Seixas F. Chiral β‐arylchalcogenium azide induce apoptosis and regulate Oxidative Damage on Human Bladder Cancer Cells. ChemistrySelect 2022. [DOI: 10.1002/slct.202203207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Martha Lucia Ruiz Benitez
- Laboratory of Cancer Biotechnology, Technology Development Center Federal University of Pelotas Pelotas Rio Grande do Sul Brazil
- School of Basic and Biomedical Sciences Universidad Simón Bolívar Barranquilla Colombia
| | - Fernanda Severo Sabedra Sousa
- Laboratory of Cancer Biotechnology, Technology Development Center Federal University of Pelotas Pelotas Rio Grande do Sul Brazil
| | - Izadora Peter Furtado
- Laboratory of Cancer Biotechnology, Technology Development Center Federal University of Pelotas Pelotas Rio Grande do Sul Brazil
| | - João Carlos Rodrigues Junior
- Laboratory of Cancer Biotechnology, Technology Development Center Federal University of Pelotas Pelotas Rio Grande do Sul Brazil
| | - Msc. Victoria Mascarenhas Borba
- Laboratory of Cancer Biotechnology, Technology Development Center Federal University of Pelotas Pelotas Rio Grande do Sul Brazil
| | - Natália Vieira Segatto
- Laboratory of Cancer Biotechnology, Technology Development Center Federal University of Pelotas Pelotas Rio Grande do Sul Brazil
| | - Greice Tabarelli
- LabSelen-NanoBio - Chemistry Department Federal University of Santa Maria, Santa Maria Rio Grande do Sul Brazil
| | - Gabriela Klein Couto
- Laboratory of Cancer Biotechnology, Technology Development Center Federal University of Pelotas Pelotas Rio Grande do Sul Brazil
| | - Msc. Júlia Damé Fonseca Paschoal
- Laboratory of Cancer Biotechnology, Technology Development Center Federal University of Pelotas Pelotas Rio Grande do Sul Brazil
| | - Bruna Silveira Pacheco
- Laboratory of Cancer Biotechnology, Technology Development Center Federal University of Pelotas Pelotas Rio Grande do Sul Brazil
| | - Oscar E. D. Rodrigues
- LabSelen-NanoBio - Chemistry Department Federal University of Santa Maria, Santa Maria Rio Grande do Sul Brazil
| | - Tiago Collares
- Laboratory of Cancer Biotechnology, Technology Development Center Federal University of Pelotas Pelotas Rio Grande do Sul Brazil
| | - Fabiana Kömmling Seixas
- Laboratory of Cancer Biotechnology, Technology Development Center Federal University of Pelotas Pelotas Rio Grande do Sul Brazil
| |
Collapse
|
12
|
James NE, Woodman M, De La Cruz P, Eurich K, Ozsoy MA, Schorl C, Hanley LC, Ribeiro JR. Adaptive transcriptomic and immune infiltrate responses in the tumor immune microenvironment following neoadjuvant chemotherapy in high grade serous ovarian cancer reveal novel prognostic associations and activation of pro-tumorigenic pathways. Front Immunol 2022; 13:965331. [PMID: 36131935 PMCID: PMC9483165 DOI: 10.3389/fimmu.2022.965331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 08/15/2022] [Indexed: 11/13/2022] Open
Abstract
The high rate of ovarian cancer recurrence and chemoresistance necessitates further research into how chemotherapy affects the tumor immune microenvironment (TIME). While studies have shown that immune infiltrate increases following neoadjuvant (NACT) chemotherapy, there lacks a comprehensive understanding of chemotherapy-induced effects on immunotranscriptomics and cancer-related pathways and their relationship with immune infiltrate and patient responses. In this study, we performed NanoString nCounter® PanCancer IO360 analysis of 31 high grade serous ovarian cancer (HGSOC) patients with matched pre-treatment biopsy and post-NACT tumor. We observed increases in pro-tumorigenic and immunoregulatory pathways and immune infiltrate following NACT, with striking increases in a cohort of genes centered on the transcription factors ATF3 and EGR1. Using quantitative PCR, we analyzed several of the top upregulated genes in HGSOC cell lines, noting that two of them, ATF3 and AREG, were consistently upregulated with chemotherapy exposure and significantly increased in platinum resistant cells compared to their sensitive counterparts. Furthermore, we observed that pre-NACT immune infiltrate and pathway scores were not strikingly related to platinum free interval (PFI), but post-NACT immune infiltrate, pathway scores, and gene expression were. Finally, we found that higher levels of a cohort of proliferative and DNA damage-related genes was related to shorter PFI. This study underscores the complex alterations in the ovarian TIME following chemotherapy exposure and begins to untangle how immunologic factors are involved in mediating chemotherapy response, which will allow for the future development of novel immunologic therapies to combat chemoresistance.
