1
|
Chen J, Bao J, Jiang X, Yu W, Han Y, Zhang X, Zhang Y, Deng G. Astragaloside IV protects brain cells from ischemia-reperfusion injury by inhibiting ryanodine receptor expression and reducing the expression of P-Src and P-GRK2. Sci Rep 2024; 14:17497. [PMID: 39080440 PMCID: PMC11289356 DOI: 10.1038/s41598-024-68462-z] [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: 12/29/2022] [Accepted: 07/24/2024] [Indexed: 08/02/2024] Open
Abstract
Astragaloside IV, a prime active component of Astragalus membranaceus, has potential as a neuroprotectant. We aimed to identify the active ingredients in A. membranaceus and assess if Astragaloside IV can improve cerebral ischemia-reperfusion injury (CIRI) cell apoptosis by reducing P-Src and P-GRK2 via ryanodine receptor (RyR) expression inhibition. We used bioinformatics analysis to examine the effects of A. membranaceus on ischemic stroke. We studied brain samples from middle cerebral artery occlusion (MCAO) mice treated with normal saline, Astragaloside IV, and sham mice for pathology and Western blot tests. We also tested PC12 cells in vitro with or without Astragaloside IV or GSK180736A using Western blotting and fluorescence assays. Our bioinformatics analysis suggested a possible association between A. membranaceus, calcium ion pathways, and apoptosis pathways. Western blot data indicated Astragaloside IV significantly decreased RyR, p-Src, and downstream phosphorylated GRK2, PLC, CaMKII, and IP3R levels in MCAO mice brains. Astragaloside IV also considerably inhibited pro-apoptotic and oxidative stress-associated proteins' expression while boosting anti-apoptotic protein expression. The results suggest Astragaloside IV can inhibit RyR expression, subsequently reducing brain cell apoptosis.
Collapse
Affiliation(s)
- Juan Chen
- College of Basic Medicine, Hebei University of Chinese Medicine, No.3 Xingyuan Road, Shijiazhuang, 050200, Hebei, China
- Shenzhen Hospital of Guangzhou University of Chinese Medicine, Shenzhen, 518034, Guangdong, China
| | - Jun Bao
- College of Basic Medicine, Hebei University of Chinese Medicine, No.3 Xingyuan Road, Shijiazhuang, 050200, Hebei, China
| | - Xiujuan Jiang
- College of Basic Medicine, Hebei University of Chinese Medicine, No.3 Xingyuan Road, Shijiazhuang, 050200, Hebei, China
| | - Wentao Yu
- College of Basic Medicine, Hebei University of Chinese Medicine, No.3 Xingyuan Road, Shijiazhuang, 050200, Hebei, China
| | - Yunpeng Han
- College of Basic Medicine, Hebei University of Chinese Medicine, No.3 Xingyuan Road, Shijiazhuang, 050200, Hebei, China
| | - Xia Zhang
- College of Basic Medicine, Hebei University of Chinese Medicine, No.3 Xingyuan Road, Shijiazhuang, 050200, Hebei, China
| | - Ying Zhang
- College of Nursing, Hebei University of Chinese Medicine, No.3 Xingyuan Road, Shijiazhuang, 050200, Hebei, China.
- Hebei Key Laboratory of Health Care with Traditional Chinese Medicine, Shijiazhuang, 050200, Hebei, China.
| | - Guoxing Deng
- College of Basic Medicine, Hebei University of Chinese Medicine, No.3 Xingyuan Road, Shijiazhuang, 050200, Hebei, China.
| |
Collapse
|
2
|
Zhu C, Fan F, Li CY, Xiong Y, Liu X. Caspase-3 promotes oncogene-induced malignant transformation via EndoG-dependent Src-STAT3 phosphorylation. Cell Death Dis 2024; 15:486. [PMID: 38977663 PMCID: PMC11231138 DOI: 10.1038/s41419-024-06884-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 06/26/2024] [Accepted: 07/02/2024] [Indexed: 07/10/2024]
Abstract
Accumulating evidence suggests that caspase-3 plays critical roles beyond apoptosis, serving pro-survival functions in malignant transformation and tumorigenesis. However, the mechanism of non-apoptotic action of caspase-3 in oncogenic transformation remains unclear. In the present study, we show that caspase-3 is consistently activated in malignant transformation induced by exogenous expression of oncogenic cocktail (c-Myc, p53DD, Oct-4, and H-Ras) in vitro as well as in the mouse mammary tumor virus-polyomavirus middle T antigen (MMTV-PyMT) mouse model of breast cancer. Genetic ablation of caspase-3 significantly attenuated oncogene-induced transformation of mammalian cells and delayed breast cancer progression in MMTV-PyMT transgenic mice. Mechanistically, active caspase-3 triggers the translocation of endonuclease G (EndoG) from mitochondria, which migrates to the nucleus, thereby induces phosphorylation of Src-STAT3 signaling pathway to facilitate oncogenic transformation. Taken together, our data suggest that caspase-3 plays pivotal role in facilitating rather than suppressing oncogene-induced malignant transformation of mammalian cells.
Collapse
Affiliation(s)
- Chenchen Zhu
- Department of Biochemistry, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Fushun Fan
- BeBetter Med Inc., Guangzhou, Guangdong, China
| | - Chuan-Yuan Li
- Department of Dermatology, Duke University Medical Center, Durham, NC, USA
| | - Yan Xiong
- Guangzhou Consen Pharmaceutical Technology Co. Ltd, Guangzhou, Guangdong, China.
| | - Xinjian Liu
- Department of Biochemistry, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, China.
| |
Collapse
|
3
|
Li R, Wang D, Yang H, Pu L, Li X, Yang F, Zhu R. Important role and underlying mechanism of non‑SMC condensin I complex subunit G in tumours (Review). Oncol Rep 2024; 51:77. [PMID: 38639175 DOI: 10.3892/or.2024.8736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 03/21/2024] [Indexed: 04/20/2024] Open
Abstract
At present, the incidence of tumours is increasing on a yearly basis, and tumourigenesis is usually associated with chromosomal instability and cell cycle dysregulation. Moreover, abnormalities in the chromosomal structure often lead to DNA damage, further exacerbating gene mutations and chromosomal rearrangements. However, the non‑SMC condensin I complex subunit G (NCAPG) of the structural maintenance of chromosomes family is known to exert a key role in tumour development. It has been shown that high expression of NCAPG is closely associated with tumour development and progression. Overexpression of NCAPG variously affects chromosome condensation and segregation during cell mitosis, influences cell cycle regulation, promotes tumour cell proliferation and invasion, and inhibits apoptosis. In addition, NCAPG has been associated with tumour cell stemness, tumour resistance and recurrence. The aim of the present review was to explore the underlying mechanisms of NCAPG during tumour development, with a view towards providing novel targets and strategies for tumour therapy, and through the elucidation of the mechanisms involved, to lay the foundation for future developments in health.
Collapse
Affiliation(s)
- Ruobing Li
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Dechun Wang
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Hong Yang
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Leilei Pu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Xiaohong Li
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Fumei Yang
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Rong Zhu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| |
Collapse
|
4
|
Inoue M, Ekimoto T, Yamane T, Ikeguchi M. Computational Analysis of Activation of Dimerized Epidermal Growth Factor Receptor Kinase Using the String Method and Markov State Model. J Chem Inf Model 2024; 64:3884-3895. [PMID: 38670929 DOI: 10.1021/acs.jcim.4c00172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
Abstract
Epidermal growth factor receptor (EGFR) activation is accompanied by dimerization. During the activation of the intracellular kinase domain, two EGFR kinases form an asymmetric dimer, and one side of the dimer (receiver) is activated. Using the string method and Markov state model (MSM), we performed a computational analysis of the structural changes in the activation of the EGFR dimer in this study. The string method reveals the minimum free-energy pathway (MFEP) from the inactive to active structure. The MSM was constructed from numerous trajectories of molecular dynamics simulations around the MFEP, which revealed the free-energy map of structural changes. In the activation of the receiver kinase, the unfolding of the activation loop (A-loop) is followed by the rearrangement of the C-helix, as observed in other kinases. However, unlike other kinases, the free-energy map of EGFR at the asymmetric dimer showed that the active state yielded the highest stability and revealed how interactions at the dimer interface induced receiver activation. As the H-helix of the activator approaches the C-helix of the receiver during activation, the A-loop unfolds. Subsequently, L782 of the receiver enters the pocket between the G- and H-helices of the activator, leading to a rearrangement of the hydrophobic residues around L782 of the receiver, which constitutes a structural rearrangement of the C-helix of the receiver from an outward to an inner position. The MSM analysis revealed long-time scale trajectories via kinetic Monte Carlo.
Collapse
Affiliation(s)
- Masao Inoue
- Graduate School of Medical Life Science, Yokohama City University, 1-7-29, Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
| | - Toru Ekimoto
- Graduate School of Medical Life Science, Yokohama City University, 1-7-29, Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
| | - Tsutomu Yamane
- HPC- and AI-driven Drug Development Platform Division, Center for Computational Science, RIKEN, 1-7-22, Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
| | - Mitsunori Ikeguchi
- Graduate School of Medical Life Science, Yokohama City University, 1-7-29, Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
- HPC- and AI-driven Drug Development Platform Division, Center for Computational Science, RIKEN, 1-7-22, Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
| |
Collapse
|
5
|
Ramos R, Vale N. Dual Drug Repurposing: The Example of Saracatinib. Int J Mol Sci 2024; 25:4565. [PMID: 38674150 PMCID: PMC11050334 DOI: 10.3390/ijms25084565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/11/2024] [Accepted: 04/20/2024] [Indexed: 04/28/2024] Open
Abstract
Saracatinib (AZD0530) is a dual Src/Abl inhibitor initially developed by AstraZeneca for cancer treatment; however, data from 2006 to 2024 reveal that this drug has been tested not only for cancer treatment, but also for the treatment of other diseases. Despite the promising pre-clinical results and the tolerability shown in phase I trials, where a maximum tolerated dose of 175 mg was defined, phase II clinical data demonstrated a low therapeutic action against several cancers and an elevated rate of adverse effects. Recently, pre-clinical research aimed at reducing the toxic effects and enhancing the therapeutic performance of saracatinib using nanoparticles and different pharmacological combinations has shown promising results. Concomitantly, saracatinib was repurposed to treat Alzheimer's disease, targeting Fyn. It showed great clinical results and required a lower daily dose than that defined for cancer treatment, 125 mg and 175 mg, respectively. In addition to Alzheimer's disease, this Src inhibitor has also been studied in relation to other health conditions such as pulmonary and liver fibrosis and even for analgesic and anti-allergic functions. Although saracatinib is still not approved by the Food and Drug Administration (FDA), the large number of alternative uses for saracatinib and the elevated number of pre-clinical and clinical trials performed suggest the huge potential of this drug for the treatment of different kinds of diseases.
Collapse
Affiliation(s)
- Raquel Ramos
- PerMed Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal;
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Nuno Vale
- PerMed Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal;
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Department of Community Medicine, Health Information and Decision (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
| |
Collapse
|
6
|
Yao L, Fang J, Zhao J, Yu J, Zhang X, Chen W, Han L, Peng D, Chen Y. Dendrobium huoshanense in the treatment of ulcerative colitis: Network pharmacology and experimental validation. JOURNAL OF ETHNOPHARMACOLOGY 2024; 323:117729. [PMID: 38190953 DOI: 10.1016/j.jep.2024.117729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 12/31/2023] [Accepted: 01/05/2024] [Indexed: 01/10/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Dendrobium huoshanense C. Z. Tang et S. J. Cheng (DH) is a traditional medicinal herb with a long history of medicinal use. DH has been recorded as protecting the gastrointestinal function. Modern pharmacology research shows that DH regulates intestinal flora, intestinal mucosal immunity, gastrointestinal peristalsis and secretion of digestive juices. At the same time, some studies have shown that DH has a good therapeutic effect on ulcerative colitis, but its mechanism of action has not been fully elucidated. AIMS OF THIS STUDY To investigate the mechanism and effect of Dendrobium huoshanense C. Z. Tang et S. J. Cheng (DH) in the treatment of ulcerative colitis (UC) by combining network pharmacology and in vivo experimental validation. METHODS A network pharmacology approach was used to perform component screening, target prediction, PPI network interaction analysis, GO and KEGG enrichment analysis to initially predict the mechanism of DH treatment for UC. Then, the mechanism was validated with the UC mouse model induced by 3% DSS. RESULTS Based on the network pharmacological analysis, a comprehensive of 101 active components were identified, with 19 of them potentially serving as the crucial elements in DH's effectiveness against UC treatment. Additionally, the study revealed 314 potential core therapeutic targets along with the top 5 key targets: SRC, STAT3, AKT1, HSP90AA1, and PIK3CA. In experiments conducted on live mice with UC, DH was found to decrease the levels of IL-6 and TNF-α in the blood, while increasing the levels of IL-10 and TGF-β. This led to notable improvements in colon length, injury severity, and an up-regulation of SRC, STAT3, HSP90AA1, PIK3CA, p-AKT1 and PI3K/AKT signaling pathway expression in the colon tissue. CONCLUSIONS In this study, the active components and main targets of DH for UC treatment were initially forecasted, and the potential mechanism was investigated through network pharmacology. These findings offer an experimental foundation for the clinical utilization of DH.
