1
|
Koval A, Mamadalieva NZ, Mamadalieva R, Jalilov F, Katanaev VL. Success and Controversy of Natural Products as Therapeutic Modulators of Wnt Signaling and Its Interplay with Oxidative Stress: Comprehensive Review Across Compound Classes and Experimental Systems. Antioxidants (Basel) 2025; 14:591. [PMID: 40427472 DOI: 10.3390/antiox14050591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2025] [Revised: 05/05/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
The highly conserved Wnt signaling pathway, a complex network critical for embryonic development and adult tissue homeostasis, regulates diverse cellular processes, ultimately influencing tissue organization and organogenesis; its dysregulation is implicated in numerous diseases, and it is known to be affected by oxidative pathways. This report reviews the recent literature on major classes of natural products with pronounced anti-oxidant properties, such as cardiac glycosides, steroid saponins, ecdysteroids, withanolides, cucurbitacins, triterpenes, flavonoids, and iridoids, that modulate its activity in various pathological conditions, summarizing and critically analyzing their effects on the Wnt pathway in various therapeutically relevant experimental models and highlighting the role of ROS-mediated crosstalk with Wnt signaling in these studies. Models reviewed include not only cancer but also stroke, ischemia, bone or kidney diseases, and regenerative medicine, such as re-epithelialization, cardiac maintenance, and hair loss. It highlights the paramount importance of modulating this signaling by natural products to define future research directions. We also discuss controversies identified in the mode of action of several compounds in different models and directions on how to further improve the quality and depth of such studies.
Collapse
Affiliation(s)
- Alexey Koval
- Translational Research Center in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Nilufar Z Mamadalieva
- Translational Research Center in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
- Faculty of Medicine, Alfraganus University, Yuqori Qoraqamish Str. 2a, Tashkent 100190, Uzbekistan
- Institute of the Chemistry of Plant Substances, Uzbekistan Academy of Sciences, Mirzo Ulugbek Str. 77, Tashkent 100170, Uzbekistan
| | - Rano Mamadalieva
- Faculty of Medicine, Namangan University of Business and Science, Beshkapa Str. 111, Namangan 160100, Uzbekistan
| | - Fazliddin Jalilov
- Faculty of Medicine, Alfraganus University, Yuqori Qoraqamish Str. 2a, Tashkent 100190, Uzbekistan
| | - Vladimir L Katanaev
- Translational Research Center in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
- Translational Oncology Research Center, Qatar Biomedical Research Institute (QBRI), College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha P.O. Box 34110, Qatar
| |
Collapse
|
2
|
Ren Z, Su R, Liu D, Wang Q, Liu S, Kong D, Qiu Y. Yes-associated protein indispensably mediates hirsutine-induced inhibition on cell growth and Wnt/β-catenin signaling in colorectal cancer. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156156. [PMID: 39437684 DOI: 10.1016/j.phymed.2024.156156] [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: 07/04/2024] [Revised: 09/26/2024] [Accepted: 10/12/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND AND PURPOSE Targeting Wnt/β-catenin signaling emerges as one of the promising strategies for colorectal cancer (CRC) treatment, as this signaling is highly activated in CRC progression. Despite reports on the cytotoxic effects of hirsutine (HT), an indole alkaloid found in herbal medicines from the genus Uncaria, its therapeutic potential for CRC and the involved mechanisms are poorly understood. This study investigates the anticancer efficacy and the probable mechanisms of HT against CRC. METHODS To evaluate in vitro anticancer activity of HT, cell growth examined by MTT and colony formation assay, and apoptosis examined by flow cytometry were analyzed. To explore the mechanisms, RNA-sequencing, western blotting, dual-luciferase reporter assays, immunofluorescence, and co-immunoprecipitation were performed. Mouse model of azoxymethane/dextran sodium sulfate (AOM/DSS)-induced colon cancer was utilized to assess HT's in vivo anticancer efficacy. RESULTS HT significantly inhibited CRC cell proliferation with IC50 values of 22.25 ± 3.27 μM for SW620 cells and 22.24 ± 2.36 μM for HCT116 cells, and induced apoptosis. HT decreased protein levels of Wnt3a and β-catenin dose- and time-dependently, and inhibited TOP/FOP FLASH reporter activity, nuclear travel of β-catenin, and downstream targets like c-Myc, Cyclin D1, VEGF. HT reduced β-catenin protein half-life, and the reversal of this effect by MG132 indicated that HT facilitated proteasome-dependent degradation of β-catenin in these two cell lines. HT also increased β-catenin ubiquitination without affecting Axin and β-TrCP levels. HT treatment for 24 h induced YAP cytoplasmic retention, enhanced YAP interacting with β-catenin and β-TrCP, triggering destruction complex formation and β-catenin ubiquitination and degradation, while YAP siRNA impaired these effects. Additionally, β-catenin overexpression and LiCl treatment counteracted HT-induced inhibition on cell growth and Wnt/β-catenin cascade. In model of AOM/DSS-induced mouse colon cancer, compared with AOM/DSS treatment group, HT recovered colon length, reduced tumor numbers and radius, and downregulated β-catenin and Ki-67, while upregulated cleaved PARP in the colorectal tissue with tumors. CONCLUSION HT exhibits anticancer activity against CRC probably by inhibiting Wnt/β-catenin signaling, with YAP playing an indispensible role during the process, highlighting HT as a potential novel candidate drug for CRC therapy.