Collapse
Affiliation(s)
- Nicole E. James
- Department of Obstetrics and Gynecology, Program in Women’s Oncology, Women and Infants Hospital, Providence, RI, United States
- Department of Obstetrics and Gynecology, Warren-Alpert Medical School of Brown University, Providence, RI, United States
- *Correspondence: Nicole E. James,
| | - Morgan Woodman
- Department of Obstetrics and Gynecology, Program in Women’s Oncology, Women and Infants Hospital, Providence, RI, United States
| | - Payton De La Cruz
- Pathobiology Graduate Program, Brown University, Providence, RI, United States
| | - Katrin Eurich
- Department of Obstetrics and Gynecology, Program in Women’s Oncology, Women and Infants Hospital, Providence, RI, United States
- Department of Obstetrics and Gynecology, Warren-Alpert Medical School of Brown University, Providence, RI, United States
| | - Melih Arda Ozsoy
- Department of Biochemistry, Brown University, Providence, RI, United States
| | - Christoph Schorl
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI, United States
| | - Linda C. Hanley
- Department of Pathology, Women and Infants Hospital, Providence, RI, United States
| | - Jennifer R. Ribeiro
- Department of Obstetrics and Gynecology, Program in Women’s Oncology, Women and Infants Hospital, Providence, RI, United States
- Department of Obstetrics and Gynecology, Warren-Alpert Medical School of Brown University, Providence, RI, United States
| |
Collapse
|
13
|
Th17-Gene Expression Profile in Patients with Chronic Venous Disease and Venous Ulcers: Genetic Modulations and Preliminary Clinical Evidence. Biomolecules 2022; 12:biom12070902. [PMID: 35883458 PMCID: PMC9312858 DOI: 10.3390/biom12070902] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/20/2022] [Accepted: 06/25/2022] [Indexed: 11/22/2022] Open
Abstract
Chronic venous disease is a condition globally widespread, resulting in a disabling pathological disorder. The CD4 + Th17+ (Cluster Differentiation 4) lymphocytes represent a regulative factor for innate immunity related to the development of complex diseases. Recently, these mechanisms have been associated with vascular disease. The aim of this work is to validate whether the Th17 response correlates with the development of CVI (Chronic venous insufficiency)and CVLUs (chronic venous limbs ulcers) and whether Th17 markers can be used, both as intrinsic risk factors and diagnostic markers, for disease development. PBL derived from peripheral blood samples of patients and controls were subjected to gene expression analysis for IL23R, IL17, SGK1, TGFβ, RORγ, FOXO1, and RANBP1 by qRT-PCR and immunoblot. A post hoc correlation, the diagnostic performance of the target genes, and multivariable analyses were properly conducted. The main expression markers of the CD4 + Th17+ switch were strongly activated in chronic venous insufficiency and in advanced ulceration. The correlation analysis demonstrated the inter-dependence on Th17’s signature modulation. ROC (Receiver Operating Characteristic) analysis defined, for the examined genes, a clinical value as the potential diagnostic markers. Multi-logistic regression studies showed that Th17 markers behave as empirical risk factors for CVD (chronic venous disease) development. Taken together, the present data provide a new hypothesis for the TH17-dependent pathogenesis of CVD, favoring the possibility for the development of new diagnostic, preventive, and therapeutic approaches.