Collapse
Affiliation(s)
- Liang Yao
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China; MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, 230012, Anhui, China.
| | - Jing Fang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China.
| | - Junwei Zhao
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China.
| | - Jiao Yu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China.
| | - Xiaoqian Zhang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China.
| | - Weidong Chen
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China; MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, 230012, Anhui, China; Key Laboratory of Modern Traditional Chinese Medicines of Anhui Higher Education Institutes, Anhui University of Chinese Medicine, Hefei, 230038, Anhui, China.
| | - Lan Han
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China; MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, 230012, Anhui, China; Key Laboratory of Modern Traditional Chinese Medicines of Anhui Higher Education Institutes, Anhui University of Chinese Medicine, Hefei, 230038, Anhui, China.
| | - Daiyin Peng
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China; MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, 230012, Anhui, China; Xin'an Medicine, Key Laboratory of Chinese Ministry of Education, Anhui University of Chinese Medicine, Hefei, 230038, Anhui, China; Key Laboratory of Modern Traditional Chinese Medicines of Anhui Higher Education Institutes, Anhui University of Chinese Medicine, Hefei, 230038, Anhui, China.
| | - Yunna Chen
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China.
| |
Collapse
|
7
|
Wang C, Zhang R, Chen X, Yuan M, Wu J, Sun Q, Miao C, Jing Y. The potential effect and mechanism of Saikosaponin A against gastric cancer. BMC Complement Med Ther 2023; 23:295. [PMID: 37608281 PMCID: PMC10463516 DOI: 10.1186/s12906-023-04108-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 07/28/2023] [Indexed: 08/24/2023] Open
Abstract
BACKGROUND Saikosaponin A (SSA) shows a series of pharmacological activities, such as anti-inflammatory, antioxidant and antitumor. However, there is a lack of comprehensive research or sufficient evidence regarding the efficacy of SSA in treating gastric cancer (GC), and the specific mechanisms by which it inhibits GC growth and progression are still not fully understood. METHODS MTT and clonogenic assays were employed to detect the effect of SSA on the proliferation of GC cells. Bioinformatics predicted the SSA targets in the treatment of GC. The core genes and the underlying mechanism of SSA in anti-GC were obtained by analyzing the intersecting targets; molecular docking and Western blot were used to check the reliability of core genes. Flow cytometry was used to analyze apoptosis and cell cycle in GC cells treated with varying concentrations of SSA. Western blot was employed to detect the expression levels of related proteins. RESULTS SSA significantly blocked GC cells in the S phase of the cell cycle and induced apoptosis to suppress the proliferation of GC cells. Network pharmacology revealed that the underlying mechanisms through which SSA acts against GC involve the modulation of several signaling pathways, including the PI3K-Akt, MAPK, RAS, and T-cell signaling pathways. Molecular docking showed pivotal target genes with a high affinity to SSA, including STAT3, MYC, TNF, STAT5B, Caspase-3 and SRC. Furthermore, western blot results revealed that SSA significantly increased the protein levels of Bax and Cleaved Caspase-3, whereas decreased the expression levels of p-JAK, p-STAT3, MYC, Bcl-2, p-PI3K, p-AKT and p-mTOR, confirming that the reliability of hub targets and SSA could promote GC cell apoptosis by suppressing PI3K/AKT/mTOR pathway. CONCLUSIONS The results suggest that SSA has the ability to trigger apoptosis in GC cells by blocking the PI3K/AKT/mTOR pathway. These findings highlight the potential of SSA as a promising natural therapeutic agent for the treatment of GC.
Collapse
Affiliation(s)
- Chao Wang
- China Pharmaceutical University, Nanjing Drum Tower Hospital, Nanjing, 210008, Jiangsu Province, China
| | - Ruijuan Zhang
- Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Xu Chen
- Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Mengyun Yuan
- Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Jian Wu
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China
| | - Qingmin Sun
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China
| | - Chunrun Miao
- Department of Gastroenterology, Dongtai Hospital of Traditional Chinese Medicine, Dongtai, 224299, Jiangsu, China.
| | - Yali Jing
- China Pharmaceutical University, Nanjing Drum Tower Hospital, Nanjing, 210008, Jiangsu Province, China.
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, No. 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China.
| |
Collapse
|
8
|
Suresh K. "Lyn"king Emphysema and Cancer Development: Insights from Src Family Kinase Gain-of-Function Models. Am J Respir Cell Mol Biol 2023; 69:8-9. [PMID: 37079882 PMCID: PMC10324041 DOI: 10.1165/rcmb.2023-0122ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2023] Open
Affiliation(s)
- Karthik Suresh
- Department of Medicine Johns Hopkins University School of Medicine Baltimore, Maryland
| |
Collapse
|
9
|
Desjardins-Lecavalier N, Annis MG, Nowakowski A, Kiepas A, Binan L, Roy J, Modica G, Hébert S, Kleinman CL, Siegel PM, Costantino S. Migration speed of captured breast cancer subpopulations correlates with metastatic fitness. J Cell Sci 2023; 136:jcs260835. [PMID: 37313743 PMCID: PMC10657211 DOI: 10.1242/jcs.260835] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 06/02/2023] [Indexed: 06/15/2023] Open
Abstract
The genetic alterations contributing to migration proficiency, a phenotypic hallmark of metastatic cells required for colonizing distant organs, remain poorly defined. Here, we used single-cell magneto-optical capture (scMOCa) to isolate fast cells from heterogeneous human breast cancer cell populations, based on their migratory ability alone. We show that captured fast cell subpopulations retain higher migration speed and focal adhesion dynamics over many generations as a result of a motility-related transcriptomic profile. Upregulated genes in isolated fast cells encoded integrin subunits, proto-cadherins and numerous other genes associated with cell migration. Dysregulation of several of these genes correlates with poor survival outcomes in people with breast cancer, and primary tumors established from fast cells generated a higher number of circulating tumor cells and soft tissue metastases in pre-clinical mouse models. Subpopulations of cells selected for a highly migratory phenotype demonstrated an increased fitness for metastasis.
Collapse
Affiliation(s)
- Nicolas Desjardins-Lecavalier
- Maisonneuve-Rosemont Hospital Research Center, 5415, boulevard de l'Assomption, Montréal, QC H1T 2M4, Canada
- Institut de genie biomedical, University of Montreal, Pavillon Paul-G.-Desmarais, 2960, chemin de la Tour, Montréal, QC H3T 1J4, Canada
| | - Matthew G. Annis
- Goodman Cancer Institute, McGill University, 1160 Pine Avenue West, Montreal, QC H3A 1A3, Canada
- Department of Medicine, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada
| | - Alexander Nowakowski
- Goodman Cancer Institute, McGill University, 1160 Pine Avenue West, Montreal, QC H3A 1A3, Canada
- Department of Medicine, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada
| | - Alexander Kiepas
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health Bethesda, MA 20892-4370, USA
| | - Loïc Binan
- Maisonneuve-Rosemont Hospital Research Center, 5415, boulevard de l'Assomption, Montréal, QC H1T 2M4, Canada
| | - Joannie Roy
- Maisonneuve-Rosemont Hospital Research Center, 5415, boulevard de l'Assomption, Montréal, QC H1T 2M4, Canada
| | - Graziana Modica
- Maisonneuve-Rosemont Hospital Research Center, 5415, boulevard de l'Assomption, Montréal, QC H1T 2M4, Canada
| | - Steven Hébert
- Lady Davis Institute, McGill University, Montréal, QC H3T 1E2, Canada
| | - Claudia L. Kleinman
- Lady Davis Institute, McGill University, Montréal, QC H3T 1E2, Canada
- Department of Human Genetics, McGill University, Montréal, QC H3T 1E2, Canada
| | - Peter M. Siegel
- Goodman Cancer Institute, McGill University, 1160 Pine Avenue West, Montreal, QC H3A 1A3, Canada
- Department of Medicine, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada
| | - Santiago Costantino
- Maisonneuve-Rosemont Hospital Research Center, 5415, boulevard de l'Assomption, Montréal, QC H1T 2M4, Canada
- Department of Ophthalmology, University of Montreal, Pavillon Roger-Gaudry, Bureau S-700, 2900, boul. Édouard-Montpetit, Montréal, QC H3T 1J4, Canada
| |
Collapse
|
10
|
Ichikawa-Tomikawa N, Sugimoto K, Kashiwagi K, Chiba H. The Src-Family Kinases SRC and BLK Contribute to the CLDN6-Adhesion Signaling. Cells 2023; 12:1696. [PMID: 37443730 PMCID: PMC10341166 DOI: 10.3390/cells12131696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/16/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Cell adhesion molecules, including integrins, cadherins, and claudins (CLDNs), are known to activate Src-family kinases (SFKs) that organize a variety of physiological and pathological processes; however, the underlying molecular basis remains unclear. Here, we identify the SFK members that are coupled with the CLDN6-adhesion signaling. Among SFK subtypes, BLK, FGR, HCK, and SRC were highly expressed in F9 cells and concentrated with CLDN6 along cell borders during epithelial differentiation. Immunoprecipitation assay showed that BLK and SRC, but not FGR or HCK, form a complex with CLDN6 via the C-terminal cytoplasmic domain. We also demonstrated, by pull-down assay, that recombinant BLK and SRC proteins directly bind to the C-terminal cytoplasmic domain of CLDN6 (CLDN6C). Unexpectedly, both recombinant SFK proteins recognized the CLDN6C peptide in a phosphotyrosine-independent manner. Furthermore, by comparing phenotypes of F9:Cldn6:Blk-/- and F9:Cldn6:Src-/- cells with those of wild-type F9 and F9:Cldn6 cells, we revealed that BLK and SRC are essential for CLDN6-triggered cellular events, namely epithelial differentiation and the expression of retinoid acid receptor target genes. These results indicate that selective SFK members appear to participate in the CLDN-adhesion signaling.
Collapse
Affiliation(s)
| | | | | | - Hideki Chiba
- Department of Basic Pathology, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan; (N.I.-T.); (K.S.); (K.K.)
| |
Collapse
|
11
|
Pan J, Tang J, Gai J, Jin Y, Tang B, Fan X. Exploring the mechanism of Ginkgo biloba L. leaves in the treatment of vascular dementia based on network pharmacology, molecular docking, and molecular dynamics simulation. Medicine (Baltimore) 2023; 102:e33877. [PMID: 37233418 PMCID: PMC10219709 DOI: 10.1097/md.0000000000033877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 05/08/2023] [Indexed: 05/27/2023] Open
Abstract
BACKGROUND Ginkgo biloba L. leaves (GBLs) play a substantial role in the treatment of vascular dementia (VD); however, the underlying mechanisms of action are unclear. OBJECTIVE This study was conducted to investigate the mechanisms of action of GBLs in the treatment of VD through network pharmacology, molecular docking, and molecular dynamics simulations. METHODS The active ingredients and related targets of GBLs were screened using the traditional Chinese medicine systems pharmacology, Swiss Target Prediction and GeneCards databases, and the VD-related targets were screened using the OMIM, DrugBank, GeneCards, and DisGeNET databases, and the potential targets were identified using a Venn diagram. We used Cytoscape 3.8.0 software and the STRING platform to construct traditional Chinese medicine-active ingredient-potential target and protein-protein interaction networks, respectively. After gene ontology and Kyoto Encyclopedia of Genes and Genomes pathway analysis of potential targets using the DAVID platform, the binding affinity between key active ingredients and targets was analyzed by molecular docking, and finally, the top 3 proteins-ligand pairs with the best binding were simulated by molecular dynamics to verify the molecular docking results. RESULTS A total of 27 active ingredients of GBLs were screened and 274 potential targets involved in the treatment of VD were identified. Quercetin, luteolin, kaempferol, and ginkgolide B were the core ingredients for treatment, and AKT1, TNF, IL6, VEGFA, IL1B, TP53, CASP3, SRC, EGFR, JUN, and EGFR were the main targets of action. The main biological processes involved apoptosis, inflammatory response, cell migration, lipopolysaccharide response, hypoxia response, and aging. PI3K/Akt appeared to be a key signaling pathway for GBLs in the treatment of VD. Molecular docking displayed strong binding affinity between the active ingredients and the targets. Molecular dynamics simulation results further verified the stability of their interactions. CONCLUSION SUBSECTIONS This study revealed the potential molecular mechanisms involved in the treatment of VD by GBLs using multi-ingredient, multi-target, and multi-pathway interactions, providing a theoretical basis for the clinical treatment and lead drug development of VD.
Collapse
Affiliation(s)
- Jienuo Pan
- School of Rehabilitation Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jiqin Tang
- School of Rehabilitation Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jialin Gai
- School of Rehabilitation Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yilan Jin
- School of International Education, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Bingshun Tang
- School of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiaohua Fan
- Department of Rehabilitation Medicine, Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
12
|
Yang N, Qi X, Hu J, Teng J, Wang Y, Li C. Exploring the mechanism of astragalus membranaceus in the treatment of multiple system atrophy based on network pharmacology and molecular docking. Medicine (Baltimore) 2023; 102:e32523. [PMID: 36749251 PMCID: PMC9901982 DOI: 10.1097/md.0000000000032523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 12/09/2022] [Indexed: 02/08/2023] Open
Abstract
Multiple system atrophy (MSA) is a fatal neurodegenerative disease, it causes functional degradation of multiple organs and systems throughout the body. Astragalus membranaceus (AM), a well-known traditional Chinese medicine, has been used to improve muscle wasting-related disorders for a long history. In this study, we used network pharmacology and molecular docking to predict the mechanism underlying AM for the treatment of MSA. We screened the active compounds of AM and its related targets, as well as the target proteins of MSA. We made a Venn diagram to obtain the intersecting targets and then constructed a protein-protein interaction network to find the core targets and build an active ingredient-target network map. After subjecting the intersecting targets to gene ontology and Kyoto encyclopedia of genes and genomes analysis, the binding ability of core compounds and core target proteins were validated by molecular docking. A total of 20 eligible compounds and 274 intersecting targets were obtained. The core components of treatment are quercetin, kaempferol, and isorhamnetin, and the core targets are TP53, RELA, and TNF. The main biological processes are related to cellular responses and regulation. Molecular functions are mainly associated with apoptosis, inflammation, and tumorigenesis. Molecular docking results show good and standard binding abilities. This study illustrates that AM treats MSA through multiple targets and pathways, and provides a reference for subsequent research.
Collapse
Affiliation(s)
- Ni Yang
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xianghua Qi
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jing Hu
- Shandong Public Health Clinical Center, Jinan, China
| | - Jing Teng
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yuangeng Wang
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chunlin Li
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
13
|
Kuo MY, Yang WT, Ho YJ, Chang GM, Chang HH, Hsu CY, Chang CC, Chen YH. Hispolon Methyl Ether, a Hispolon Analog, Suppresses the SRC/STAT3/Survivin Signaling Axis to Induce Cytotoxicity in Human Urinary Bladder Transitional Carcinoma Cell Lines. Int J Mol Sci 2022; 24:ijms24010138. [PMID: 36613579 PMCID: PMC9820424 DOI: 10.3390/ijms24010138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/14/2022] [Accepted: 12/17/2022] [Indexed: 12/24/2022] Open
Abstract
Bladder cancer is a leading human malignancy worldwide. Signal transducer and activator of transcription (STAT) 3 is an oncogenic transcription factor commonly hyperactivated in most human cancers, including bladder cancer. Notably, preclinical evidence has validated STAT3 blockade as a promising therapeutic strategy for bladder cancer. Hispolon Methyl Ether (HME) is a structural analog of hispolon, an anticancer component of the medicinal mushroom Phellinus linteus. Thus far, HME's anticancer activity and mechanisms remain largely unknown. We herein report HME was cytotoxic, more potent than cisplatin, and proapoptotic to various human bladder transitional carcinoma cell lines. Of note, HME blocked STAT3 activation, evidenced by HME-elicited reduction in tyrosine 705-phosphorylated STAT3 levels constitutively expressed or induced by interleukin-6. Significantly, HME-induced cytotoxicity was abrogated in cells expressing a dominant-active STAT3 mutant (STAT3-C), confirming STAT3 blockage as a pivotal mechanism of HME's cytotoxic action. We further revealed that survivin was downregulated by HME, while its levels were rescued in STAT3-C-expressing cells. Moreover, survivin overexpression abolished HME-induced cytotoxicity, illustrating survivin as a central downstream mediator of STAT3 targeted by HME. Lastly, HME was shown to lower tyrosine 416-phosphorylated SRC levels, suggesting that HME inhibits STAT3 by repressing the activation of SRC, a STAT3 upstream kinase. In conclusion, we present the first evidence of HME's anti-bladder cancer effect, likely proceeding by evoking apoptosis through suppression of the antiapoptotic SRC/STAT3/survivin signaling axis.