Collapse
Affiliation(s)
- Zehao Ren
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Ruixin Su
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Donghui Liu
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Qian Wang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Shanshan Liu
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Dexin Kong
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China.
| | - Yuling Qiu
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
3
|
Reissland M, Hartmann O, Tauch S, Bugter JM, Prieto-Garcia C, Schulte C, Loebbert S, Solvie D, Bitman-Lotan E, Narain A, Jacomin AC, Schuelein-Voelk C, Fuss CT, Pahor N, Ade C, Buck V, Potente M, Li V, Beliu G, Wiegering A, Grossmann T, Eilers M, Wolf E, Maric H, Rosenfeldt M, Maurice MM, Dikic I, Gallant P, Orian A, Diefenbacher ME. USP10 drives cancer stemness and enables super-competitor signalling in colorectal cancer. Oncogene 2024; 43:3645-3659. [PMID: 39443725 PMCID: PMC11611742 DOI: 10.1038/s41388-024-03141-x] [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: 12/12/2023] [Revised: 07/30/2024] [Accepted: 08/21/2024] [Indexed: 10/25/2024]
Abstract
The contribution of deubiquitylating enzymes (DUBs) to β-Catenin stabilization in intestinal stem cells and colorectal cancer (CRC) is poorly understood. Here, and by using an unbiassed screen, we discovered that the DUB USP10 stabilizes β-Catenin specifically in APC-truncated CRC in vitro and in vivo. Mechanistic studies, including in vitro binding together with computational modelling, revealed that USP10 binding to β-Catenin is mediated via the unstructured N-terminus of USP10 and is outcompeted by intact APC, favouring β-catenin degradation. However, in APC-truncated cancer cells USP10 binds to β-catenin, increasing its stability which is critical for maintaining an undifferentiated tumour identity. Elimination of USP10 reduces the expression of WNT and stem cell signatures and induces the expression of differentiation genes. Remarkably, silencing of USP10 in murine and patient-derived CRC organoids established that it is essential for NOTUM signalling and the APC super competitor-phenotype, reducing tumorigenic properties of APC-truncated CRC. These findings are clinically relevant as patient-derived organoids are highly dependent on USP10, and abundance of USP10 correlates with poorer prognosis of CRC patients. Our findings reveal, therefore, a role for USP10 in CRC cell identity, stemness, and tumorigenic growth by stabilising β-Catenin, leading to aberrant WNT signalling and degradation resistant tumours. Thus, USP10 emerges as a unique therapeutic target in APC truncated CRC.
Collapse
Affiliation(s)
- Michaela Reissland
- Protein Stability and Cancer Group, University of Wuerzburg, Department of Biochemistry and Molecular Biology, Wuerzburg, Germany
- Mildred-Scheel Early Career Cancer Center, Wuerzburg, Germany
- Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Oliver Hartmann
- Protein Stability and Cancer Group, University of Wuerzburg, Department of Biochemistry and Molecular Biology, Wuerzburg, Germany
- Mildred-Scheel Early Career Cancer Center, Wuerzburg, Germany
| | - Saskia Tauch
- Protein Stability and Cancer Group, University of Wuerzburg, Department of Biochemistry and Molecular Biology, Wuerzburg, Germany
- Division of Signal Transduction and Growth Control, DKFZ/ZMBH Alliance, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jeroen M Bugter
- Oncode Institute and Department of Cell Biology, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, Netherlands
| | - Cristian Prieto-Garcia
- Protein Stability and Cancer Group, University of Wuerzburg, Department of Biochemistry and Molecular Biology, Wuerzburg, Germany
- Molecular Signalling Group, Institute of Biochemistry II, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Clemens Schulte
- Rudolf-Virchow-Center for Integrative and Translational Imaging, University of Wuerzburg, Wuerzburg, Germany
| | - Sinah Loebbert
- Mildred-Scheel Early Career Cancer Center, Wuerzburg, Germany
- Department of Biochemistry and Molecular Biology, University of Wuerzburg, Wuerzburg, Germany
| | - Daniel Solvie
- Department of Biochemistry and Molecular Biology, University of Wuerzburg, Wuerzburg, Germany
| | | | - Ashwin Narain
- Cancer Systems Biology Group, University of Wuerzburg, Am Hubland, Wuerzburg, Germany
| | - Anne-Claire Jacomin
- Molecular Signalling Group, Institute of Biochemistry II, Goethe University Frankfurt, Frankfurt am Main, Germany
| | | | - Carmina T Fuss
- Protein Stability and Cancer Group, University of Wuerzburg, Department of Biochemistry and Molecular Biology, Wuerzburg, Germany
- Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Wuerzburg, Wuerzburg, Germany
| | - Nikolett Pahor
- Protein Stability and Cancer Group, University of Wuerzburg, Department of Biochemistry and Molecular Biology, Wuerzburg, Germany
- Mildred-Scheel Early Career Cancer Center, Wuerzburg, Germany
| | - Carsten Ade
- Department of Biochemistry and Molecular Biology, University of Wuerzburg, Wuerzburg, Germany
| | - Viktoria Buck
- Institute for Pathology, University of Würzburg, Wuerzburg, Germany
| | - Michael Potente
- Angiogenesis & Metabolism Laboratory, Berlin Institute of Health at Charité, Universitätsmedizin Berlin, Berlin Germany and Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Vivian Li