Collapse
|
14
|
Cicenas J, Meskinyte-Kausiliene E, Jukna V, Rimkus A, Simkus J, Soderholm D. SGK1 in Cancer: Biomarker and Drug Target. Cancers (Basel) 2022; 14:2385. [PMID: 35625991 PMCID: PMC9139822 DOI: 10.3390/cancers14102385] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/06/2022] [Accepted: 05/09/2022] [Indexed: 12/29/2022] Open
Abstract
Serum- and glucocorticoid-regulated kinases (SGKs) are members of the AGC family of serine/threonine kinases, consisting of three isoforms: SGK1, SGK2, and SGK3. SGK1 was initially cloned as a gene transcriptionally stimulated by serum and glucocorticoids in rat mammary tumor cells. It is upregulated in some cancers and downregulated in others. SGK1 increases tumor cell survival, adhesiveness, invasiveness, motility, and epithelial to mesenchymal transition. It stimulates tumor growth by mechanisms such as activation of K+ channels and Ca2+ channels, Na+/H+ exchanger, amino acid and glucose transporters, downregulation of Foxo3a and p53, and upregulation of β-catenin and NFκB. This chapter focuses on major aspects of SGK1 involvement in cancer, its use as biomarker as well as potential therapeutic target.
Collapse
Affiliation(s)
- Jonas Cicenas
- Proteomics Centre, Institute of Biochemistry, Vilnius University, Sauletekio al. 7, LT-10257 Vilnius, Lithuania
- MAP Kinase Resource, Bioinformatics, Melchiorstrasse 9, CH-3027 Bern, Switzerland; (A.R.); (J.S.)
- Center of Animal Husbandry Selections, Breeding Values and Dissemination, Agriculture Academy, Vytautas Magnus University, Studentų g. 11, LT-53361 Akademija, Lithuania; (E.M.-K.); (V.J.)
| | - Edita Meskinyte-Kausiliene
- Center of Animal Husbandry Selections, Breeding Values and Dissemination, Agriculture Academy, Vytautas Magnus University, Studentų g. 11, LT-53361 Akademija, Lithuania; (E.M.-K.); (V.J.)
| | - Vigilijus Jukna
- Center of Animal Husbandry Selections, Breeding Values and Dissemination, Agriculture Academy, Vytautas Magnus University, Studentų g. 11, LT-53361 Akademija, Lithuania; (E.M.-K.); (V.J.)
| | - Arnas Rimkus
- MAP Kinase Resource, Bioinformatics, Melchiorstrasse 9, CH-3027 Bern, Switzerland; (A.R.); (J.S.)
| | - Jokubas Simkus
- MAP Kinase Resource, Bioinformatics, Melchiorstrasse 9, CH-3027 Bern, Switzerland; (A.R.); (J.S.)
| | - Diana Soderholm
- Walker Art Center, 752 Vineland PI, Mineapolis, MN 55403, USA;
| |
Collapse
|
15
|
Abstract
BACKGROUND The serum and glucocorticoid-induced kinase-1 (SGK1) belonging to the AGC protein kinase family phosphorylates serine and threonine residues of target proteins. It regulates numerous ion channels and transporters and promotes survival under cellular stress. Unique to SGK1 is a tight control at transcriptional and post-transcriptional levels. SGK1 regulates multiple signal transduction pathways related to tumor development. Several studies have reported that SGK1 is upregulated in different types of human malignancies and induces resistance against inhibitors, drugs, and targeted therapies. RESULTS AND CONCLUSION This review highlights the cellular functions of SGK1, its crucial role in cancer development, and clinical insights for SGK1 targeted therapies. Furthermore, the role of SGK1-mediated autophagy as a potential therapeutic target for cancer has been discussed.