Collapse
Affiliation(s)
- Min-Yung Kuo
- Pediatric Surgery Division, Department of Surgery, Tungs’ Taichung MetroHarbor Hospital, Taichung 402202, Taiwan
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung 402202, Taiwan
| | - Wei-Ting Yang
- Department of Life Sciences, National Chung Hsing University, Taichung 402202, Taiwan
| | - Yann-Jen Ho
- Department of Life Sciences, National Chung Hsing University, Taichung 402202, Taiwan
| | - Ge-Man Chang
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung 402202, Taiwan
| | - Hsiung-Hao Chang
- Department of Life Sciences, National Chung Hsing University, Taichung 402202, Taiwan
| | - Chao-Yu Hsu
- Division of Urology, Department of Surgery, Tungs’ Taichung MetroHarbor Hospital, Taichung 402202, Taiwan
- Ph.D. Program in Translational Medicine, Rong Hsing Research Center for Translational Medicine, The iEGG and Animal Biotechnology Research Center, National Chung Hsing University, Taichung 402202, Taiwan
| | - Chia-Che Chang
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung 402202, Taiwan
- Department of Life Sciences, National Chung Hsing University, Taichung 402202, Taiwan
- Ph.D. Program in Translational Medicine, Rong Hsing Research Center for Translational Medicine, The iEGG and Animal Biotechnology Research Center, National Chung Hsing University, Taichung 402202, Taiwan
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung 413305, Taiwan
- Department of Medical Research, China Medical University Hospital, Taichung 404327, Taiwan
- Traditional Herbal Medicine Research Center, Taipei Medical University Hospital, Taipei 110301, Taiwan
- Correspondence: or (C.-C.C.); or (Y.-H.C.)
| | - Yi-Hsin Chen
- Department of Nephrology, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung 427213, Taiwan
- School of Medicine, Tzu Chi University, Hualein 970374, Taiwan
- Correspondence: or (C.-C.C.); or (Y.-H.C.)
| |
Collapse
|
14
|
Yan Q, Yang J, Yao Y, Jia Z, Wang Y, Cheng M, Yan X, Xu Y. Research of the Active Components and Potential Mechanisms of Qingfei Gujin Decoction in the Treatment of Osteosarcoma Based on Network Pharmacology and Molecular Docking Technology. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:7994425. [PMID: 36466554 PMCID: PMC9713469 DOI: 10.1155/2022/7994425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 10/09/2022] [Accepted: 11/03/2022] [Indexed: 09/10/2024]
Abstract
Aim Qingfei Gujin Decoction (QGD) has been shown to be effective against osteosarcoma. This research was aimed at investigating the main active ingredients and potential mechanisms of QGD acting on osteosarcoma through network pharmacology and molecular docking techniques. Methods The active ingredients and targets of QGD were screened from the TCMSP database, and the predicted targets were obtained from the PharmMapper database. Meanwhile, the targets of osteosarcoma were collected using OMIM, PharmGKB, and DisGeNET databases. Then, GO and KEGG enrichment analyses were performed by RStudio. PPI and drug-ingredient-target networks were constructed using Cytoscape 3.2.1 to screen the major active ingredients, key networks, and targets. Finally, molecular docking of key genes and their regulatory active ingredients was performed using AutoDockTools 1.5.6 software. Results 38 active ingredients were collected, generating 89 cross-targets; quercetin, luteolin, β-sitosterol, and kaempferol were the main active ingredients of QGD acting on osteosarcoma, and major signaling pathways such as PI3K-Akt signaling pathway, MAPK signaling pathway, and IL-17 signaling pathway were observed. TP53, SRC, and ESR1 were identified as key proteins that docked well with their regulated compounds. Conclusion QGD is effective against osteosarcoma through multicomponent, multitarget, and multipathway. This study was helpful for finding effective targets and compounds for osteosarcoma treatment.
Collapse
Affiliation(s)
- Qingying Yan
- Department of Oncology, Hangzhou Third People's Hospital, Hangzhou, China
- Department of Oncology, Affiliated Hangzhou Dermatology Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiewen Yang
- Department of Oncology, Hangzhou Third People's Hospital, Hangzhou, China
| | - Yongwei Yao
- Department of Oncology, Hangzhou Third People's Hospital, Hangzhou, China
| | - Zhen Jia
- Department of Oncology, Hangzhou Third People's Hospital, Hangzhou, China
| | - Yiqing Wang
- Department of Oncology, Hangzhou Third People's Hospital, Hangzhou, China
| | - Miao Cheng
- Department of Oncology, Hangzhou Third People's Hospital, Hangzhou, China
| | - Xiaobo Yan
- Department of Orthopedics, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yefeng Xu
- Department of Oncology, Hangzhou Third People's Hospital, Hangzhou, China
| |
Collapse
|
15
|
Zhang W, Yuan Y, Huang G, Xiao J. Potential Molecular Mechanism of Guishen Huoxue Decoction against Intrauterine Adhesion Based on Network Pharmacology. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:4049147. [PMID: 36193142 PMCID: PMC9525774 DOI: 10.1155/2022/4049147] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/31/2022] [Accepted: 09/06/2022] [Indexed: 11/18/2022]
Abstract
Objective Intrauterine adhesion (IUA) represents an endometrial repair disorder that is associated with menstrual disorders, recurrent pregnancy loss, and infertility. This study aimed to explore the underlying biological mechanisms of Guishen Huoxue decoction for the treatment of IUA based on network pharmacology. Methods The selection of active compounds for Guishen Huoxue decoction and prediction of relevant targets were performed by the TCMSP and Swiss Target Prediction databases, respectively. The targets of IUA were obtained by three databases, including Online Mendelian Inheritance in Man (OMIM), DisGeNET, and GeneCards. The drug-disease regulatory network was constructed via Cytoscape software, following the acquisition of common genes of active compounds of drug Guishen Huoxue decoction and disease IUA, which was carried out through Venny software. Protein-protein interaction (PPI) network and function enrichment analyses were performed. Results According to the data obtained from TCMSP, a total of 200 potential active compounds of Guishen Huoxue decoction and their related targets (1068) were screened by the Swiss Target Prediction database. 1303 disease targets and 134 common targets were identified. The drug-disease regulatory network showed that 165 active compounds were found to be involved in the treatment of IUA. Among 134 common targets, AKT1, SRC, TP53, VEGFA, and IL-6 were predicted as core genes against IUA. PI3K-Akt, Rap1, Ras, and AGE-RAGE were the main signaling pathways that participated in the treatment of Guishen Huoxue decoction for IUA. Conclusion The active compounds of Guishen Huoxue decoction confer therapeutic effects against IUA by regulating fibrosis, inflammation, and oxidative stress through major signaling pathways such as PI3K-Akt and AGE-RAGE.
Collapse
Affiliation(s)
- Wenyan Zhang
- Department of Gynaecology, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Gynaecology, Shenzhen Bao'an Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Yuan Yuan
- Department of Gynaecology, Shenzhen Bao'an Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Guangrong Huang
- Department of Gynaecology, Shenzhen Bao'an Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Jing Xiao
- Department of Gynaecology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
16
|
Jeong JH, Park HJ, Park SH, Choi YH, Chi GY. β2-Adrenergic Receptor Signaling Pathway Stimulates the Migration and Invasion of Cancer Cells via Src Activation. Molecules 2022; 27:molecules27185940. [PMID: 36144682 PMCID: PMC9503488 DOI: 10.3390/molecules27185940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/08/2022] [Accepted: 09/10/2022] [Indexed: 11/16/2022] Open
Abstract
Chronic stress has been reported to stimulate the release of catecholamines, including norepinephrine (NE) and epinephrine (E), which promote cancer progression by activating the adrenergic receptor (AR). Although previous studies showed that β2-AR mediated chronic stress-induced tumor growth and metastasis, the underlying mechanism has not been fully explored. In this study, we aimed to investigate the molecular mechanism by which β2-AR exerts a pro-metastatic function in hepatocarcinoma (HCC) cells and breast cancer (BC) cells. Our results showed that Hep3B human HCC cells and MDA-MB-231 human BC cells exhibited the highest ADRB2 expression among diverse HCC and BC cell lines. NE, E, and isoprenaline (ISO), adrenergic agonists commonly increased the migration and invasion of Hep3B cells and MDA-MB-231 cells. The phosphorylation level of Src was significantly increased by E/NE. Dasatinib, a Src kinase inhibitor, blocked E/NE-induced migration and invasion, indicating that AR agonists enhanced the mobility of cancer cells by activating Src. ADRB2 knockdown attenuated E/NE-induced Src phosphorylation, as well as the metastatic ability of cancer cells, suggesting the essential role of β2-AR. Taken together, our results demonstrate that chronic stress-released catecholamines promoted the migration and invasion of HCC cells and BC cells via β2-AR-mediated Src activation.
Collapse
Affiliation(s)
- Jae-Hoon Jeong
- Department of Pathology, College of Korean Medicine, Dong-eui University, Busan 47227, Korea
| | - Hyun-Ji Park
- Department of Pathology, College of Korean Medicine, Dong-eui University, Busan 47227, Korea
| | - Shin-Hyung Park
- Department of Pathology, College of Korean Medicine, Dong-eui University, Busan 47227, Korea
- Correspondence: ; Tel.: +82-51-890-3332
| | - Yung-Hyun Choi
- Department of Biochemistry, College of Korean Medicine, Dong-eui University, Busan 47227, Korea
| | - Gyoo-Yong Chi
- Department of Pathology, College of Korean Medicine, Dong-eui University, Busan 47227, Korea
| |
Collapse
|
17
|
Huang CF, Gottardi CJ, Mrksich M. Tyrosine phosphatase activity is restricted by basic charge substituting mutation of substrates. Sci Rep 2022; 12:15095. [PMID: 36064958 PMCID: PMC9445012 DOI: 10.1038/s41598-022-19133-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 08/24/2022] [Indexed: 11/09/2022] Open
Abstract
Phosphorylation controls important cellular signals and its dysregulation leads to disease. While most phospho-regulation studies are focused on kinases, phosphatases are comparatively overlooked. Combining peptide arrays with SAMDI mass spectrometry, we show that tyrosine phosphatase activity is restricted by basic amino acids adjacent to phosphotyrosines. We validate this model using two β-catenin mutants associated with cancer (T653R/K) and a mouse model for intellectual disability (T653K). These mutants introduce a basic residue next to Y654, an established phosphorylation site where modification shifts β-catenin from cell-cell adhesions and towards its essential nuclear role as Wnt-signaling effector. We show that T653-basic mutant β-catenins are less efficiently dephosphorylated by phosphatases, leading to sustained Y654 phosphorylation and elevated Wnt signals, similar to those observed for Y654E phospho-mimic mutant mice. This model rationalizes how basic mutations proximal to phosphotyrosines can restrict counter-regulation by phosphatases, providing new mechanismistic and treatment insights for 6000+ potentially relevant cancer mutations.
Collapse
Affiliation(s)
- Che-Fan Huang
- Department of Chemistry, Northwestern University, Evanston, IL, 60208, USA
| | - Cara J Gottardi
- Division of Pulmonary and Critical Care, Department of Medicine, Northwestern University, Chicago, IL, 60611, USA.
- Biochemistry and Molecular Genetics, Northwestern University, Chicago, IL, 60611, USA.
| | - Milan Mrksich
- Department of Chemistry, Northwestern University, Evanston, IL, 60208, USA.
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, 60208, USA.
- Department of Cell & Developmental Biology, Northwestern University, Chicago, IL, 60611, USA.
| |
Collapse
|
18
|
Novel inhibitor of hematopoietic cell kinase as a potential therapeutic agent for acute myeloid leukemia. Cancer Immunol Immunother 2022; 71:1909-1921. [DOI: 10.1007/s00262-021-03111-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 11/10/2021] [Indexed: 10/19/2022]
|
19
|
Wang H, Chi L, Yu F, Dai H, Si X, Gao C, Wang Z, Liu L, Zheng J, Ke Y, Liu H, Zhang Q. The overview of Mitogen-activated extracellular signal-regulated kinase (MEK)-based dual inhibitor in the treatment of cancers. Bioorg Med Chem 2022; 70:116922. [PMID: 35849914 DOI: 10.1016/j.bmc.2022.116922] [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: 05/24/2022] [Revised: 07/01/2022] [Accepted: 07/06/2022] [Indexed: 11/02/2022]
Abstract
Mitogen-activated extracellular signal-regulated kinase 1 and 2 (MEK1/2) are the critical components of the mitogen-activated protein kinase/extracellular signal-regulated kinase 1 and 2 (MAPK/ERK1/2) signaling pathway which is one of the well-characterized kinase cascades regulating cell proliferation, differentiation, growth, metabolism, survival and mobility both in normal and cancer cells. The aberrant activation of MAPK/ERK1/2 pathway is a hallmark of numerous human cancers, therefore targeting the components of this pathway to inhibit its dysregulation is a promising strategy for cancer treatment. Enormous efforts have been done in the development of MEK1/2 inhibitors and encouraging advancements have been made, including four inhibitors approved for clinical use. However, due to the multifactorial property of cancer and rapidly arising drug resistance, the clinical efficacy of these MEK1/2 inhibitors as monotherapy are far from ideal. Several alternative strategies have been developed to improve the limited clinical efficacy, including the dual inhibitor which is a single drug molecule able to simultaneously inhibit two targets. In this review, we first introduced the activation and function of the MAPK/ERK1/2 components and discussed the advantages of MEK1/2-based dual inhibitors compared with the single inhibitors and combination therapy in the treatment of cancers. Then, we overviewed the MEK1/2-based dual inhibitors for the treatment of cancers and highlighted the theoretical basis of concurrent inhibition of MEK1/2 and other targets for development of these dual inhibitors. Besides, the status and results of these dual inhibitors in both preclinical and clinical studies were also the focus of this review.