- Stem Cell and Cancer Biology Laboratory, The Francis Crick Institute, London, UK
| | - Gerti Beliu
- Rudolf-Virchow-Center for Integrative and Translational Imaging, University of Wuerzburg, Wuerzburg, Germany
| | - Armin Wiegering
- Department of Biochemistry and Molecular Biology, University of Wuerzburg, Wuerzburg, Germany
- Department of General, Visceral, Transplantation, Vascular and Paediatric Surgery, University Hospital, Julius-Maximilians-University of Wuerzburg, Wuerzburg, Germany
| | - Tom Grossmann
- Amsterdam Institute of Molecular and Life Sciences, Amsterdam, Netherlands
| | - Martin Eilers
- Mildred-Scheel Early Career Cancer Center, Wuerzburg, Germany
- Department of Biochemistry and Molecular Biology, University of Wuerzburg, Wuerzburg, Germany
| | - Elmar Wolf
- Mildred-Scheel Early Career Cancer Center, Wuerzburg, Germany
- Cancer Systems Biology Group, University of Wuerzburg, Am Hubland, Wuerzburg, Germany
| | - Hans Maric
- Rudolf-Virchow-Center for Integrative and Translational Imaging, University of Wuerzburg, Wuerzburg, Germany
| | - Mathias Rosenfeldt
- Mildred-Scheel Early Career Cancer Center, Wuerzburg, Germany
- Institute for Pathology, University of Würzburg, Wuerzburg, Germany
| | - Madelon M Maurice
- Oncode Institute and Department of Cell Biology, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, Netherlands
| | - Ivan Dikic
- Molecular Signalling Group, Institute of Biochemistry II, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Peter Gallant
- Department of Biochemistry and Molecular Biology, University of Wuerzburg, Wuerzburg, Germany
| | - Amir Orian
- Faculty of Medicine, TICC, Technion Haifa, Haifa, Israel
| | - Markus E Diefenbacher
- Protein Stability and Cancer Group, University of Wuerzburg, Department of Biochemistry and Molecular Biology, Wuerzburg, Germany.
- Mildred-Scheel Early Career Cancer Center, Wuerzburg, Germany.
- Institute of Lung Health and Immunity, Helmholtz Center, Munich, Germany.
- German Center for Lung Research, DZL, Germany.
- Ludwig Maximilian University Munich, Munich, Germany.
| |
Collapse
|
4
|
Gelsomino L, Caruso A, Tasan E, Leonetti AE, Malivindi R, Naimo GD, Giordano F, Panza S, Gu G, Perrone B, Giordano C, Mauro L, Nardo B, Filippelli G, Bonofiglio D, Barone I, Fuqua SAW, Catalano S, Andò S. Evidence that CRISPR-Cas9 Y537S-mutant expressing breast cancer cells activate Yes-associated protein 1 to driving the conversion of normal fibroblasts into cancer-associated fibroblasts. Cell Commun Signal 2024; 22:545. [PMID: 39543704 PMCID: PMC11566413 DOI: 10.1186/s12964-024-01918-x] [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: 07/29/2024] [Accepted: 10/31/2024] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND Endocrine therapy (ET) has improved the clinical outcomes of Estrogen receptor alpha-positive (ERɑ +) breast cancer (BC) patients, even though resistance to ET remains a clinical issue. Mutations in the hormone-binding domain of ERɑ represent an acquired intrinsic mechanism of ET resistance. However, the latter also depends on the multiple functional interactions between BC cells and the tumor microenvironment (TME). Here, we investigated how the most common Y537S-ERɑ mutation may influence the behavior of fibroblasts, the most prominent component of the TME. METHODS We conducted coculture experiments with normal human foreskin fibroblasts BJ1-hTERT (NFs), cancer-associated fibroblasts (CAFs), isolated from human BC specimens, and Y537S CRISPR-expressing MCF-7 BC cells (MCF-7YS). Mass spectrometry (MS) and Metacore analyses were performed to investigate how the functional interactions between BC cells/fibroblasts may affect their proteomic profile. The impact of fibroblasts on BC tumor growth and metastatic potential was evaluated in nude mice. RESULTS Mutant BC conditioned medium (CM) affected the morphology/proliferation/migration of both NFs and CAFs. 198 deregulated proteins signed the proteomic similarity profile of NFs exposed to the YS-CM and CAFs. Among the upregulated proteins, Yes-associated protein 1 (YAP1) was the main central hub in the direct interaction network. Increased YAP1 protein expression and activity were confirmed in NFs treated with MCF-7YS-CM. However, YAP1 activation appears to crosstalk with the insulin growth factor-1 receptor (IGF-1R). Higher amount of IGF-1 were noticed in the MCF-7YS-CM cells compared to the MCF-7P, and IGF-1 immunodepletion reversed the enhanced YAP1 expression and activity. Mutant cells upon exposure to the NF- and CAF-CM exhibited an enhanced proliferation/growth/migration/invasion compared to the MCF-7P. MCF-7YS cells when implanted with CAFs showed an early relative increased tumor volume compared to YS alone. No changes were observed when MCF-7P cells were co-implanted with CAFs. Compared with that in MCF-7P cells, the metastatic burden of MCF-7YS cells was intrinsically greater, and this effect was augmented upon treatment with NF-CM and further increased with CAF-CM. CONCLUSIONS YS mutant BC cells induced the conversion of fibroblasts into CAFs, via YAP, which represent a potential therapeutic target which interrupt the functional interactions between mutant cells/TME and to be implemented in the novel therapeutic strategy of a subset of metastatic BC patients carrying the frequent Y537S mutations.