Collapse
|
16
|
Greenstein AE, Hunt HJ. Glucocorticoid receptor antagonism promotes apoptosis in solid tumor cells. Oncotarget 2021; 12:1243-1255. [PMID: 34194622 PMCID: PMC8238250 DOI: 10.18632/oncotarget.27989] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 06/02/2021] [Indexed: 11/25/2022] Open
Abstract
Background: Resistance to antiproliferative chemotherapies remains a significant challenge in the care of patients with solid tumors. Glucocorticoids, including endogenous cortisol, have been shown to induce pro-survival pathways in epithelial tumor cells. While pro-apoptotic effects of glucocorticoid receptor (GR) antagonism have been demonstrated under select conditions, the breadth and nature of these effects have not been fully established. Materials and Methods: To guide studies in cancer patients, relacorilant, an investigational selective GR modulator (SGRM) that antagonizes cortisol activity, was assessed in various tumor types, with multiple cytotoxic combination partners, and in the presence of physiological cortisol concentrations. Results: In the MIA PaCa-2 cell line, paclitaxel-driven apoptosis was blunted by cortisol and restored by relacorilant. In the OVCAR5 cell line, relacorilant improved the efficacy of paclitaxel and the potency of platinum agents. A screen to identify optimal combination partners for relacorilant showed that microtubule-targeted agents consistently benefited from combination with relacorilant. These findings were confirmed in xenograft models, including MIA PaCa-2, HeLa, and a cholangiocarcinoma patient-derived xenograft. In vivo, tumor-cell apoptosis was increased when relacorilant was added to paclitaxel in multiple models. Conclusions: These observations support recently reported findings of clinical benefit when relacorilant is added to paclitaxel-containing therapy in patients with ovarian and pancreatic cancers and provide a new rationale for combining relacorilant with additional cytotoxic agents.
Collapse
|
17
|
Rango E, D'Antona L, Iovenitti G, Brai A, Mancini A, Zamperini C, Trivisani CI, Marianelli S, Fallacara AL, Molinari A, Cianciusi A, Schenone S, Perrotti N, Dreassi E, Botta M. Si113-prodrugs selectively activated by plasmin against hepatocellular and ovarian carcinoma. Eur J Med Chem 2021; 223:113653. [PMID: 34161866 DOI: 10.1016/j.ejmech.2021.113653] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 06/01/2021] [Accepted: 06/13/2021] [Indexed: 11/25/2022]
Abstract
Si113, a pyrazolo[3,4-d]pyrimidine derivative, gained more attention as an anticancer agent due to its potent anticancer activity on both in vitro and in vivo hepatocellular carcinomas (HCC) and ovarian carcinoma models. But the drawback is the low water solubility which prevents its further development. In this context, we successfully overcame this limitation by synthesizing two novel prodrugs introducing the amino acid sequence D-Ala-Leu-Lys (TP). Moreover, TP sequence has a high affinity with plasmin, a protease recognized as overexpressed in many solid cancers, including HCC and ovarian carcinoma. The prodrugs were synthesized and fully characterized in terms of in vitro ADME properties, plasma stability and plasmin-induced release of the parent drug. The inhibitory activity against Sgk1 was evaluated and in vitro growth inhibition was evaluated on ovarian carcinoma and HCC cell lines in the presence and absence of human plasmin. In vivo pharmacokinetic properties and preliminary tissue distribution confirmed a better profile highlighting the importance of the prodrug approach. Finally, the prodrug antitumor efficacy was evaluated in an HCC xenografted murine model, where a significant reduction (around 90%) in tumor growth was observed. Treatment with ProSi113-TP in combination with paclitaxel in a paclitaxel-resistant ovarian carcinoma xenografted murine model, resulted in an impressive reduction of tumor volume greater than 95%. Our results revealed a promising activity of Si113 prodrugs and pave the way for their further development against resistant cancer.