Collapse
Affiliation(s)
- Hao Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation of Henan Province, Zhengzhou 450001, China
| | - Lingling Chi
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation of Henan Province, Zhengzhou 450001, China
| | - Fuqiang Yu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation of Henan Province, Zhengzhou 450001, China
| | - Hongling Dai
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation of Henan Province, Zhengzhou 450001, China
| | - Xiaojie Si
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation of Henan Province, Zhengzhou 450001, China
| | - Chao Gao
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation of Henan Province, Zhengzhou 450001, China
| | - Zhengjie Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation of Henan Province, Zhengzhou 450001, China
| | - Limin Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation of Henan Province, Zhengzhou 450001, China
| | - Jiaxin Zheng
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation of Henan Province, Zhengzhou 450001, China
| | - Yu Ke
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation of Henan Province, Zhengzhou 450001, China.
| | - Hongmin Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation of Henan Province, Zhengzhou 450001, China; State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou 450052, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou 450001, China.
| | - Qiurong Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation of Henan Province, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou 450001, China.
| |
Collapse
|
20
|
Cheng P, Zhang X, Wang X, Liu C, Zhao X, Fan J, Xu C. Identification of evodiamine as a suppressor of prostate cancer progression by reducing AR transcriptional activity via targeting Src. Endocrine 2022; 75:635-645. [PMID: 34713388 DOI: 10.1007/s12020-021-02907-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 10/10/2021] [Indexed: 12/24/2022]
Abstract
Evodiamine (EVO) is a bioactive alkaloid that exerts antitumor activity in various cancers, including prostate cancer (PCa). In this paper, we further investigated the molecular mechanisms underlying the anti-PCa effect of evodiamine. In the present study, cell proliferation, colony formation, migration, and invasion were assessed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), colony formation, and transwell assays, respectively. Animal studies were used to evaluate the effect of evodiamine on the tumorigenicity of LNCaP cells in vivo. The expression levels of steroid receptor coactivator (Src), androgene receptor (AR), and prostate-specific antigen (PSA) were detected by western blot, quantitative real-time PCR (qRT-PCR) or ELISA assay. Association between Src and AR was examined by Co-Immunoprecipitation (CoIP). The impact of evodiamine on AR-mediated transcriptional activity was confirmed by dual-luciferase reporter assay. The results showed that evodiamine reduced LNCaP and 22Rv1 cell proliferation, colony formation, migration, and invasion induced by dihydrotestosterone (DHT) in vitro, as well as diminished tumor growth in vivo. Mechanistically, evodiamine directly targeted Src and reduced DHT-induced Src activation. Moreover, the restoration of Src activation abolished evodiamine-mediated suppression of proliferation, migration, and invasion of DHT-treated LNCaP and 22Rv1 cells. Furthermore, evodiamine inhibited DHT-induced AR transcriptional activity through targeting Src. As a conclusion, our findings demonstrate the antitumor property of evodiamine in PCa by blocking AR transcriptional activity through targeting Src and provide a rationale for developing evodiamine as a promising antitumor agent against PCa.
Collapse
Affiliation(s)
- Pei Cheng
- Department of Urinary Surgery, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, PR China
| | - Xiaofan Zhang
- Department of Urinary Surgery, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, PR China
| | - Xiaofu Wang
- Department of Urinary Surgery, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, PR China
| | - Changwei Liu
- Department of Urinary Surgery, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, PR China
| | - Xinghua Zhao
- Department of Urinary Surgery, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, PR China
| | - Junfang Fan
- Department of Urinary Surgery, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, PR China
| | - Changbao Xu
- Department of Urinary Surgery, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, PR China.
| |
Collapse
|
21
|
Garcia-Mayea Y, Mir C, Carballo L, Sánchez-García A, Bataller M, LLeonart ME. TSPAN1, a novel tetraspanin member highly involved in carcinogenesis and chemoresistance. Biochim Biophys Acta Rev Cancer 2021; 1877:188674. [PMID: 34979155 DOI: 10.1016/j.bbcan.2021.188674] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/22/2021] [Accepted: 12/27/2021] [Indexed: 12/11/2022]
Abstract
The tetraspanin (TSPAN) family constitutes a poorly explored family of membrane receptors involved in various physiological processes, with relevant roles in anchoring multiple proteins, acting as scaffolding proteins, and cell signaling. Recent studies have increasingly demonstrated the involvement of TSPANs in cancer. In particular, tetraspanin 1 (also known as TSPAN1, NET-1, TM4C, C4.8 or GEF) has been implicated in cell survival, proliferation and invasion. Recently, our laboratory revealed a key role of TSPAN1 in the acquired resistance of tumor cells to conventional chemotherapy (e.g., cisplatin). In this review, we summarize and discuss the latest research on the physiological mechanisms of TSPANs in cancer and, in particular, on TSPAN1 regulating resistance to chemotherapy. A model of TSPAN1 action is proposed, and the potential of targeting TSPAN1 in anticancer therapeutic strategies is discussed.
Collapse
Affiliation(s)
- Yoelsis Garcia-Mayea
- Biomedical Research in Cancer Stem Cells Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Cristina Mir
- Biomedical Research in Cancer Stem Cells Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Laia Carballo
- Biomedical Research in Cancer Stem Cells Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Almudena Sánchez-García
- Biomedical Research in Cancer Stem Cells Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Marina Bataller
- Biomedical Research in Cancer Stem Cells Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Matilde E LLeonart
- Biomedical Research in Cancer Stem Cells Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain; Spanish Biomedical Research Network Center in Oncology, CIBERONC, Spain.
| |
Collapse
|
22
|
Protein Tyrosine Phosphatases: Mechanisms in Cancer. Int J Mol Sci 2021; 22:ijms222312865. [PMID: 34884670 PMCID: PMC8657787 DOI: 10.3390/ijms222312865] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 11/22/2021] [Accepted: 11/24/2021] [Indexed: 12/12/2022] Open
Abstract
Protein tyrosine kinases, especially receptor tyrosine kinases, have dominated the cancer therapeutics sphere as proteins that can be inhibited to selectively target cancer. However, protein tyrosine phosphatases (PTPs) are also an emerging target. Though historically known as negative regulators of the oncogenic tyrosine kinases, PTPs are now known to be both tumor-suppressive and oncogenic. This review will highlight key protein tyrosine phosphatases that have been thoroughly investigated in various cancers. Furthermore, the different mechanisms underlying pro-cancerous and anti-cancerous PTPs will also be explored.
Collapse
|
23
|
Mir MA, Qayoom H, Mehraj U, Nisar S, Bhat B, Wani NA. Targeting Different Pathways Using Novel Combination Therapy in Triple Negative Breast Cancer. Curr Cancer Drug Targets 2021; 20:586-602. [PMID: 32418525 DOI: 10.2174/1570163817666200518081955] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 03/22/2020] [Accepted: 03/24/2020] [Indexed: 02/07/2023]
Abstract
Triple negative breast cancer (TNBC) is one of the most aggressive subtypes of breast cancer accounting for 15-20% of cases and is defined by the lack of hormonal receptors viz., estrogen receptor (ER), progesterone receptor (PR) and expression of human epidermal growth receptor 2 (HER2). Treatment of TNBC is more challenging than other subtypes of breast cancer due to the lack of markers for the molecularly targeted therapies (ER, PR, and HER-2/ Neu), the conventional chemotherapeutic agents are still the mainstay of the therapeutic protocols of its patients. Despite, TNBC being more chemo-responsive than other subtypes, unfortunately, the initial good response to the chemotherapy eventually turns into a refractory drug-resistance. Using a monotherapy for the treatment of cancer, especially high-grade tumors like TNBC, is mostly worthless due to the inherent genetic instability of tumor cells to develop intrinsic and acquired resistance. Thus, a cocktail of two or more drugs with different mechanisms of action is more effective and could successfully control the disease. Furthermore, combination therapy reveals more, or at least the same, effectiveness with lower doses of every single agent and decreases the likelihood of chemoresistance. Herein, we shed light on the novel combinatorial approaches targeting PARP, EGFR, PI3K pathway, AR, and wnt signaling, HDAC, MEK pathway for efficient treatment of high-grade tumors like TNBC and decreasing the onset of resistance.
Collapse
Affiliation(s)
- Manzoor A Mir
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar, India
| | - Hina Qayoom
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar, India
| | - Umar Mehraj
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar, India
| | - Safura Nisar
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar, India
| | - Basharat Bhat
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar, India
| | - Nissar A Wani
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar, India
| |
Collapse
|
24
|
Li ZY, Sun XY. Recent progress in SRC targeted therapy for pancreatic cancer. Shijie Huaren Xiaohua Zazhi 2021; 29:621-627. [DOI: 10.11569/wcjd.v29.i12.621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer (PC) is a highly lethal malignancy with a 5-year survival rate of only 10% and is extremely resistant to chemotherapy. Therefore, developing effective therapeutic drugs is urgently needed. SRC is a proto-oncogenic tyrosine protein kinase and highly expressed in more than 70% of PCs. SRC is involved in regulating the proliferation, infiltration, and metastasis of PC cells as well as tumor angiogenesis, thus representing one of the most promising molecular targets for developing novel drugs. Preclinical studies demonstrate that small-molecule SRC inhibitors display significant anti-cancer activities in vitro and in vivo, and have a synergistic effect with conventional chemotherapy drugs against PC. Some SRC inhibitors have been evaluated in clinical trials. This article analyzes the regulatory mechanism of SRC and the recent progress and problems in developing drugs targeting SRC for the treatment of PC. Unfortunately, up to date no SRC inhibitor or regiment containing SRC inhibitors has been approved for the clinical treatment of PC. In the authors' opinion, the introduction of precision medicine principles to carry out SRC inhibitor clinical trials, combination of immunotherapy and SRC inhibitors, seeking more selective and effective SRC inhibitors, and further exploration of the SRC regulatory network may be the future directions for developing SRC-targeted therapies against PC.
Collapse
Affiliation(s)
- Zi-Yi Li
- Hepatosplenic Surgery Center, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Xue-Ying Sun
- Hepatosplenic Surgery Center, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| |
Collapse
|
25
|
Close DA, Kirkwood JM, Fecek RJ, Storkus WJ, Johnston PA. Unbiased High-Throughput Drug Combination Pilot Screening Identifies Synergistic Drug Combinations Effective against Patient-Derived and Drug-Resistant Melanoma Cell Lines. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2021; 26:712-729. [PMID: 33208016 PMCID: PMC8128935 DOI: 10.1177/2472555220970917] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
We describe the development, optimization, and validation of 384-well growth inhibition assays for six patient-derived melanoma cell lines (PDMCLs), three wild type (WT) for BRAF and three with V600E-BRAF mutations. We conducted a pilot drug combination (DC) high-throughput screening (HTS) of 45 pairwise 4×4 DC matrices prepared from 10 drugs in the PDMCL assays: two B-Raf inhibitors (BRAFi), a MEK inhibitor (MEKi), and a methylation agent approved for melanoma; cytotoxic topoisomerase II and DNA methyltransferase chemotherapies; and drugs targeting the base excision DNA repair enzyme APE1 (apurinic/apyrimidinic endonuclease-1/redox effector factor-1), SRC family tyrosine kinases, the heat shock protein 90 (HSP90) molecular chaperone, and histone deacetylases.Pairwise DCs between dasatinib and three drugs approved for melanoma therapy-dabrafenib, vemurafenib, or trametinib-were flagged as synergistic in PDMCLs. Exposure to fixed DC ratios of the SRC inhibitor dasatinib with the BRAFis or MEKis interacted synergistically to increase PDMCL sensitivity to growth inhibition and enhance cytotoxicity independently of PDMCL BRAF status. These DCs synergistically inhibited the growth of mouse melanoma cell lines that either were dabrafenib-sensitive or had acquired resistance to dabrafenib with cross resistance to vemurafenib, trametinib, and dasatinib. Dasatinib DCs with dabrafenib, vemurafenib, or trametinib activated apoptosis and increased cell death in melanoma cells independently of their BRAF status or their drug resistance phenotypes. These preclinical in vitro studies provide a data-driven rationale for the further investigation of DCs between dasatinib and BRAFis or MEKis as candidates for melanoma combination therapies with the potential to improve outcomes and/or prevent or delay the emergence of disease resistance.
Collapse
Affiliation(s)
- David A. Close
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - John M. Kirkwood
- Departments of Medicine, Dermatology, Translational Science, and Melanoma and Skin Cancer Program University of Pittsburgh School of Medicine, Pittsburgh, PA 15232, USA
- University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, PA 15232, USA
| | - Ronald J. Fecek
- Department of Microbiology, Lake Erie College of Osteopathic Medicine at Seton Hill, Greensburg, PA 15601, USA
| | - Walter J. Storkus
- University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, PA 15232, USA
- Departments of Dermatology, Immunology, Bioengineering and Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Paul A. Johnston
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
- University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, PA 15232, USA
| |
Collapse
|
26
|
Bhuyan F, de Jesus AA, Mitchell J, Leikina E, VanTries R, Herzog R, Onel KB, Oler A, Montealegre Sanchez GA, Johnson KA, Bichell L, Marrero B, De Castro LF, Huang Y, Calvo KR, Collins MT, Ganesan S, Chernomordik LV, Ferguson PJ, Goldbach-Mansky R. Novel Majeed Syndrome-Causing LPIN2 Mutations Link Bone Inflammation to Inflammatory M2 Macrophages and Accelerated Osteoclastogenesis. Arthritis Rheumatol 2021; 73:1021-1032. [PMID: 33314777 PMCID: PMC8252456 DOI: 10.1002/art.41624] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 12/10/2020] [Indexed: 12/29/2022]
Abstract
Objective To identify novel heterozygous LPIN2 mutations in a patient with Majeed syndrome and characterize the pathomechanisms that lead to the development of sterile osteomyelitis. Methods Targeted genetic analysis and functional studies assessing monocyte responses, macrophage differentiation, and osteoclastogenesis were conducted to compare the pathogenesis of Majeed syndrome to interleukin‐1 (IL‐1)–mediated diseases including neonatal‐onset multisystem inflammatory disease (NOMID) and deficiency of the IL‐1 receptor antagonist (DIRA). Results A 4‐year‐old girl of mixed ethnic background presented with sterile osteomyelitis and elevated acute‐phase reactants. She had a 17.8‐kb deletion on the maternal LPIN2 allele and a splice site mutation, p.R517H, that variably spliced out exons 10 and 11 on the paternal LPIN2 allele. The patient achieved long‐lasting remission receiving IL‐1 blockade with canakinumab. Compared to controls, monocytes and monocyte‐derived M1‐like macrophages from the patient with Majeed syndrome and those with NOMID or DIRA had elevated caspase 1 activity and IL‐1β secretion. In contrast, lipopolysaccharide‐stimulated, monocyte‐derived, M2‐like macrophages from the patient with Majeed syndrome released higher levels of osteoclastogenic mediators (IL‐8, IL‐6, tumor necrosis factor, CCL2, macrophage inflammatory protein 1α/β, CXCL8, and CXCL1) compared to NOMID patients and healthy controls. Accelerated osteoclastogenesis in the patient with Majeed syndrome was associated with higher NFATc1 levels, enhanced JNK/MAPK, and reduced Src kinase activation, and partially responded to JNK inhibition and IL‐1 (but not IL‐6) blockade. Conclusion We report 2 novel compound heterozygous disease‐causing mutations in LPIN2 in an American patient with Majeed syndrome. LPIN2 deficiency drives differentiation of proinflammatory M2‐like macrophages and enhances intrinsic osteoclastogenesis. This provides a model for the pathogenesis of sterile osteomyelitis which differentiates Majeed syndrome from other IL‐1–mediated autoinflammatory diseases.