Collapse
Affiliation(s)
- Luca Gelsomino
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende (CS), Cosenza, Italy
- Health Center, University of Calabria, Arcavacata Di Rende (CS), Cosenza, Italy
| | - Amanda Caruso
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende (CS), Cosenza, Italy
| | - Emine Tasan
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende (CS), Cosenza, Italy
| | - Adele Elisabetta Leonetti
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende (CS), Cosenza, Italy
| | - Rocco Malivindi
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende (CS), Cosenza, Italy
- Clinical Laboratory Unit, A.O. "Annunziata", Cosenza, Italy
| | - Giuseppina Daniela Naimo
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende (CS), Cosenza, Italy
| | - Francesca Giordano
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende (CS), Cosenza, Italy
| | - Salvatore Panza
- Department of Experimental and Clinical Medicine, "Magna Graecia", University of Catanzaro, Catanzaro, Italy
| | - Guowei Gu
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
| | - Benedetta Perrone
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende (CS), Cosenza, Italy
- Health Center, University of Calabria, Arcavacata Di Rende (CS), Cosenza, Italy
| | - Cinzia Giordano
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende (CS), Cosenza, Italy
- Health Center, University of Calabria, Arcavacata Di Rende (CS), Cosenza, Italy
- Clinical Laboratory Unit, A.O. "Annunziata", Cosenza, Italy
| | - Loredana Mauro
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende (CS), Cosenza, Italy
| | - Bruno Nardo
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende (CS), Cosenza, Italy
- Department of Surgery, General Surgery Unit, Annunziata Hospital, Cosenza, Italy
| | | | - Daniela Bonofiglio
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende (CS), Cosenza, Italy
- Health Center, University of Calabria, Arcavacata Di Rende (CS), Cosenza, Italy
| | - Ines Barone
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende (CS), Cosenza, Italy
- Health Center, University of Calabria, Arcavacata Di Rende (CS), Cosenza, Italy
| | - Suzanne A W Fuqua
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
| | - Stefania Catalano
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende (CS), Cosenza, Italy
- Health Center, University of Calabria, Arcavacata Di Rende (CS), Cosenza, Italy
- Clinical Laboratory Unit, A.O. "Annunziata", Cosenza, Italy
| | - Sebastiano Andò
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende (CS), Cosenza, Italy.
- Health Center, University of Calabria, Arcavacata Di Rende (CS), Cosenza, Italy.
| |
Collapse
|
5
|
Jones I, Arias-Garcia M, Pascual-Vargas P, Beykou M, Dent L, Chaudhuri TP, Roumeliotis T, Choudhary J, Sero J, Bakal C. YAP activation is robust to dilution. Mol Omics 2024; 20:554-569. [PMID: 39282972 PMCID: PMC11403994 DOI: 10.1039/d4mo00100a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 08/07/2024] [Indexed: 09/22/2024]
Abstract
The concentration of many transcription factors exhibits high cell-to-cell variability due to differences in synthesis, degradation, and cell size. Whether the functions of these factors are robust to fluctuations in concentration, and how this may be achieved, is poorly understood. Across two independent panels of breast cancer cells, we show that the average whole cell concentration of YAP decreases as a function of cell area. However, the nuclear concentration distribution remains constant across cells grouped by size, across a 4-8 fold size range, implying unperturbed nuclear translocation despite the falling cell wide concentration. Both the whole cell and nuclear concentration was higher in cells with more DNA and CycA/PCNA expression suggesting periodic synthesis of YAP across the cell cycle offsets dilution due to cell growth and/or cell spreading. The cell area - YAP scaling relationship extended to melanoma and RPE cells. Integrative analysis of imaging and phospho-proteomic data showed the average nuclear YAP concentration across cell lines was predicted by differences in RAS/MAPK signalling, focal adhesion maturation, and nuclear transport processes. Validating the idea that RAS/MAPK and cell cycle regulate YAP translocation, chemical inhibition of MEK or CDK4/6 increased the average nuclear YAP concentration. Together, this study provides an example case, where cytoplasmic dilution of a protein, for example through cell growth, does not limit a cognate cellular function. Here, that same proteins translocation into the nucleus.