Collapse
Affiliation(s)
- Enrico Rango
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via Aldo Moro 2, 53100, Siena, Italy
| | - Lucia D'Antona
- Dipartimento di Scienze della Salute, Università"Magna Graecia" di Catanzaro, Viale Europa, 88100, Catanzaro, Italy
| | - Giulia Iovenitti
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via Aldo Moro 2, 53100, Siena, Italy
| | - Annalaura Brai
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via Aldo Moro 2, 53100, Siena, Italy
| | - Arianna Mancini
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via Aldo Moro 2, 53100, Siena, Italy
| | - Claudio Zamperini
- Lead Discovery Siena S.r.l., Via Vittorio Alfieri 31, 53019, Castelnuovo Berardenga, Siena, Italy
| | - Claudia Immacolata Trivisani
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via Aldo Moro 2, 53100, Siena, Italy
| | - Stefano Marianelli
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via Aldo Moro 2, 53100, Siena, Italy
| | - Anna Lucia Fallacara
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via Aldo Moro 2, 53100, Siena, Italy
| | - Alessio Molinari
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via Aldo Moro 2, 53100, Siena, Italy
| | - Annarita Cianciusi
- Dipartimento di Farmacia, Università degli Studi di Genova, Viale Benedetto XV 3, Genoa, 16132, Italy
| | - Silvia Schenone
- Dipartimento di Farmacia, Università degli Studi di Genova, Viale Benedetto XV 3, Genoa, 16132, Italy
| | - Nicola Perrotti
- Dipartimento di Scienze della Salute, Università"Magna Graecia" di Catanzaro, Viale Europa, 88100, Catanzaro, Italy.
| | - Elena Dreassi
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via Aldo Moro 2, 53100, Siena, Italy.
| | - Maurizio Botta
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via Aldo Moro 2, 53100, Siena, Italy; Lead Discovery Siena S.r.l., Via Vittorio Alfieri 31, 53019, Castelnuovo Berardenga, Siena, Italy; Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology Temple University, BioLife Science Building, Suite 333, 1900 North 12th Street, Philadelphia, PA, 19122, United States
| |
Collapse
|
18
|
Pyrazole[3,4-d]pyrimidine derivatives loaded into halloysite as potential CDK inhibitors. Int J Pharm 2021; 599:120281. [PMID: 33524522 DOI: 10.1016/j.ijpharm.2021.120281] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 01/11/2021] [Accepted: 01/12/2021] [Indexed: 12/15/2022]
Abstract
Uncontrolled cell proliferation is a hallmark of cancer as a result of rapid and deregulated progression through the cell cycle. The inhibition of cyclin-dependent kinases (CDKs) activities is a promising therapeutic strategy to block cell cycle of tumor cells. In this work we reported a new example of nanocomposites based on halloysite nanotubes (HNTs)/pyrazolo[3,4-d]pyrimidine derivatives (Si306 and Si113) as anticancer agents and CDK inhibitors. HNTs/Si306 and HNTs/Si113 nanocomposites were synthesized and characterized. The release kinetics were also investigated. Antitumoral activity was evaluated on three cancer cell lines (HeLa, MDA-MB-231 and HCT116) and the effects on cell cycle arrest in HCT116 cells were evaluated. Finally, molecular dynamics simulations were performed of the complexes between Si113 or Si306 and the active site of both CDK 1 and 2.
Collapse
|
19
|
Sang Y, Kong P, Zhang S, Zhang L, Cao Y, Duan X, Sun T, Tao Z, Liu W. SGK1 in Human Cancer: Emerging Roles and Mechanisms. Front Oncol 2021; 10:608722. [PMID: 33542904 PMCID: PMC7851074 DOI: 10.3389/fonc.2020.608722] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 11/24/2020] [Indexed: 12/12/2022] Open
Abstract
Serum and glucocorticoid-induced protein kinase 1 (SGK1) is a member of the "AGC" subfamily of protein kinases, which shares structural and functional similarities with the AKT family of kinases and displays serine/threonine kinase activity. Aberrant expression of SGK1 has profound cellular consequences and is closely correlated with human cancer. SGK1 is considered a canonical factor affecting the expression and signal transduction of multiple genes involved in the genesis and development of many human cancers. Abnormal expression of SGK1 has been found in tissue and may hopefully become a useful indicator of cancer progression. In addition, SGK1 acts as a prognostic factor for cancer patient survival. This review systematically summarizes and discusses the role of SGK1 as a diagnostic and prognostic biomarker of diverse cancer types; focuses on its essential roles and functions in tumorigenesis, cancer cell proliferation, apoptosis, invasion, metastasis, autophagy, metabolism, and therapy resistance and in the tumor microenvironment; and finally summarizes the current understanding of the regulatory mechanisms of SGK1 at the molecular level. Taken together, this evidence highlights the crucial role of SGK1 in tumorigenesis and cancer progression, revealing why it has emerged as a potential target for cancer therapy.