Collapse
Affiliation(s)
- Farzana Bhuyan
- National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland
| | - Adriana A de Jesus
- National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland
| | - Jacob Mitchell
- National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland
| | - Evgenia Leikina
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland
| | - Rachel VanTries
- National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland
| | | | | | - Andrew Oler
- National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland
| | | | - Kim A Johnson
- National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland
| | - Lena Bichell
- National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland
| | - Bernadette Marrero
- National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland
| | | | - Yan Huang
- National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland
| | | | - Michael T Collins
- National Institute of Dental and Craniofacial Research, NIH, Bethesda, Maryland
| | - Sundar Ganesan
- National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland
| | - Leonid V Chernomordik
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland
| | | | | |
Collapse
|
27
|
Mayoral-Varo V, Sánchez-Bailón MP, Calcabrini A, García-Hernández M, Frezza V, Martín ME, González VM, Martín-Pérez J. The Relevance of the SH2 Domain for c-Src Functionality in Triple-Negative Breast Cancer Cells. Cancers (Basel) 2021; 13:cancers13030462. [PMID: 33530373 PMCID: PMC7865352 DOI: 10.3390/cancers13030462] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/15/2021] [Accepted: 01/19/2021] [Indexed: 02/05/2023] Open
Abstract
The role of Src family kinases (SFKs) in human tumors has been always associated with tyrosine kinase activity and much less attention has been given to the SH2 and SH3 adapter domains. Here, we studied the role of the c-Src-SH2 domain in triple-negative breast cancer (TNBC). To this end, SUM159PT and MDA-MB-231 human cell lines were employed as model systems. These cells conditionally expressed, under tetracycline control (Tet-On system), a c-Src variant with point-inactivating mutation of the SH2 adapter domain (R175L). The expression of this mutant reduced the self-renewal capability of the enriched population of breast cancer stem cells (BCSCs), demonstrating the importance of the SH2 adapter domain of c-Src in the mammary gland carcinogenesis. In addition, the analysis of anchorage-independent growth, proliferation, migration, and invasiveness, all processes associated with tumorigenesis, showed that the SH2 domain of c-Src plays a very relevant role in their regulation. Furthermore, the transfection of two different aptamers directed to SH2-c-Src in both SUM159PT and MDA-MB-231 cells induced inhibition of their proliferation, migration, and invasiveness, strengthening the hypothesis that this domain is highly involved in TNBC tumorigenesis. Therefore, the SH2 domain of c-Src could be a promising therapeutic target and combined treatments with inhibitors of c-Src kinase enzymatic activity may represent a new therapeutic strategy for patients with TNBC, whose prognosis is currently very negative.
Collapse
Affiliation(s)
- Víctor Mayoral-Varo
- Instituto de Investigaciones Biomédicas A, Sols/Dpto. Bioquímica (CSIC/UAM), Arturo Duperier 4, 28029 Madrid, Spain; (V.M.-V.); (M.P.S.-B.); (A.C.)
| | - María Pilar Sánchez-Bailón
- Instituto de Investigaciones Biomédicas A, Sols/Dpto. Bioquímica (CSIC/UAM), Arturo Duperier 4, 28029 Madrid, Spain; (V.M.-V.); (M.P.S.-B.); (A.C.)
- Max Delbrück Center for Molecular Medicine (MDC), Robert-Rössle-Str. 10, 13092 Berlin, Germany
| | - Annarica Calcabrini
- Instituto de Investigaciones Biomédicas A, Sols/Dpto. Bioquímica (CSIC/UAM), Arturo Duperier 4, 28029 Madrid, Spain; (V.M.-V.); (M.P.S.-B.); (A.C.)
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Marta García-Hernández
- Grupo de Aptámeros, Servicio Bioquímica-Investigación, IRYCIS-Hospital Ramón y Cajal. Ctra. Colmenar Viejo km 9100, 28034 Madrid, Spain; (M.G.-H.); (V.F.); (M.E.M.); (V.M.G.)
| | - Valerio Frezza
- Grupo de Aptámeros, Servicio Bioquímica-Investigación, IRYCIS-Hospital Ramón y Cajal. Ctra. Colmenar Viejo km 9100, 28034 Madrid, Spain; (M.G.-H.); (V.F.); (M.E.M.); (V.M.G.)
| | - María Elena Martín
- Grupo de Aptámeros, Servicio Bioquímica-Investigación, IRYCIS-Hospital Ramón y Cajal. Ctra. Colmenar Viejo km 9100, 28034 Madrid, Spain; (M.G.-H.); (V.F.); (M.E.M.); (V.M.G.)
| | - Víctor M. González
- Grupo de Aptámeros, Servicio Bioquímica-Investigación, IRYCIS-Hospital Ramón y Cajal. Ctra. Colmenar Viejo km 9100, 28034 Madrid, Spain; (M.G.-H.); (V.F.); (M.E.M.); (V.M.G.)
| | - Jorge Martín-Pérez
- Instituto de Investigaciones Biomédicas A, Sols/Dpto. Bioquímica (CSIC/UAM), Arturo Duperier 4, 28029 Madrid, Spain; (V.M.-V.); (M.P.S.-B.); (A.C.)
- Instituto de Investigaciones Sanitarias del Hospital La Paz (IdiPAZ), Paseo de la Castellana 261, 28046 Madrid, Spain
- Correspondence: or ; Tel.: +34-91-585-4416; Fax: +34-91-585-4401
| |
Collapse
|
28
|
Luo Y, Hu J, Liu Y, Li L, Li Y, Sun B, Kong R. Invadopodia: A potential target for pancreatic cancer therapy. Crit Rev Oncol Hematol 2021; 159:103236. [PMID: 33482351 DOI: 10.1016/j.critrevonc.2021.103236] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 01/05/2021] [Accepted: 01/16/2021] [Indexed: 02/08/2023] Open
Abstract
Dissemination of cancer cells is an intricate multistep process that represents the most deadly aspect of cancer. Cancer cells form F-actin-rich protrusions known as invadopodia to invade surrounding tissues, blood vessels and lymphatics. A number of studies have demonstrated the significant roles of invadopodia in cancer. Therefore, the specific cells and molecules involved in invadopodia activity can provide as therapeutic targets. In this review, we included a thorough overview of studies in invadopodia and discussed their functions in cancer metastasis. We then presented the specific cells and molecules involved in invadopodia activity in pancreatic cancer and analyzed their suitability to be effective therapeutic targets. Currently, drugs targeting invadopodia and relevant clinical trials are negligible. Here, we highlighted the significance of potential drugs and discussed future obstacles in implementing clinical trials. This review presents a new perspective on invadopodia-induced pancreatic cancer metastasis and may prosper the development of targeted therapeutics against pancreatic cancer.
Collapse
Affiliation(s)
- Yan Luo
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jisheng Hu
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yong Liu
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Le Li
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yilong Li
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bei Sun
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Rui Kong
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
29
|
Yuan G, Yao M, Lv H, Jia X, Chen J, Xue J. Novel Targeted Photosensitizer as an Immunomodulator for Highly Efficient Therapy of T-Cell Acute Lymphoblastic Leukemia. J Med Chem 2020; 63:15655-15667. [PMID: 33300796 DOI: 10.1021/acs.jmedchem.0c01072] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Dasatinib is a kinase-targeted drug used in the treatment of leukemia. Regrettably, it remains far from optimal medicine due to insurmountable drug resistance and side effects. Photodynamic therapy (PDT) has proven that it can induce systemic immune responses. However, conventional photosensitizers as immunomodulators produce anticancer immunities, which are inadequate to eliminate residual cancer cells. Herein, a novel compound 4 was synthesized and investigated, which introduces dasatinib and zinc(II) phthalocyanine as the targeting and photodynamic moiety, respectively. Compound 4 exhibits a high affinity to CCRF-CEM cells/tumor tissues, which overexpress lymphocyte-specific protein tyrosine kinase (LCK), and preferential elimination from the body. Meanwhile, compound 4 shows excellent photocytotoxicity and tumor regression. Significantly, compound 4-induced PDT can obviously enhance immune responses, resulting in the production of more immune cells. We believe that the proposed manner is a potential strategy for the treatment of T-cell acute lymphoblastic leukemia.
Collapse
Affiliation(s)
- Gankun Yuan
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, 2 Xueyuan Road, University Town, Fuzhou 350116, Fujian, P. R. China
| | - Mengyu Yao
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, 2 Xueyuan Road, University Town, Fuzhou 350116, Fujian, P. R. China
| | - Huihui Lv
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, 2 Xueyuan Road, University Town, Fuzhou 350116, Fujian, P. R. China
| | - Xiao Jia
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, 2 Xueyuan Road, University Town, Fuzhou 350116, Fujian, P. R. China
| | - Juanjuan Chen
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, 2 Xueyuan Road, University Town, Fuzhou 350116, Fujian, P. R. China
| | - Jinping Xue
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, 2 Xueyuan Road, University Town, Fuzhou 350116, Fujian, P. R. China
| |
Collapse
|
30
|
Maloney SM, Hoover CA, Morejon-Lasso LV, Prosperi JR. Mechanisms of Taxane Resistance. Cancers (Basel) 2020; 12:E3323. [PMID: 33182737 PMCID: PMC7697134 DOI: 10.3390/cancers12113323] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 10/30/2020] [Accepted: 11/06/2020] [Indexed: 12/17/2022] Open
Abstract
The taxane family of chemotherapy drugs has been used to treat a variety of mostly epithelial-derived tumors and remain the first-line treatment for some cancers. Despite the improved survival time and reduction of tumor size observed in some patients, many have no response to the drugs or develop resistance over time. Taxane resistance is multi-faceted and involves multiple pathways in proliferation, apoptosis, metabolism, and the transport of foreign substances. In this review, we dive deeper into hypothesized resistance mechanisms from research during the last decade, with a focus on the cancer types that use taxanes as first-line treatment but frequently develop resistance to them. Furthermore, we will discuss current clinical inhibitors and those yet to be approved that target key pathways or proteins and aim to reverse resistance in combination with taxanes or individually. Lastly, we will highlight taxane response biomarkers, specific genes with monitored expression and correlated with response to taxanes, mentioning those currently being used and those that should be adopted. The future directions of taxanes involve more personalized approaches to treatment by tailoring drug-inhibitor combinations or alternatives depending on levels of resistance biomarkers. We hope that this review will identify gaps in knowledge surrounding taxane resistance that future research or clinical trials can overcome.
Collapse
Affiliation(s)
- Sara M. Maloney
- Harper Cancer Research Institute, South Bend, IN 46617, USA;
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, South Bend, IN 46617, USA
| | - Camden A. Hoover
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA; (C.A.H.); (L.V.M.-L.)
| | - Lorena V. Morejon-Lasso
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA; (C.A.H.); (L.V.M.-L.)
| | - Jenifer R. Prosperi
- Harper Cancer Research Institute, South Bend, IN 46617, USA;
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, South Bend, IN 46617, USA
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA; (C.A.H.); (L.V.M.-L.)
| |
Collapse
|
31
|
Exploration in the Mechanism of Kaempferol for the Treatment of Gastric Cancer Based on Network Pharmacology. BIOMED RESEARCH INTERNATIONAL 2020; 2020:5891016. [PMID: 33145355 PMCID: PMC7596434 DOI: 10.1155/2020/5891016] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 09/07/2020] [Accepted: 10/09/2020] [Indexed: 12/11/2022]
Abstract
Background Kaempferol is a natural polyphenol in lots of Chinese herbs, which has shown promising treatment for gastric cancer (GC). However, the molecular mechanisms of its action have not been systematically revealed yet. In this work, a network pharmacology approach was used to elucidate the potential mechanisms of kaempferol in the treatment of GC. Methods The kaempferol was input into the PharmMapper and SwissTargetPrediction database to get its targets, and the targets of GC were obtained by retrieving the Online Mendelian Inheritance in Man (OMIM) database, MalaCards database, Therapeutic Target Database (TTD), and Coolgen database. The molecular docking was performed to assess the interactions between kaempferol and these targets. Next, the overlap targets of kaempferol and GC were identified for GO and KEGG enrichment analyses. Afterward, a protein-protein interaction (PPI) network was constructed to get the hub targets, and the expression and overall survival analysis of the hub target were investigated. Finally, the overall survival (OS) analysis of hub targets was performed using the Kaplan-Meier Plotter online tool. Results A total of 990 genes related to GC and 10 overlapping genes were determined through matching the 24 potential targets of kaempferol with disease-associated genes. The result of molecular docking indicated that kaempferol can bind with these hub targets with good binding scores. These targets were further mapped to 140 GO biological process terms and 11 remarkable pathways. In the PPI network analysis, 3 key targets were identified, including ESR1, EGFR, and SRC. The mRNA and protein expression levels of EGFR and SRC were obviously higher in GC tissues. High expression of these targets was related to poor OS in GC patients. Conclusions This study provided a novel approach to reveal the therapeutic mechanisms of kaempferol on GC, which will ease the future clinical application of kaempferol in the treatment of GC.
Collapse
|
32
|
Zhang H, Shen Z, Zhou Y, Zhang Z, Wang Q, Zhang M, Jiang K, Wang S, Ye Y, Wang B. Downregulation of miR-654-3p in Colorectal Cancer Indicates Poor Prognosis and Promotes Cell Proliferation and Invasion by Targeting SRC. Front Genet 2020; 11:577948. [PMID: 33193697 PMCID: PMC7554538 DOI: 10.3389/fgene.2020.577948] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 09/10/2020] [Indexed: 12/21/2022] Open
Abstract
Background MicroRNAs (miRNAs), such as miR-654-3p, regulate gene expression at the post-transcriptional level affecting malignant tumor behavior. However, the expression levels, function, and mechanism of miR-654-3p in colorectal cancer (CRC) are unknown. Methods The expression levels of miR-654-3p and SRC in 103 CRC tissues and matched normal colorectal tissues were detected by a quantitative real-time polymerase chain reaction (qRT-PCR). miR-654-3p was overexpressed by RNA mimics and SRC knockdown by siRNA. Function-based experiments were carried out to detect the proliferation and migration abilities in CRC cell lines. Flow cytometry assay was performed to evaluate the effect of miR-654-3p on cell apoptosis and cycle distribution. Xenograft tumor models in nude mice were utilized to evaluate miR-654-3p functions in vivo. Dual-fluorescence reporter assay was used to verify the direct binding between miR-654-3p and SRC. Results miR-654-3p was downregulated in CRC tissues as compared to matched normal colorectal tissues. The expression levels of miR-654-3p were closely associated with distant metastasis. In addition, elevated expression of miR-654-3p in CRC patients prolonged the overall survival. Upregulated miR-654-3p significantly suppressed the proliferation and migration capacity of CRC cells by enhancing apoptosis and promoting G0/G1 phase arrest. The direct binding between miR-654-3p and SRC was verified by the dual-luciferase reporter gene. Furthermore, the suppression of proliferation and migration capacity by elevated miR-654-3p level could be reversed by overexpressing SRC. Conclusion miR-654-3p acts as a tumor suppressor through regulating SRC. It might also serve as a diagnostic and prognostic indicator and a novel molecular target for CRC therapy.