Collapse
Affiliation(s)
- Ian Jones
- Chester Beatty Laboratories, Division of Cancer Biology, Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK.
| | - Mar Arias-Garcia
- Chester Beatty Laboratories, Division of Cancer Biology, Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK.
| | - Patricia Pascual-Vargas
- Chester Beatty Laboratories, Division of Cancer Biology, Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK.
| | - Melina Beykou
- Chester Beatty Laboratories, Division of Cancer Biology, Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK.
| | - Lucas Dent
- Chester Beatty Laboratories, Division of Cancer Biology, Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK.
| | - Tara Pal Chaudhuri
- Chester Beatty Laboratories, Division of Cancer Biology, Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK.
| | - Theodoros Roumeliotis
- Chester Beatty Laboratories, Division of Cancer Biology, Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK.
| | - Jyoti Choudhary
- Chester Beatty Laboratories, Division of Cancer Biology, Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK.
| | - Julia Sero
- Institute for Mathematical Innovation, Department of Life Sciences, University of Bath, Claverton Down, Bath BA2 7AY, UK
| | - Chris Bakal
- Chester Beatty Laboratories, Division of Cancer Biology, Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK.
| |
Collapse
|
6
|
Bai Y, Li R, Hao JF, Chen LW, Liu ST, Zhang XL, Lip GYH, Yang JK, Zou YX, Wang H. Accumulated β-catenin is associated with human atrial fibrosis and atrial fibrillation. J Transl Med 2024; 22:734. [PMID: 39103891 PMCID: PMC11302159 DOI: 10.1186/s12967-024-05558-0] [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: 12/01/2023] [Accepted: 07/29/2024] [Indexed: 08/07/2024] Open
Abstract
BACKGROUND Atrial fibrillation (AF) is associated with increased risk of stroke and mortality. It has been reported that the process of atrial fibrosis was regulated by β-catenin in rats with AF. However, pathophysiological mechanisms of this process in human with AF remain unclear. This study aims to investigate the possible mechanisms of β-catenin in participating in the atrial fibrosis using human right atrial appendage (hRAA) tissues . METHODS We compared the difference of β-catenin expression in hRAA tissues between the patients with AF and sinus rhythm (SR). The possible function of β-catenin in the development of AF was also explored in mice and primary cells. RESULTS Firstly, the space between the membrane of the gap junctions of cardiomyocytes was wider in the AF group. Secondly, the expression of the gap junction function related proteins, Connexin40 and Connexin43, was decreased, while the expression of β-catenin and its binding partner E-cadherin was increased in hRAA and cardiomyocytes of the AF group. Thirdly, β-catenin colocalized with E-cadherin on the plasma membrane of cardiomyocytes in the SR group, while they were dissociated and accumulated intracellularly in the AF group. Furthermore, the expression of glycogen synthase kinase 3β (GSK-3β) and Adenomatous Polyposis Coli (APC), which participated in the degradation of β-catenin, was decreased in hRAA tissues and cardiomyocytes of the AF group. Finally, the development of atrial fibrosis and AF were proved to be prevented after inhibiting β-catenin expression in the AF model mice. CONCLUSIONS Based on human atrial pathological and molecular analyses, our findings provided evidence that β-catenin was associated with atrial fibrosis and AF progression.
Collapse
Affiliation(s)
- Ying Bai
- Cardiovascular Center, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Rui Li
- Department of Pathology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Jun-Feng Hao
- Core Facility for Protein Research, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Lian-Wan Chen
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Si-Tong Liu
- Cardiovascular Center, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Xi-Lin Zhang
- Cardiovascular Center, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool Heart & Chest Hospital, Liverpool, UK
- Danish Center for Health Services Research, Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Jin-Kui Yang
- Department of Endocrinology, Beijing Key Laboratory of Diabetes Research and Care, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Yi-Xi Zou
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China.
| | - Hao Wang
- Beijing Key Laboratory of Diabetes Research and Care, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| |
Collapse
|
7
|
Ashique S, Bhowmick M, Pal R, Khatoon H, Kumar P, Sharma H, Garg A, Kumar S, Das U. Multi drug resistance in Colorectal Cancer- approaches to overcome, advancements and future success. ADVANCES IN CANCER BIOLOGY - METASTASIS 2024; 10:100114. [DOI: 10.1016/j.adcanc.2024.100114] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
|
8
|
Zhao Y, Sun B, Fu X, Zuo Z, Qin H, Yao K. YAP in development and disease: Navigating the regulatory landscape from retina to brain. Biomed Pharmacother 2024; 175:116703. [PMID: 38713948 DOI: 10.1016/j.biopha.2024.116703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 04/30/2024] [Accepted: 05/01/2024] [Indexed: 05/09/2024] Open
Abstract
The distinctive role of Yes-associated protein (YAP) in the nervous system has attracted widespread attention. This comprehensive review strategically uses the retina as a vantage point, embarking on an extensive exploration of YAP's multifaceted impact from the retina to the brain in development and pathology. Initially, we explore the crucial roles of YAP in embryonic and cerebral development. Our focus then shifts to retinal development, examining in detail YAP's regulatory influence on the development of retinal pigment epithelium (RPE) and retinal progenitor cells (RPCs), and its significant effects on the hierarchical structure and functionality of the retina. We also investigate the essential contributions of YAP in maintaining retinal homeostasis, highlighting its precise regulation of retinal cell proliferation and survival. In terms of retinal-related diseases, we explore the epigenetic connections and pathophysiological regulation of YAP in diabetic retinopathy (DR), glaucoma, and proliferative vitreoretinopathy (PVR). Lastly, we broaden our exploration from the retina to the brain, emphasizing the research paradigm of "retina: a window to the brain." Special focus is given to the emerging studies on YAP in brain disorders such as Alzheimer's disease (AD) and Parkinson's disease (PD), underlining its potential therapeutic value in neurodegenerative disorders and neuroinflammation.