Collapse
Affiliation(s)
- Yiwen Sang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Piaoping Kong
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Shizhen Zhang
- The Cancer Institute of the Second Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Lingyu Zhang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Ying Cao
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xiuzhi Duan
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Tao Sun
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Zhihua Tao
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Weiwei Liu
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
20
|
Activation of Serum/Glucocorticoid Regulated Kinase 1/Nuclear Factor-κB Pathway Are Correlated with Low Sensitivity to Bortezomib and Ixazomib in Resistant Multiple Myeloma Cells. Biomedicines 2021; 9:biomedicines9010033. [PMID: 33406639 PMCID: PMC7823718 DOI: 10.3390/biomedicines9010033] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/14/2020] [Accepted: 12/30/2020] [Indexed: 11/16/2022] Open
Abstract
Multiple myeloma (MM) is an incurable malignancy often associated with primary and acquired resistance to therapeutic agents, such as proteasome inhibitors. However, the mechanisms underlying the proteasome inhibitor resistance are poorly understood. Here, we elucidate the mechanism of primary resistance to bortezomib and ixazomib in the MM cell lines, KMS-20, KMS-26, and KMS-28BM. We find that low bortezomib and ixazomib concentrations induce cell death in KMS-26 and KMS-28BM cells. However, high bortezomib and ixazomib concentrations induce cell death only in KMS-20 cells. During Gene Expression Omnibus analysis, KMS-20 cells exhibit high levels of expression of various genes, including anti-phospho-fibroblast growth factor receptor 1 (FGFR1), chemokine receptor type (CCR2), and serum and glucocorticoid regulated kinase (SGK)1. The SGK1 inhibitor enhances the cytotoxic effects of bortezomib and ixazomib; however, FGFR1 and CCR2 inhibitors do not show such effect in KMS-20 cells. Moreover, SGK1 activation induces the phosphorylation of NF-κB p65, and an NF-κB inhibitor enhances the sensitivity of KMS-20 cells to bortezomib and ixazomib. Additionally, high levels of expression of SGK1 and NF-κB p65 is associated with a low sensitivity to bortezomib and a poor prognosis in MM patients. These results indicate that the activation of the SGK1/NF-κB pathway correlates with a low sensitivity to bortezomib and ixazomib, and a combination of bortezomib and ixazomib with an SGK1 or NF-κB inhibitor may be involved in the treatment of MM via activation of the SGK1/NF-κB pathway.
Collapse
|
21
|
Greco C, Catania R, Balacco DL, Taresco V, Musumeci F, Alexander C, Huett A, Schenone S. Synthesis and Antibacterial Evaluation of New Pyrazolo[3,4- d]pyrimidines Kinase Inhibitors. Molecules 2020; 25:molecules25225354. [PMID: 33207806 PMCID: PMC7696985 DOI: 10.3390/molecules25225354] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/06/2020] [Accepted: 11/09/2020] [Indexed: 12/13/2022] Open
Abstract
Pyrazolo[3,4-d]pyrimidines represent an important class of heterocyclic compounds well-known for their anticancer activity exerted by the inhibition of eukaryotic protein kinases. Recently, pyrazolo[3,4-d]pyrimidines have become increasingly attractive for their potential antimicrobial properties. Here, we explored the activity of a library of in-house pyrazolo[3,4-d]pyrimidines, targeting human protein kinases, against Staphylococcus aureus and Escherichia coli and their interaction with ampicillin and kanamycin, representing important classes of clinically used antibiotics. Our results represent a first step towards the potential application of dual active pyrazolo[3,4-d]pyrimidine kinase inhibitors in the prevention and treatment of bacterial infections in cancer patients.