Collapse
Affiliation(s)
- Haoran Zhang
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, China
| | - Zhanlong Shen
- Laboratory of Surgical Oncology, Peking University People's Hospital, Beijing, China.,Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Beijing, China
| | - Yushi Zhou
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, China
| | - Zhen Zhang
- Laboratory of Surgical Oncology, Peking University People's Hospital, Beijing, China
| | - Quan Wang
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, China
| | - Mengmeng Zhang
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, China
| | - Kewei Jiang
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, China
| | - Shan Wang
- Laboratory of Surgical Oncology, Peking University People's Hospital, Beijing, China.,Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Beijing, China
| | - Yingjiang Ye
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, China
| | - Bo Wang
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, China.,Laboratory of Surgical Oncology, Peking University People's Hospital, Beijing, China
| |
Collapse
|
33
|
Systematic Elucidation of the Mechanism of Quercetin against Gastric Cancer via Network Pharmacology Approach. BIOMED RESEARCH INTERNATIONAL 2020; 2020:3860213. [PMID: 32964029 PMCID: PMC7486643 DOI: 10.1155/2020/3860213] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 06/25/2020] [Indexed: 12/24/2022]
Abstract
This study was aimed at elucidating the potential mechanisms of quercetin in the treatment of gastric cancer (GC). A network pharmacology approach was used to analyze the targets and pathways of quercetin in treating GC. The predicted targets of quercetin against GC were obtained through database mining, and the correlation of these targets with GC was analyzed by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses. Next, the protein-protein interaction (PPI) network was constructed, and overall survival (OS) analysis of hub targets was performed using the Kaplan–Meier Plotter online tool. Finally, the mechanism was further analyzed via molecular docking of quercetin with the hub targets. Thirty-six quercetin-related genes were identified, 15 of which overlapped with GC-related targets. These targets were further mapped to 319 GO biological process terms and 10 remarkable pathways. In the PPI network analysis, six hub targets were identified, including AKT1, EGFR, SRC, IGF1R, PTK2, and KDR. The high expression of these targets was related to poor OS in GC patients. Molecular docking analysis confirmed that quercetin can bind to these hub targets. In conclusion, this study provided a novel approach to reveal the therapeutic mechanisms of quercetin on GC, which will ease the future clinical application of quercetin in the treatment of GC.
Collapse
|
34
|
Three-Dimensional Interactions Analysis of the Anticancer Target c-Src Kinase with Its Inhibitors. Cancers (Basel) 2020; 12:cancers12082327. [PMID: 32824733 PMCID: PMC7466017 DOI: 10.3390/cancers12082327] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 08/07/2020] [Accepted: 08/16/2020] [Indexed: 12/13/2022] Open
Abstract
Src family kinases (SFKs) constitute the biggest family of non-receptor tyrosine kinases considered as therapeutic targets for cancer therapy. An aberrant expression and/or activation of the proto-oncogene c-Src kinase, which is the oldest and most studied member of the family, has long been demonstrated to play a major role in the development, growth, progression and metastasis of numerous human cancers, including colon, breast, gastric, pancreatic, lung and brain carcinomas. For these reasons, the pharmacological inhibition of c-Src activity represents an effective anticancer strategy and a few compounds targeting c-Src, together with other kinases, have been approved as drugs for cancer therapy, while others are currently undergoing preclinical studies. Nevertheless, the development of potent and selective inhibitors of c-Src aimed at properly exploiting this biological target for the treatment of cancer still represents a growing field of study. In this review, the co-crystal structures of c-Src kinase in complex with inhibitors discovered in the past two decades have been described, highlighting the key ligand-protein interactions necessary to obtain high potency and the features to be exploited for addressing selectivity and drug resistance issues, thus providing useful information for the design of new and potent c-Src kinase inhibitors.
Collapse
|
35
|
Mayoral-Varo V, Calcabrini A, Sánchez-Bailón MP, Martínez-Costa ÓH, González-Páramos C, Ciordia S, Hardisson D, Aragón JJ, Fernández-Moreno MÁ, Martín-Pérez J. c-Src functionality controls self-renewal and glucose metabolism in MCF7 breast cancer stem cells. PLoS One 2020; 15:e0235850. [PMID: 32673341 PMCID: PMC7365443 DOI: 10.1371/journal.pone.0235850] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 06/23/2020] [Indexed: 06/11/2023] Open
Abstract
Deregulation of Src kinases is associated with cancer. We previously showed that SrcDN conditional expression in MCF7 cells reduces tumorigenesis and causes tumor regression in mice. However, it remained unclear whether SrcDN affected breast cancer stem cell functionality or it reduced tumor mass. Here, we address this question by isolating an enriched population of Breast Cancer Stem Cells (BCSCs) from MCF7 cells with inducible expression of SrcDN. Induction of SrcDN inhibited self-renewal, and stem-cell marker expression (Nanog, Oct3-4, ALDH1, CD44). Quantitative proteomic analyses of mammospheres from MCF7-Tet-On-SrcDN cells (data are available via ProteomeXchange with identifier PXD017789, project DOI: 10.6019/PXD017789) and subsequent GSEA showed that SrcDN expression inhibited glycolysis. Indeed, induction of SrcDN inhibited expression and activity of hexokinase, pyruvate kinase and lactate dehydrogenase, resulting in diminished glucose consumption and lactate production, which restricted Warburg effect. Thus, c-Src functionality is important for breast cancer stem cell maintenance and renewal, and stem cell transcription factor expression, effects linked to glucose metabolism reduction.
Collapse
Affiliation(s)
| | | | | | | | | | - Sergio Ciordia
- Servicio de Espectrometría de Masas, Centro Nacional de Biotecnología (CSIC), Madrid, Spain
| | - David Hardisson
- Servicio de Anatomía Patológica, Hospital Universitario La Paz, Madrid
- Departamento de Anatomía Patológica, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Instituto de investigaciones sanitarias del hospital La Paz (IdiPAZ), Madrid, Spain
| | - Juan J. Aragón
- Instituto de Investigaciones Biomédicas A. Sols (CSIC/UAM), Madrid, Spain
| | - Miguel Ángel Fernández-Moreno
- Instituto de Investigaciones Biomédicas A. Sols (CSIC/UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Raras (CIBERER), Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Jorge Martín-Pérez
- Instituto de Investigaciones Biomédicas A. Sols (CSIC/UAM), Madrid, Spain
- Instituto de investigaciones sanitarias del hospital La Paz (IdiPAZ), Madrid, Spain
| |
Collapse
|
36
|
Rainey L, Deevi RK, McClements J, Khawaja H, Watson CJ, Roudier M, Van Schaeybroeck S, Campbell FC. Fundamental control of grade-specific colorectal cancer morphology by Src regulation of ezrin-centrosome engagement. J Pathol 2020; 251:310-322. [PMID: 32315081 DOI: 10.1002/path.5452] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 02/27/2020] [Accepted: 04/07/2020] [Indexed: 11/11/2022]
Abstract
The phenotypic spectrum of colorectal cancer (CRC) is remarkably diverse, with seemingly endless variations in cell shape, mitotic figures and multicellular configurations. Despite this morphological complexity, histological grading of collective phenotype patterns provides robust prognostic stratification in CRC. Although mechanistic understanding is incomplete, previous studies have shown that the cortical protein ezrin controls diversification of cell shape, mitotic figure geometry and multicellular architecture, in 3D organotypic CRC cultures. Because ezrin is a substrate of Src tyrosine kinase that is frequently overexpressed in CRC, we investigated Src regulation of ezrin and morphogenic growth in 3D CRC cultures. Here we show that Src perturbations disrupt CRC epithelial spatial organisation. Aberrant Src activity suppresses formation of the cortical ezrin cap that anchors interphase centrosomes. In CRC cells with a normal centrosome number, these events lead to mitotic spindle misorientation, perturbation of cell cleavage, abnormal epithelial stratification, apical membrane misalignment, multilumen formation and evolution of cribriform multicellular morphology, a feature of low-grade cancer. In isogenic CRC cells with centrosome amplification, aberrant Src signalling promotes multipolar mitotic spindle formation, pleomorphism and morphological features of high-grade cancer. Translational studies in archival human CRC revealed associations between Src intensity, multipolar mitotic spindle frequency and high-grade cancer morphology. Collectively, our study reveals Src regulation of CRC morphogenic growth via ezrin-centrosome engagement and uncovers combined perturbations underlying transition to high-grade CRC morphology. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Lisa Rainey
- Centre for Cancer Research and Cell Biology, Queen's University Belfast and Belfast Health and Social Care Trust, Belfast, UK
| | - Ravi K Deevi
- Centre for Cancer Research and Cell Biology, Queen's University Belfast and Belfast Health and Social Care Trust, Belfast, UK
| | - Jane McClements
- Centre for Cancer Research and Cell Biology, Queen's University Belfast and Belfast Health and Social Care Trust, Belfast, UK
| | - Hajrah Khawaja
- Centre for Cancer Research and Cell Biology, Queen's University Belfast and Belfast Health and Social Care Trust, Belfast, UK
| | - Chris J Watson
- Wellcome Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Martine Roudier
- Molecular Pathology Laboratory, AstraZeneca Oncology Translational Science, Cambridge, UK
| | - Sandra Van Schaeybroeck
- Centre for Cancer Research and Cell Biology, Queen's University Belfast and Belfast Health and Social Care Trust, Belfast, UK
| | - Frederick C Campbell
- Centre for Cancer Research and Cell Biology, Queen's University Belfast and Belfast Health and Social Care Trust, Belfast, UK
| |
Collapse
|
37
|
Dasatinib prevents skeletal metastasis of osteotropic MDA-MB-231 cells in a xenograft mouse model. Arch Gynecol Obstet 2020; 301:1493-1502. [PMID: 32170411 DOI: 10.1007/s00404-020-05496-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 03/05/2020] [Indexed: 10/25/2022]
Abstract
PURPOSE Bone metastasis in breast cancer has been linked to activity of c-Src kinase, one of the extensively explored tyrosine kinases in cell biology. The impact of TNF-related apoptosis inducing ligand (TRAIL) and TRAIL receptors has just recently been integrated into this conception. METHODS An osteotropic clone of MDA-MB-231 cells simulated a model for bone metastasis of triple-negative breast cancer (TNBC). The effects of Dasatinib, a clinically established inhibitor of Src kinases family and Abl were evaluated in vitro and in vivo. In vivo effects of Dasatinib treatment on the occurrence of skeletal metastases were tested in a xenograft mouse model after intra-cardiac injection of osteotropic MDA-MB-231-cells. Ex vivo analyses of the bone sections confirmed intraosseous growth of metastases and allowed determination of osteoclastic activity. RESULTS Treatment of osteotropic MDA-MB-231 cells with Dasatinib inhibited proliferation rates in vitro. A shift in TRAIL-receptor expression towards an induction of oncogenic TRAIL-R2 was observed. In vivo, 15 of 30 mice received an intra-peritoneal treatment with Dasatinib. These mice showed significantly less skeletal metastases in bioluminescence scans. Moreover, a pronounced increase in bone volume was observed in the treatment group, as detected by µ-Computed Tomography. Dasatinib treatment also led to a greater increase in bone density in tibiae without metastatic affection, which was accompanied by reduced recruitment of osteoclasts. CONCLUSION Our observations support the concept of utilizing Dasatinib in targeting early-stage bone metastatic TNBC and sustaining bone health.
Collapse
|
38
|
Montenegro RC, Howarth A, Ceroni A, Fedele V, Farran B, Mesquita FP, Frejno M, Berger BT, Heinzlmeir S, Sailem HZ, Tesch R, Ebner D, Knapp S, Burbano R, Kuster B, Müller S. Identification of molecular targets for the targeted treatment of gastric cancer using dasatinib. Oncotarget 2020; 11:535-549. [PMID: 32082487 PMCID: PMC7007292 DOI: 10.18632/oncotarget.27462] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Accepted: 01/13/2020] [Indexed: 12/24/2022] Open
Abstract
Gastric cancer (GC) remains the third leading cause of cancer-related death despite several improvements in targeted therapy. There is therefore an urgent need to investigate new treatment strategies, including the identification of novel biomarkers for patient stratification. In this study, we evaluated the effect of FDA-approved kinase inhibitors on GC. Through a combination of cell growth, migration and invasion assays, we identified dasatinib as an efficient inhibitor of GC proliferation. Mass-spectrometry-based selectivity profiling and subsequent knockdown experiments identified members of the SRC family of kinases including SRC, FRK, LYN and YES, as well as other kinases such as DDR1, ABL2, SIK2, RIPK2, EPHA2, and EPHB2 as dasatinib targets. The expression levels of the identified kinases were investigated on RNA and protein level in 200 classified tumor samples from patients, who had undergone gastrectomy, but had received no treatment. Levels of FRK, DDR1 and SRC expression on both mRNA and protein level were significantly higher in metastatic patient samples regardless of the tumor stage, while expression levels of SIK2 correlated with tumor size. Collectively, our data suggest dasatinib for treatment of GC based on its unique property, inhibiting a small number of key kinases (SRC, FRK, DDR1 and SIK2), highly expressed in GC patients.