Collapse
Affiliation(s)
- Yaqin Zhao
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan 430065, China; College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Bin Sun
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan 430065, China; College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Xuefei Fu
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan 430065, China; College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Zhuan Zuo
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan 430065, China; College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Huan Qin
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan 430065, China; College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China.
| | - Kai Yao
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan 430065, China; College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China.
| |
Collapse
|
9
|
Cai Y, Zhu J, Zhu L, Hong L, Zhang J, Kong L, Chen C, Luo J. Physalin H ameliorates LPS-induced acute lung injury via KEAP1/NRF2 axis. Int Immunopharmacol 2024; 131:111789. [PMID: 38484668 DOI: 10.1016/j.intimp.2024.111789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 02/19/2024] [Accepted: 02/29/2024] [Indexed: 04/10/2024]
Abstract
Physalin H (PH), a withanolide isolated from Physalisangulata L. has been reported to have anti-inflammatory effect. However, its impact on acute lung injury (ALI) remains unexplored. In this study, we observed that PH significantly alleviated inflammation in LPS-stimulated macrophages by suppressing the release of proinflammatory cytokines (TNF-α, IL-1β, and IL-6) and down-regulating the expression of the inflammation-related genes. RNA sequencing analysis revealed a significant up-regulation of the NRF2 pathway by PH. Further investigation elucidated that PH attenuated the ubiquitination of NRF2 by impeding the interaction between NRF2 and KEAP1, thereby facilitating NRF2 nuclear translocation and up-regulating the expression of target genes. Consequently, it regulated redox system and exerted anti-inflammatory effect. Consistently, PH also significantly alleviated pathological damage and inflammation in LPS-induced ALI mice model, which could be reversed by administration of an NRF2 inhibitor. Collectively, these results suggest that PH ameliorates ALI by activating the KEAP1/NRF2 pathway. These findings provide a foundation for further development of pH as a new anti-inflammatory agent for ALI therapy.
Collapse
Affiliation(s)
- Yuxing Cai
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province, China
| | - Jiangmin Zhu
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province, China
| | - Ling Zhu
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province, China
| | - Lihong Hong
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province, China
| | - Jianfei Zhang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province, China
| | - Lingyi Kong
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province, China
| | - Chen Chen
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province, China.
| | - Jianguang Luo
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province, China.
| |
Collapse
|
10
|
Kang HW, Kim JH, Lee DE, Lee YS, Kim MJ, Kim HS, Fang S, Lee BE, Lee KJ, Yoo J, Kim HJ, Park JS. Combination therapy of niclosamide with gemcitabine inhibited cell proliferation and apoptosis via Wnt/β-catenin/c-Myc signaling pathway by inducing β-catenin ubiquitination in pancreatic cancer. Cancer Biol Ther 2023; 24:2272334. [PMID: 37917550 PMCID: PMC10623893 DOI: 10.1080/15384047.2023.2272334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 10/10/2023] [Indexed: 11/04/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a type of cancer with high morbidity and mortality rates worldwide. Owing to a lack of therapeutic options, the overall survival rate of patients with pancreatic cancer is low. Gemcitabine has been mainly used to treat patients with pancreatic cancer, but its efficacy is limited by chemoresistance. Therefore, a novel therapeutic agent for PDAC therapy is urgently needed. An anthelminthic drug, niclosamide, has already been researched in breast, lung, colon, and pancreatic cancer as an anti-cancer purpose by re-positioning its original purpose. However, combination therapy of gemcitabine and niclosamide was not informed yet. Here, we found that niclosamide co-administered with gemcitabine significantly inhibited tumorigenesis of pancreatic cancer compared to gemcitabine alone. Further, combining niclosamide and gemcitabine inhibited cell proliferation and induced apoptosis. Niclosamide induced cell cycle arrest at the G1 phase, and the levels of CDK4/6 and cyclin D1 were lowered after gemcitabine treatment. In addition, the combination of these chemical compounds more effectively increased the binding level of activated β-catenin destruction complex and β-catenin to enable phosphorylation, compared to gemcitabine alone. After phosphorylation, niclosamide - gemcitabine upregulated the ubiquitin level, which caused phosphorylated β-catenin to undergo proteasomal degradation; the combination was more potent than gemcitabine alone. Finally, the combination more effectively suppressed tumor growth in vivo, compared to gemcitabine alone. Altogether, our results indicate that niclosamide synergistically enhances the antitumor effect of gemcitabine in pancreatic cancer, by inducing the degradation of β-catenin with ubiquitination. Therefore, this drug combination can potentially be used in PDAC therapy.