Collapse
Affiliation(s)
- Chiara Greco
- Dipartimento di Farmacia, Università di Genova, Viale Benedetto XV 3, 16132 Genova, Italy; (C.G.); (F.M.)
| | - Rosa Catania
- School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK;
| | - Dario Leonardo Balacco
- School of Dentistry, Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B5 7EG, UK;
| | - Vincenzo Taresco
- School of Chemistry, University of Nottingham, University Park, Nottingham NG7 2RD, UK;
| | - Francesca Musumeci
- Dipartimento di Farmacia, Università di Genova, Viale Benedetto XV 3, 16132 Genova, Italy; (C.G.); (F.M.)
| | - Cameron Alexander
- School of Pharmacy, University of Nottingham, University Park, Nottingham NG7 2RD, UK;
| | - Alan Huett
- School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK;
- Correspondence: (A.H.); (S.S.)
| | - Silvia Schenone
- Dipartimento di Farmacia, Università di Genova, Viale Benedetto XV 3, 16132 Genova, Italy; (C.G.); (F.M.)
- Correspondence: (A.H.); (S.S.)
| |
Collapse
|
22
|
Dattilo V, Amato R, Perrotti N, Gennarelli M. The Emerging Role of SGK1 (Serum- and Glucocorticoid-Regulated Kinase 1) in Major Depressive Disorder: Hypothesis and Mechanisms. Front Genet 2020; 11:826. [PMID: 32849818 PMCID: PMC7419621 DOI: 10.3389/fgene.2020.00826] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 07/09/2020] [Indexed: 12/28/2022] Open
Abstract
Major depressive disorder (MDD) is a heterogeneous psychiatric disease characterized by persistent low mood, diminished interests, and impaired cognitive and social functions. The multifactorial etiology of MDD is still largely unknown because of the complex genetic and environmental interactions involved. Therefore, no established mechanism can explain all the aspects of the disease. In this light, an extensive research about the pathophysiology of MDD has been carried out. Several pathogenic hypotheses, such as monoamines deficiency and neurobiological alterations in the stress-responsive system, including the hypothalamic-pituitary-adrenal (HPA) axis and the immune system, have been proposed for MDD. Over time, remarkable studies, mainly on preclinical rodent models, linked the serum- and glucocorticoid-regulated kinase 1 (SGK1) to the main features of MDD. SGK1 is a serine/threonine kinase belonging to the AGK Kinase family. SGK1 is ubiquitously expressed, which plays a pivotal role in the hormonal regulation of several ion channels, carriers, pumps, and transcription factors or regulators. SGK1 expression is modulated by cell stress and hormones, including gluco- and mineralocorticoids. Compelling evidence suggests that increased SGK1 expression or function is related to the pathogenic stress hypothesis of major depression. Therefore, the first part of the present review highlights the putative role of SGK1 as a critical mediator in the dysregulation of the HPA axis, observed under chronic stress conditions, and its controversial role in the neuroinflammation as well. The second part depicts the negative regulation exerted by SGK1 in the expression of both the brain-derived neurotrophic factor (BDNF) and the vascular endothelial growth factor (VEGF), resulting in an anti-neurogenic activity. Finally, the review focuses on the antidepressant-like effects of anti-oxidative nutraceuticals in several preclinical model of depression, resulting from the restoration of the physiological expression and/or activity of SGK1, which leads to an increase in neurogenesis. In summary, the purpose of this review is a systematic analysis of literature depicting SGK1 as molecular junction of the complex mechanisms underlying the MDD in an effort to suggest the kinase as a potential biomarker and strategic target in modern molecular antidepressant therapy.