Collapse
Affiliation(s)
| | - Alison Howarth
- Novo Nordisk Research Centre Oxford (NNRCO), Discovery Technologies and Genomics, Oxford, UK
| | - Alessandro Ceroni
- Novo Nordisk Research Centre Oxford (NNRCO), Discovery Technologies and Genomics, Oxford, UK
| | - Vita Fedele
- Novo Nordisk Research Centre Oxford (NNRCO), Discovery Technologies and Genomics, Oxford, UK
| | - Batoul Farran
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Felipe Pantoja Mesquita
- Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE, Brazil
| | - Martin Frejno
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany
| | - Benedict-Tilman Berger
- Structural Genomics Consortium, Buchmann Institute for Life Sciences, Goethe-University Frankfurt, Frankfurt, Germany.,Institute of Pharmaceutical Chemistry, Goethe-University Frankfurt, Frankfurt, Germany
| | - Stephanie Heinzlmeir
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Heba Z Sailem
- Institute of Biomedical Engineering, Department of Engineering, University of Oxford, Oxford, UK.,Big Data Institute, University of Oxford, Li Ka Shing Centre for Health Information and Discovery, Old Road Campus Research Building, Oxford, UK
| | - Roberta Tesch
- Structural Genomics Consortium, Buchmann Institute for Life Sciences, Goethe-University Frankfurt, Frankfurt, Germany.,Institute of Pharmaceutical Chemistry, Goethe-University Frankfurt, Frankfurt, Germany
| | - Daniel Ebner
- Novo Nordisk Research Centre Oxford (NNRCO), Discovery Technologies and Genomics, Oxford, UK
| | - Stefan Knapp
- Structural Genomics Consortium, Buchmann Institute for Life Sciences, Goethe-University Frankfurt, Frankfurt, Germany.,Institute of Pharmaceutical Chemistry, Goethe-University Frankfurt, Frankfurt, Germany
| | | | - Bernhard Kuster
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Bavarian Center for Biomolecular Mass Spectrometry (BayBioMS), Technische Universität München, Freising, Germany
| | - Susanne Müller
- Structural Genomics Consortium, Buchmann Institute for Life Sciences, Goethe-University Frankfurt, Frankfurt, Germany
| |
Collapse
|
39
|
Nelson LJ, Wright HJ, Dinh NB, Nguyen KD, Razorenova OV, Heinemann FS. Src Kinase Is Biphosphorylated at Y416/Y527 and Activates the CUB-Domain Containing Protein 1/Protein Kinase C δ Pathway in a Subset of Triple-Negative Breast Cancers. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 190:484-502. [PMID: 31843498 DOI: 10.1016/j.ajpath.2019.10.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 09/20/2019] [Accepted: 10/15/2019] [Indexed: 01/07/2023]
Abstract
Targeted therapeutics are needed for triple-negative breast cancer (TNBC). In this study, we investigated the activation of Src family of cytoplasmic tyrosine kinases (SFKs) and two SFK substrates-CUB-domain containing protein 1 (CDCP1) and protein kinase C δ (PKCδ)-in 56 formalin-fixed, paraffin-embedded (FFPE) TNBCs. Expression of SFK phosphorylated at Y416 (SFK_pY416+) in tumor cells was strongly associated with phosphorylation of CDCP1 and PKCδ (CDCP1_ pY743+ and PKCδ_pY311+), as assessed by immunohistochemistry, indicating increased SFK activity in situ. To enable biochemical analysis, protein extraction from FFPE tissue was optimized. Cleaved CDCP1 isoform (70 kDa) was expressed to a varying degree in all samples but only phosphorylated in TNBC tumor cells that expressed SFK_pY416. Interestingly, active SFK was found to be biphosphorylated (SFK_pY416+/pY527+). Biphosphorylated active SFK was observed more frequently in forkhead box protein A1 (FOXA1)- TNBCs. In addition, in SFK_pY416- samples, FOXA1+ TNBC tended to be SFK_pY527+ (classic inactive SFK), and FOXA1- TNBC tended to be SFK_pY527- (SFK poised for activation). Strong SFK_pY416 staining was also observed in tumor-infiltrating lymphocytes in a subset of TNBCs with high tumor-infiltrating lymphocyte content. This report will facilitate protein biochemical analysis of FFPE tumor samples and justifies the development of therapies targeting the SFK/CDCP1/PKCδ pathway for TNBC treatment.
Collapse
Affiliation(s)
- Luke J Nelson
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, California
| | - Heather J Wright
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, California
| | - Nguyen B Dinh
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, California
| | - Kevin D Nguyen
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, California
| | - Olga V Razorenova
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, California.
| | - F Scott Heinemann
- Department of Pathology, Hoag Memorial Hospital Presbyterian, Newport Beach, California.
| |
Collapse
|
40
|
'Piperazining' the catalytic gatekeepers: unraveling the pan-inhibition of SRC kinases; LYN, FYN and BLK by masitinib. Future Med Chem 2019; 11:2365-2380. [PMID: 31516031 DOI: 10.4155/fmc-2018-0354] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Aim: Blocking oncogenic signaling of B-cell receptor (BCR) has been explored as a viable strategy in the treatment of diffuse large B-cell lymphoma. Masitinib is shown to multitarget LYN, FYN and BLK kinases that propagate BCR signals to downstream effectors. However, the molecular mechanisms of its selectivity and pan-inhibition remain elusive. Materials & methods: This study therefore employed molecular dynamics simulations coupled with advanced post-molecular dynamics simulation techniques to unravel the structural mechanisms that inform the reported multitargeting ability of masitinib. Results: Molecular dynamics simulations revealed initial selective targeting of catalytic residues (Asp334/Glu335 - LYN; Asp130/Asp148/Glu54 - FYN; Asp89 - BLK) by masitinib, with high-affinity interactions via its piperazine ring at the entrance of the ATP-binding pockets, before systematic access into the hydrophobic deep pocket grooves. Conclusion: Identification of these 'gatekeeper' residues could open up a novel paradigm of structure-based design of highly selective pan-inhibitors of BCR signaling in the treatment of diffuse large B-cell lymphoma.
Collapse
|
41
|
Parkin A, Man J, Timpson P, Pajic M. Targeting the complexity of Src signalling in the tumour microenvironment of pancreatic cancer: from mechanism to therapy. FEBS J 2019; 286:3510-3539. [PMID: 31330086 PMCID: PMC6771888 DOI: 10.1111/febs.15011] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Revised: 05/26/2019] [Accepted: 07/19/2019] [Indexed: 02/06/2023]
Abstract
Pancreatic cancer, a disease with extremely poor prognosis, has been notoriously resistant to virtually all forms of treatment. The dynamic crosstalk that occurs between tumour cells and the surrounding stroma, frequently mediated by intricate Src/FAK signalling, is increasingly recognised as a key player in pancreatic tumourigenesis, disease progression and therapeutic resistance. These important cues are fundamental for defining the invasive potential of pancreatic tumours, and several components of the Src and downstream effector signalling have been proposed as potent anticancer therapeutic targets. Consequently, numerous agents that block this complex network are being extensively investigated as potential antiinvasive and antimetastatic therapeutic agents for this disease. In this review, we will discuss the latest evidence of Src signalling in PDAC progression, fibrotic response and resistance to therapy. We will examine future opportunities for the development and implementation of more effective combination regimens, targeting key components of the oncogenic Src signalling axis, and in the context of a precision medicine-guided approach.
Collapse
Affiliation(s)
- Ashleigh Parkin
- The Kinghorn Cancer CentreThe Garvan Institute of Medical ResearchSydneyAustralia
| | - Jennifer Man
- The Kinghorn Cancer CentreThe Garvan Institute of Medical ResearchSydneyAustralia
| | - Paul Timpson
- The Kinghorn Cancer CentreThe Garvan Institute of Medical ResearchSydneyAustralia
- Faculty of MedicineSt Vincent's Clinical SchoolUniversity of NSWSydneyAustralia
| | - Marina Pajic
- The Kinghorn Cancer CentreThe Garvan Institute of Medical ResearchSydneyAustralia
- Faculty of MedicineSt Vincent's Clinical SchoolUniversity of NSWSydneyAustralia
| |
Collapse
|
42
|
Cai ML, Wang MY, Zhang CH, Wang JX, Liu H, He HW, Zhao WL, Xia GM, Shao RG. Role of co- and post-translational modifications of SFKs in their kinase activation. J Drug Target 2019; 28:23-32. [PMID: 31094236 DOI: 10.1080/1061186x.2019.1616297] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Src family kinases (SFKs) are non-receptor tyrosine kinases and are involved in various cellular functions (proliferation, differentiation, migration, survival and invasion) by regulating downstream pathways. Considerable evidence suggests that co- and post-translational modifications are highly related to the activation of SFKs and their downstream signals. How SFKs are activated and how their subsequent cascades were regulated has been reviewed in previous reports. However, the contribution of co- and post-translational modification to SFKs activation has not been fully elucidated. This review focuses on the effect of these modifications on SFKs activity according to structural and biochemical studies and uncovers the significance of co-and post-translational modifications in the regulation of SFKs activity.
Collapse
Affiliation(s)
- Mei-Lian Cai
- China Academy of Medical Sciences, Key Laboratory of Antibiotic Bioengineering, Ministry of Health, Laboratory of Oncology, Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Meng-Yan Wang
- China Academy of Medical Sciences, Key Laboratory of Antibiotic Bioengineering, Ministry of Health, Laboratory of Oncology, Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Cong-Hui Zhang
- China Academy of Medical Sciences, Key Laboratory of Antibiotic Bioengineering, Ministry of Health, Laboratory of Oncology, Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Jun-Xia Wang
- China Academy of Medical Sciences, Key Laboratory of Antibiotic Bioengineering, Ministry of Health, Laboratory of Oncology, Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Hong Liu
- China Academy of Medical Sciences, Key Laboratory of Antibiotic Bioengineering, Ministry of Health, Laboratory of Oncology, Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Hong-Wei He
- China Academy of Medical Sciences, Key Laboratory of Antibiotic Bioengineering, Ministry of Health, Laboratory of Oncology, Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Wu-Li Zhao
- China Academy of Medical Sciences, Key Laboratory of Antibiotic Bioengineering, Ministry of Health, Laboratory of Oncology, Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Gui-Ming Xia
- China Academy of Medical Sciences, Key Laboratory of Antibiotic Bioengineering, Ministry of Health, Laboratory of Oncology, Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Rong-Guang Shao
- China Academy of Medical Sciences, Key Laboratory of Antibiotic Bioengineering, Ministry of Health, Laboratory of Oncology, Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
43
|
Kim S, Yang X, Yin A, Zha J, Beharry Z, Bai A, Bielawska A, Bartlett MG, Yin H, Cai H. Dietary palmitate cooperates with Src kinase to promote prostate tumor progression. Prostate 2019; 79:896-908. [PMID: 30900312 PMCID: PMC6502658 DOI: 10.1002/pros.23796] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 02/14/2019] [Accepted: 02/28/2019] [Indexed: 12/14/2022]
Abstract
Numerous genetic alterations have been identified during prostate cancer progression. The influence of environmental factors, particularly the diet, on the acceleration of tumor progression is largely unknown. Expression levels and/or activity of Src kinase are highly elevated in numerous cancers including advanced stages of prostate cancer. In this study, we demonstrate that high-fat diets (HFDs) promoted pathological transformation mediated by the synergy of Src and androgen receptor in vivo. Additionally, a diet high in saturated fat significantly enhanced proliferation of Src-mediated xenograft tumors in comparison with a diet high in unsaturated fat. The saturated fatty acid palmitate, a major constituent in a HFD, significantly upregulated the biosynthesis of palmitoyl-CoA in cancer cells in vitro and in xenograft tumors in vivo. The exogenous palmitate enhanced Src-dependent mitochondrial β-oxidation. Additionally, it elevated the amount of C16-ceramide and total saturated ceramides, increased the level of Src kinase localized in the cell membrane, and Src-mediated downstream signaling, such as the activation of mitogen-activated protein kinase and focal adhesion kinase. Our results uncover how the metabolism of dietary palmitate cooperates with elevated Src kinase in the acceleration of prostate tumor progression.
Collapse
Affiliation(s)
- Sungjin Kim
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, Georgia 30602
| | - Xiangkun Yang
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, Georgia 30602
| | - Amelia Yin
- Center for Molecular Medicine, Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia 30602
| | - Junyi Zha
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, Georgia 30602
| | - Zanna Beharry
- Department of Chemistry and Physics, Florida Gulf Coast University, Fort Myers, Florida 33965
| | - Aiping Bai
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Alicja Bielawska
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Michael G. Bartlett
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, Georgia 30602
| | - Hang Yin
- Center for Molecular Medicine, Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia 30602
| | - Houjian Cai
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, Georgia 30602
| |
Collapse
|
44
|
Zhou J, Jiang X, He S, Jiang H, Feng F, Liu W, Qu W, Sun H. Rational Design of Multitarget-Directed Ligands: Strategies and Emerging Paradigms. J Med Chem 2019; 62:8881-8914. [PMID: 31082225 DOI: 10.1021/acs.jmedchem.9b00017] [Citation(s) in RCA: 172] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Due to the complexity of multifactorial diseases, single-target drugs do not always exhibit satisfactory efficacy. Recently, increasing evidence indicates that simultaneous modulation of multiple targets may improve both therapeutic safety and efficacy, compared with single-target drugs. However, few multitarget drugs are on market or in clinical trials, despite the best efforts of medicinal chemists. This article discusses the systematic establishment of target combination, lead generation, and optimization of multitarget-directed ligands (MTDLs). Moreover, we analyze some MTDLs research cases for several complex diseases in recent years and the physicochemical properties of 117 clinical multitarget drugs, with the aim to reveal the trends and insights of the potential use of MTDLs.
Collapse
Affiliation(s)
- Junting Zhou
- Department of Medicinal Chemistry , China Pharmaceutical University , Nanjing 211198 , People's Republic of China.,Department of Natural Medicinal Chemistry , China Pharmaceutical University , Nanjing , 211198 , People's Republic of China
| | - Xueyang Jiang
- Department of Medicinal Chemistry , China Pharmaceutical University , Nanjing 211198 , People's Republic of China.,Department of Natural Medicinal Chemistry , China Pharmaceutical University , Nanjing , 211198 , People's Republic of China
| | - Siyu He
- Department of Medicinal Chemistry , China Pharmaceutical University , Nanjing 211198 , People's Republic of China
| | - Hongli Jiang
- Department of Medicinal Chemistry , China Pharmaceutical University , Nanjing 211198 , People's Republic of China.,Department of Natural Medicinal Chemistry , China Pharmaceutical University , Nanjing , 211198 , People's Republic of China
| | - Feng Feng
- Department of Natural Medicinal Chemistry , China Pharmaceutical University , Nanjing , 211198 , People's Republic of China.,Jiangsu Food and Pharmaceutical Science College , Huaian 223003 , People's Republic of China
| | - Wenyuan Liu
- Department of Analytical Chemistry , China Pharmaceutical University , Nanjing 210009 , People's Republic of China
| | - Wei Qu
- Department of Natural Medicinal Chemistry , China Pharmaceutical University , Nanjing , 211198 , People's Republic of China
| | - Haopeng Sun
- Department of Medicinal Chemistry , China Pharmaceutical University , Nanjing 211198 , People's Republic of China
| |
Collapse
|
45
|
Ren Y, He Y, Brown S, Zbornik E, Mlodzianoski MJ, Ma D, Huang F, Mattoo S, Suter DM. A single tyrosine phosphorylation site in cortactin is important for filopodia formation in neuronal growth cones. Mol Biol Cell 2019; 30:1817-1833. [PMID: 31116646 PMCID: PMC6727743 DOI: 10.1091/mbc.e18-04-0202] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Cortactin is a Src tyrosine phosphorylation substrate that regulates multiple actin-related cellular processes. While frequently studied in nonneuronal cells, the functions of cortactin in neuronal growth cones are not well understood. We recently reported that cortactin mediates the effects of Src tyrosine kinase in regulating actin organization and dynamics in both lamellipodia and filopodia of Aplysia growth cones. Here, we identified a single cortactin tyrosine phosphorylation site (Y499) to be important for the formation of filopodia. Overexpression of a 499F phospho-deficient cortactin mutant decreased filopodia length and density, whereas overexpression of a 499E phospho-mimetic mutant increased filopodia length. Using an antibody against cortactin pY499, we showed that tyrosine-phosphorylated cortactin is enriched along the leading edge. The leading edge localization of phosphorylated cortactin is Src2-dependent, F-actin-independent, and important for filopodia formation. In vitro kinase assays revealed that Src2 phosphorylates cortactin at Y499, although Y505 is the preferred site in vitro. Finally, we provide evidence that Arp2/3 complex acts downstream of phosphorylated cortactin to regulate density but not length of filopodia. In conclusion, we have characterized a tyrosine phosphorylation site in Aplysia cortactin that plays a major role in the Src/cortactin/Arp2/3 signaling pathway controlling filopodia formation.