Collapse
Affiliation(s)
- Hyeon Woong Kang
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, Republic of Korea
| | - Ju Hyun Kim
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Da Eun Lee
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yun Sun Lee
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Myeong Jin Kim
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, Republic of Korea
| | - Hyung Sun Kim
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - SungSoon Fang
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, Republic of Korea
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Bo Eun Lee
- CHA Organoid Research Center, School of Medicine, CHA University, Seoul, Republic of Korea
- ORGANOIDSCIENCES, Ltd, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Kyung Jin Lee
- CHA Organoid Research Center, School of Medicine, CHA University, Seoul, Republic of Korea
- ORGANOIDSCIENCES, Ltd, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Jongman Yoo
- CHA Organoid Research Center, School of Medicine, CHA University, Seoul, Republic of Korea
- ORGANOIDSCIENCES, Ltd, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Hyo Jung Kim
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Joon Seong Park
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
11
|
Lin Q, Cao J, Yu J, Zhu Y, Shen Y, Wang S, Wang Y, Liu Z, Chang Y. YAP-mediated trophoblast dysfunction: the common pathway underlying pregnancy complications. Cell Commun Signal 2023; 21:353. [PMID: 38098027 PMCID: PMC10722737 DOI: 10.1186/s12964-023-01371-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 10/29/2023] [Indexed: 12/17/2023] Open
Abstract
Yes-associated protein (YAP) is a pivotal regulator in cellular proliferation, survival, differentiation, and migration, with significant roles in embryonic development, tissue repair, and tumorigenesis. At the maternal-fetal interface, emerging evidence underscores the importance of precisely regulated YAP activity in ensuring successful pregnancy initiation and progression. However, despite the established association between YAP dysregulation and adverse pregnancy outcomes, insights into the impact of aberrant YAP levels in fetal-derived, particularly trophoblast cells, and the ensuing dysfunction at the maternal-fetal interface remain limited. This review comprehensively examines YAP expression and its regulatory mechanisms in trophoblast cells throughout pregnancy. We emphasize its integral role in placental development and maternal-fetal interactions and delve into the correlations between YAP dysregulation and pregnancy complications. A nuanced understanding of YAP's functions during pregnancy could illuminate intricate molecular mechanisms and pave the way for innovative prevention and treatment strategies for pregnancy complications. Video Abstract.
Collapse
Affiliation(s)
- Qimei Lin
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Nankai University Affiliated Maternity Hospital, Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin, 300100, China
| | - Jiasong Cao
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Nankai University Affiliated Maternity Hospital, Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin, 300100, China
| | - Jing Yu
- School of Clinical Medicine, Tianjin Medical University, Tianjin, 300070, China
| | - Yu Zhu
- School of Clinical Medicine, Tianjin Medical University, Tianjin, 300070, China
| | - Yongmei Shen
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Nankai University Affiliated Maternity Hospital, Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin, 300100, China
| | - Shuqi Wang
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Nankai University Affiliated Maternity Hospital, Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin, 300100, China
| | - Yixin Wang
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Zhen Liu
- Academy of Clinical Medicine, Medical College, Tianjin University, Tianjin, 300072, China
| | - Ying Chang
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Nankai University Affiliated Maternity Hospital, Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin, 300100, China.
- Academy of Clinical Medicine, Medical College, Tianjin University, Tianjin, 300072, China.
| |
Collapse
|
12
|
Zhang N, Chen R, Cao X, Wang L. Aberrantly expressed HIF-1α enhances HCC stem cell-like traits via Wnt/β-catenin signaling activation after insufficient radiofrequency ablation. J Cancer Res Ther 2023; 19:1517-1524. [PMID: 38156917 DOI: 10.4103/jcrt.jcrt_1458_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 07/01/2023] [Indexed: 01/03/2024]
Abstract
BACKGROUND Radiofrequency ablation has become a favorable treatment modality for small hepatocellular carcinoma (HCC) recently; however, insufficient radiofrequency ablation (RFA) was shown to lead to enhanced invasiveness and metastasis of HCC in our previous study, while the underlying molecular mechanism has not been understood. MATERIALS AND METHODS In order to explore the influence of the hypoxic microenvironment on residual cancer and cancer stem cell (CSC)-like characteristics of HCC cells in this process, an in vitro hypoxic model and an insufficient RFA mouse model were established with HCC cancer cell lines. Immunochemistry staining and western blot were used to examine the expression of hypoxia-inducible factor (HIF)-1α and liver CSC markers. The 3D colon formation assay, tumor cell invasion assay, and gene transfection assays were applied to test the change in liver CSC stemness and HCC cell invasion. RESULTS After insufficient RFA treatment, the upregulated HIF-1α expression was associated with an increase in the CSC-like population in residual cancer. In vitro, hypoxic tumor cells showed aggressive CSC-like properties and phenotypes. Wnt/β-catenin signaling activation was shown to be necessary for the acquisition of liver CSC-like characteristics under hypoxic conditions. CONCLUSION Overall, the aberrantly enhanced HIF-1α expression enhanced the liver CSC-like traits via abnormal Wnt/β-catenin signaling activation after insufficient RFA, and the overexpressed HIF-1α would be a vital factor and useful biomarker during the HCC recurrence and metastasis.