Collapse
Affiliation(s)
- Vincenzo Dattilo
- Genetic Unit, IRCCS Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Rosario Amato
- Department of Health Sciences, Magna Graecia University of Catanzaro, Catanzaro, Italy.,Medical Genetics Unit, Mater Domini University Hospital, Catanzaro, Italy
| | - Nicola Perrotti
- Department of Health Sciences, Magna Graecia University of Catanzaro, Catanzaro, Italy.,Medical Genetics Unit, Mater Domini University Hospital, Catanzaro, Italy
| | - Massimo Gennarelli
- Genetic Unit, IRCCS Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy.,Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| |
Collapse
|
23
|
Sharif Siam MK, Sarker A, Sayeem MMS. In silico drug design and molecular docking studies targeting Akt1 (RAC-alpha serine/threonine-protein kinase) and Akt2 (RAC-beta serine/threonine-protein kinase) proteins and investigation of CYP (cytochrome P450) inhibitors against MAOB (monoamine oxidase B) for OSCC (oral squamous cell carcinoma) treatment. J Biomol Struct Dyn 2020; 39:6467-6479. [PMID: 32746771 DOI: 10.1080/07391102.2020.1802335] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The overexpression of Akt1 (RAC-alpha serine/threonine-protein Kinase) and Akt2 (RAC-beta serine/threonine-protein Kinase) is a hallmark of Oral Squamous Cell Carcinoma (OSCC). Because of the elevated frequency of OSCC occurrence in South Asian countries, novel therapeutic approaches are indispensable. Drugs that inhibit the overexpression of Akt1 and Akt2 proteins in Akt pathway and do not cause reduced expression of MAOB can be leads for OSCC treatment. In this study, Akt1, Akt2 and MAOB were targeted and 100 CYP inhibitors were screened through several in silico approaches and Galuteolin and Linarin were identified as potential leads for OSCC treatment as they inhibited Akt1 proteins with strong binding affinities of -12.3 and -11.5 kcal/mol respectively and also Akt2 proteins with strong binding affinities of -11.4 and -11.1 kcal/mol respectively, but they did not inhibit MAOB. Decreased expression of MAOB in tissues causes OSCC but overexpression is also responsible for other types of diseases and cancers. From the investigation of CYP inhibitors against MAOB, five CYP inhibitors- Diosmetin, Acacetin, Epicatechin, Eriodictyol and Capillin have expressed inhibitory action against MAOB without any interference with Akt1 and Akt2. This study mainly represents that Galuteolin and Linarin in the Akt pathway can be perceived for OSCC treatment and other five CYP inhibitors - Diosmetin, Acacetin, Epicatechin, Eriodictyol and Capillin for the treatment of other diseases and cancers caused by overexpression of MAOB. ADMET properties of CYP inhibitors obtained from admetSAR 2.0 and were compared with reference drugs for validation. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
| | - Anusree Sarker
- Department of Pharmacy, BRAC University, Dhaka, Bangladesh
| | - Mohammad Manzur Sharif Sayeem
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
24
|
Zhu R, Yang G, Cao Z, Shen K, Zheng L, Xiao J, You L, Zhang T. The prospect of serum and glucocorticoid-inducible kinase 1 (SGK1) in cancer therapy: a rising star. Ther Adv Med Oncol 2020; 12:1758835920940946. [PMID: 32728395 PMCID: PMC7364809 DOI: 10.1177/1758835920940946] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 06/16/2020] [Indexed: 12/21/2022] Open
Abstract
Serum and glucocorticoid-inducible kinase 1 (SGK1) is an AGC kinase that has been reported to be involved in a variety of physiological and pathological processes. Recent evidence has accumulated that SGK1 acts as an essential Akt-independent mediator of phosphatidylinositol 3-kinase (PI3K)/mammalian target of rapamycin (mTOR) signaling pathway in cancer. SGK1 is overexpressed in several tumors, including prostate cancer, colorectal carcinoma, glioblastoma, breast cancer, and endometrial cancer. The functions of SGK1 include regulating tumor growth, survival, metastasis, autophagy, immunoregulation, calcium (Ca2+) signaling, cancer stem cells, cell cycle, and therapeutic resistance. In this review, we introduce the pleiotropic role of SGK1 in the development and progression of tumors, summarize its downstream targets, and integrate the knowledge provided by preclinical studies that the prospect of SGK1 inhibition as a potential therapeutic approach.
Collapse
Affiliation(s)
- Ruizhe Zhu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Gang Yang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhe Cao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Kexin Shen
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lianfang Zheng
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianchun Xiao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Taiping Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing 100730, China
| |
Collapse
|