Collapse
Affiliation(s)
- Yuan Ren
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907
| | - Yingpei He
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907
| | - Sherlene Brown
- Department of Biochemistry, Purdue University, West Lafayette, IN 47907
| | - Erica Zbornik
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907
| | - Michael J Mlodzianoski
- Department of Weldon School of Biomedical Engineering, Purdue Institutes of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN 47907
| | - Donghan Ma
- Department of Weldon School of Biomedical Engineering, Purdue Institutes of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN 47907
| | - Fang Huang
- Department of Weldon School of Biomedical Engineering, Purdue Institutes of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN 47907.,Department of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN 47907.,Department of Integrative Neuroscience, Purdue University, West Lafayette, IN 47907
| | - Seema Mattoo
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907.,Department of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN 47907
| | - Daniel M Suter
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907.,Department of Integrative Neuroscience, Purdue University, West Lafayette, IN 47907.,Department of Bindley Bioscience Center, Purdue University, West Lafayette, IN 47907.,Department of Birck Nanotechnology Center, Purdue University, West Lafayette, IN 47907
| |
Collapse
|
46
|
A Cell-Autonomous Oncosuppressive Role of Human RNASET2 Affecting ECM-Mediated Oncogenic Signaling. Cancers (Basel) 2019; 11:cancers11020255. [PMID: 30813308 PMCID: PMC6406318 DOI: 10.3390/cancers11020255] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 02/18/2019] [Accepted: 02/20/2019] [Indexed: 02/05/2023] Open
Abstract
RNASET2 is an extracellular ribonuclease endowed with a marked antitumorigenic role in several carcinomas, independent from its catalytic activity. Besides its antitumorigenic role by the recruitment to the tumor mass of immune cells from the monocyte/macrophage lineage, RNASET2 is induced by cellular stress and involved in actin cytoskeleton remodeling affecting cell interactions with the extracellular matrix (ECM). Here, we aimed to investigate the effects of RNASET2 expression modulation on cell phenotype and behavior in epithelial ovarian cancer (EOC) cellular models. In silico analysis on two publicly available datasets of gene expression from EOC patients (n = 392) indicated that increased RNASET2 transcript levels are associated with longer overall survival. In EOC biopsies (n = 101), analyzed by immunohistochemistry, RNASET2 was found heterogeneously expressed among tumors with different clinical⁻pathological characteristics and, in some cases, its expression localized to tumor-associated ECM. By characterizing in vitro two models of EOC cells in which RNASET2 was silenced or overexpressed, we report that RNASET2 expression negatively affects growth capability by conferring a peculiar cell phenotype upon the interaction of EOC cells with the ECM, resulting in decreased src activation. Altogether, these data suggest that drugs targeting activated src might represent a therapeutic approach for RNASET2-expressing EOCs.
Collapse
|
47
|
Gao Z, Zhao GS, Lv Y, Peng D, Tang X, Song H, Guo QN. Anoikis‑resistant human osteosarcoma cells display significant angiogenesis by activating the Src kinase‑mediated MAPK pathway. Oncol Rep 2018; 41:235-245. [PMID: 30542722 PMCID: PMC6278590 DOI: 10.3892/or.2018.6827] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 10/19/2018] [Indexed: 02/06/2023] Open
Abstract
Tumor cells must resist anoikis to metastasize. There is a key role of angiogenesis in the growth and metastasis of tumors. However, the relationship between anoikis resistance and angiogenesis has not been well explored in human osteosarcoma. In the present study, we reported the higher expression of vascular endothelial growth factor-A (VEGF-A) in osteosarcoma cells that were resistant to anoikis than in parental osteosarcoma cells, promoting the proliferation, tube formation, and migration of human umbilical vein endothelial cells (HUVECs). Src, JNK (Jun amino-terminal kinase) and ERK (extracellular signal-regulated kinase) signaling pathway phosphorylation was activated in anoikis-resistant cells; Src inhibitor reduced the expression of VEGF-A and angiogenesis and inhibited JNK and ERK pathway activity. Overexpression of phosphorylated (p)-Src and VEGF-A was positively correlated to the metastatic potential in human osteosarcoma tissues, as quantified by immunohistochemistry. In addition, p-Src expression was directly correlated with VEGF-A expression and microvessel density in vivo. Our findings revealed that anoikis resistance in osteosarcoma cells increased the expression of VEGF-A and angiogenesis through the Src/JNK/ERK signaling pathways. Thus, Src may be a potential therapeutic alternative in osteosarcoma angiogenesis and metastasis.
Collapse
Affiliation(s)
- Ziran Gao
- Department of Pathology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, P.R. China
| | - Guo-Sheng Zhao
- Department of Orthopedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Yangfan Lv
- Department of Pathology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, P.R. China
| | - Dongbin Peng
- Department of Pathology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, P.R. China
| | - Xuefeng Tang
- Department of Pathology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, P.R. China
| | - Hanxiang Song
- Department of Pathology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, P.R. China
| | - Qiao-Nan Guo
- Department of Pathology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, P.R. China
| |
Collapse
|
48
|
Zeng J, Zhang H, Tan Y, Sun C, Liang Y, Yu J, Zou H. Aggregation of lipid rafts activates c-met and c-Src in non-small cell lung cancer cells. BMC Cancer 2018; 18:611. [PMID: 29848294 PMCID: PMC5977465 DOI: 10.1186/s12885-018-4501-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 05/11/2018] [Indexed: 11/30/2022] Open
Abstract
Background Activation of c-Met, a receptor tyrosine kinase, induces radiation therapy resistance in non-small cell lung cancer (NSCLC). The activated residual of c-Met is located in lipid rafts (Duhon et al. Mol Carcinog 49:739-49, 2010). Therefore, we hypothesized that disturbing the integrity of lipid rafts would restrain the activation of the c-Met protein and reverse radiation resistance in NSCLC. In this study, a series of experiments was performed to test this hypothesis. Methods NSCLC A549 and H1993 cells were incubated with methyl-β-cyclodextrin (MβCD), a lipid raft inhibitor, at different concentrations for 1 h before the cells were X-ray irradiated. The following methods were used: clonogenic (colony-forming) survival assays, flow cytometry (for cell cycle and apoptosis analyses), immunofluorescence microscopy (to show the distribution of proteins in lipid rafts), Western blotting, and biochemical lipid raft isolation (purifying lipid rafts to show the distribution of proteins in lipid rafts). Results Our results showed that X-ray irradiation induced the aggregation of lipid rafts in A549 cells, activated c-Met and c-Src, and induced c-Met and c-Src clustering to lipid rafts. More importantly, MβCD suppressed the proliferation of A549 and H1993 cells, and the combination of MβCD and radiation resulted in additive increases in A549 and H1993 cell apoptosis. Destroying the integrity of lipid rafts inhibited the aggregation of c-Met and c-Src to lipid rafts and reduced the expression of phosphorylated c-Met and phosphorylated c-Src in lipid rafts. Conclusions X-ray irradiation induced the aggregation of lipid rafts and the clustering of c-Met and c-Src to lipid rafts through both lipid raft-dependent and lipid raft-independent mechanisms. The lipid raft-dependent activation of c-Met and its downstream pathways played an important role in the development of radiation resistance in NSCLC cells mediated by c-Met. Further studies are still required to explore the molecular mechanisms of the activation of c-Met and c-Src in lipid rafts induced by radiation. Electronic supplementary material The online version of this article (10.1186/s12885-018-4501-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Juan Zeng
- The First Oncology Department, Shengjing Hospital affiliated with China Medical University, Shenyang, 110004, China
| | - Heying Zhang
- The First Oncology Department, Shengjing Hospital affiliated with China Medical University, Shenyang, 110004, China
| | - Yonggang Tan
- The First Oncology Department, Shengjing Hospital affiliated with China Medical University, Shenyang, 110004, China
| | - Cheng Sun
- The First Oncology Department, Shengjing Hospital affiliated with China Medical University, Shenyang, 110004, China
| | - Yusi Liang
- The First Oncology Department, Shengjing Hospital affiliated with China Medical University, Shenyang, 110004, China
| | - Jinyang Yu
- The First Oncology Department, Shengjing Hospital affiliated with China Medical University, Shenyang, 110004, China
| | - Huawei Zou
- The First Oncology Department, Shengjing Hospital affiliated with China Medical University, Shenyang, 110004, China.
| |
Collapse
|
49
|
Woodcock VK, Clive S, Wilson RH, Coyle VM, Stratford MRL, Folkes LK, Eastell R, Barton C, Jones P, Kazmi-Stokes S, Turner H, Halford S, Harris AL, Middleton MR. A first-in-human phase I study to determine the maximum tolerated dose of the oral Src/ABL inhibitor AZD0424. Br J Cancer 2018; 118:770-776. [PMID: 29438361 PMCID: PMC5877436 DOI: 10.1038/bjc.2017.484] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 12/09/2017] [Accepted: 12/12/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Src is involved in cancer invasion and metastasis. AZD0424, an oral inhibitor of Src and ABL1, has shown evidence of anti-tumour activity in pre-clinical studies. METHODS A phase Ia, dose escalation study was performed to assess the safety of continuous oral dosing with AZD0424 in advanced solid tumours. Secondary objectives included investigation of AZD0424 pharmacokinetics, effect on Src activity using markers of bone turnover, and anti-tumour activity. RESULTS 41 patients were treated; 34 received AZD0424 once-daily at doses ranging from 5 mg to 150 mg, and 7 received 40 mg bi-daily 41.5% of patients experienced at least one AZD0424-related adverse event that was Grade 3-5 in severity, with patients treated at doses above 60 mg per day experiencing multiple treatment-related toxicities. The most commonly observed AZD0424-related adverse events were nausea, fatigue, anorexia and alopecia. Cmax and AUC increased linearly with dose and the mean±standard deviation t1/2 was 8.4±2.8 h. Clear evidence of Src target inhibition was seen at doses ⩾20 mg per day. No responses were observed and 7 patients (17.1%) achieved stable disease lasting 6 weeks or more. CONCLUSIONS AZD0424 displayed no evidence of efficacy as monotherapy despite a clear pharmacodynamic effect. Further evaluation of AZD0424 monotherapy in patients with solid tumours is not recommended.
Collapse
Affiliation(s)
- Victoria K Woodcock
- University of Oxford Department of Oncology, Churchill Hospital, Old Road, Oxford OX3 7LJ, UK
| | - Sally Clive
- Edinburgh Cancer Centre, Western General Hospital, Edinburgh EH4 2XU, UK
| | - Richard H Wilson
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Lisburn Road, Belfast BT9 7AE, Northern Ireland, UK
| | - Vicky M Coyle
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Lisburn Road, Belfast BT9 7AE, Northern Ireland, UK
| | - Michael R L Stratford
- Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Lisa K Folkes
- Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Richard Eastell
- Academic Unit of Bone Metabolism, University of Sheffield, Sheffield S10 2TN, UK
| | - Claire Barton
- Cancer Research UK Centre for Drug Development, Cancer Research UK, Angel Building, 407 St. John Street, London EC1V 4AD, UK
| | - Paul Jones
- Cancer Research UK Centre for Drug Development, Cancer Research UK, Angel Building, 407 St. John Street, London EC1V 4AD, UK
| | - Shamim Kazmi-Stokes
- Cancer Research UK Centre for Drug Development, Cancer Research UK, Angel Building, 407 St. John Street, London EC1V 4AD, UK
| | - Helen Turner
- Cancer Research UK Centre for Drug Development, Cancer Research UK, Angel Building, 407 St. John Street, London EC1V 4AD, UK
| | - Sarah Halford
- Cancer Research UK Centre for Drug Development, Cancer Research UK, Angel Building, 407 St. John Street, London EC1V 4AD, UK
| | - Adrian L Harris
- University of Oxford Department of Oncology, Churchill Hospital, Old Road, Oxford OX3 7LJ, UK
| | - Mark R Middleton
- University of Oxford Department of Oncology, Churchill Hospital, Old Road, Oxford OX3 7LJ, UK
- National Institute for Health Research Oxford Biomedical Research Centre, Oxford OX3 7LE, UK
| |
Collapse
|
50
|
Melone MAB, Valentino A, Margarucci S, Galderisi U, Giordano A, Peluso G. The carnitine system and cancer metabolic plasticity. Cell Death Dis 2018; 9:228. [PMID: 29445084 PMCID: PMC5833840 DOI: 10.1038/s41419-018-0313-7] [Citation(s) in RCA: 138] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 01/09/2018] [Accepted: 01/11/2018] [Indexed: 12/11/2022]
Abstract
Metabolic flexibility describes the ability of cells to respond or adapt its metabolism to support and enable rapid proliferation, continuous growth, and survival in hostile conditions. This dynamic character of the cellular metabolic network appears enhanced in cancer cells, in order to increase the adaptive phenotype and to maintain both viability and uncontrolled proliferation. Cancer cells can reprogram their metabolism to satisfy the energy as well as the biosynthetic intermediate request and to preserve their integrity from the harsh and hypoxic environment. Although several studies now recognize these reprogrammed activities as hallmarks of cancer, it remains unclear which are the pathways involved in regulating metabolic plasticity. Recent findings have suggested that carnitine system (CS) could be considered as a gridlock to finely trigger the metabolic flexibility of cancer cells. Indeed, the components of this system are involved in the bi-directional transport of acyl moieties from cytosol to mitochondria and vice versa, thus playing a fundamental role in tuning the switch between the glucose and fatty acid metabolism. Therefore, the CS regulation, at both enzymatic and epigenetic levels, plays a pivotal role in tumors, suggesting new druggable pathways for prevention and treatment of human cancer.
Collapse
Affiliation(s)
- Mariarosa Anna Beatrice Melone
- Department of Medical, Surgical, Neurological, Metabolic Sciences, and Aging, 2nd Division of Neurology, Center for Rare Diseases and InterUniversity Center for Research in Neurosciences, University of Campania "Luigi Vanvitelli", Naples, Italy
- Department of Biology, Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, USA
| | - Anna Valentino
- Department of Medical, Surgical, Neurological, Metabolic Sciences, and Aging, 2nd Division of Neurology, Center for Rare Diseases and InterUniversity Center for Research in Neurosciences, University of Campania "Luigi Vanvitelli", Naples, Italy
- Institute of Agro-Environmental and Forest Biology, National Research Council, IBAF-CNR, Naples, Italy
| | | | - Umberto Galderisi
- Department of Experimental Medicine, Biotechnology and Molecular Biology Section, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Antonio Giordano
- Department of Biology, Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, USA.
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy.
| | - Gianfranco Peluso
- Institute of Agro-Environmental and Forest Biology, National Research Council, IBAF-CNR, Naples, Italy.
| |
Collapse
|