Collapse
Affiliation(s)
- Ning Zhang
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Ruoxue Chen
- Zhongshan Hospital Institute of Clinical Science, Fudan University Shanghai Medical College, Shanghai, China
| | - Xin Cao
- Zhongshan Hospital Institute of Clinical Science, Fudan University Shanghai Medical College, Shanghai, China
| | - Lu Wang
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| |
Collapse
|
13
|
Koeberle SC, Kipp AP, Stuppner H, Koeberle A. Ferroptosis-modulating small molecules for targeting drug-resistant cancer: Challenges and opportunities in manipulating redox signaling. Med Res Rev 2023; 43:614-682. [PMID: 36658724 PMCID: PMC10947485 DOI: 10.1002/med.21933] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/07/2022] [Accepted: 01/03/2023] [Indexed: 01/21/2023]
Abstract
Ferroptosis is an iron-dependent cell death program that is characterized by excessive lipid peroxidation. Triggering ferroptosis has been proposed as a promising strategy to fight cancer and overcome drug resistance in antitumor therapy. Understanding the molecular interactions and structural features of ferroptosis-inducing compounds might therefore open the door to efficient pharmacological strategies against aggressive, metastatic, and therapy-resistant cancer. We here summarize the molecular mechanisms and structural requirements of ferroptosis-inducing small molecules that target central players in ferroptosis. Focus is placed on (i) glutathione peroxidase (GPX) 4, the only GPX isoenzyme that detoxifies complex membrane-bound lipid hydroperoxides, (ii) the cystine/glutamate antiporter system Xc - that is central for glutathione regeneration, (iii) the redox-protective transcription factor nuclear factor erythroid 2-related factor (NRF2), and (iv) GPX4 repression in combination with induced heme degradation via heme oxygenase-1. We deduce common features for efficient ferroptotic activity and highlight challenges in drug development. Moreover, we critically discuss the potential of natural products as ferroptosis-inducing lead structures and provide a comprehensive overview of structurally diverse biogenic and bioinspired small molecules that trigger ferroptosis via iron oxidation, inhibition of the thioredoxin/thioredoxin reductase system or less defined modes of action.
Collapse
Affiliation(s)
- Solveigh C. Koeberle
- Michael Popp Institute, Center for Molecular Biosciences Innsbruck (CMBI)University of InnsbruckTirolInnsbruckAustria
- Department of Molecular Nutritional Physiology, Institute of Nutritional SciencesFriedrich Schiller University JenaThüringenJenaGermany
| | - Anna P. Kipp
- Department of Molecular Nutritional Physiology, Institute of Nutritional SciencesFriedrich Schiller University JenaThüringenJenaGermany
| | - Hermann Stuppner
- Unit of Pharmacognosy, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck (CMBI)University of InnsbruckTirolInnsbruckAustria
| | - Andreas Koeberle
- Michael Popp Institute, Center for Molecular Biosciences Innsbruck (CMBI)University of InnsbruckTirolInnsbruckAustria
| |
Collapse
|
14
|
Khan AU, Khan A, Shal B, Khan S, Khan M, Ahmad R, Riaz M. The critical role of the phytosterols in modulating tumor microenvironment via multiple signaling: A comprehensive molecular approach. Phytother Res 2023; 37:1606-1623. [PMID: 36757068 DOI: 10.1002/ptr.7755] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/30/2022] [Accepted: 01/02/2023] [Indexed: 02/10/2023]
Abstract
Cancer is the leading cause of mortality and morbidity worldwide, and its cases are rapidly increasing every year. Several factors contribute to the development of tumorigenesis. including radiation, dietary lifestyle, smoking, environmental, and genetic factors. The cell cycle is regulated by a variety of molecular signaling proteins. However, when the proteins involved in the cell cycle regulation are altered, cellular growth and proliferation are significantly affected. Natural products provide an important source of new drug development for a variety of ailments. including cancer. Phytosterols (PSs) are an important class of natural compounds reported for numerous pharmacological activities, including cancer. Various PSs, such as ergosterol, stigmasterol, sitosterol, withaferin A, etc., have been reported for their anti-cancer activities against a variety of cancer by modulating the tumor microenvironment via molecular signaling pathways discussed within the article. These signaling pathways are associated with the production of pro-inflammatory mediators, growth factors, chemokines, and pro-apoptotic and anti-apoptotic genes. These mediators and their upstream signaling are very active within the variety of tumors and by modulating these signalings, thus PS exhibits promising anti-cancer activities. However, further high-quality studies are needed to firmly establish the clinical efficacy as well the safety of the phytosterols.
Collapse
Affiliation(s)
- Ashraf Ullah Khan
- Pharmacological Sciences Research Lab, Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.,Faculty of Pharmaceutical Sciences, Abasyn University, Peshawar, Pakistan
| | - Adnan Khan
- Pharmacological Sciences Research Lab, Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Bushra Shal
- Pharmacological Sciences Research Lab, Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.,Faculty of Health Sciences, IQRA University, Islamabad Campus, (Chak Shahzad), Islamabad, Pakistan
| | - Salman Khan
- Pharmacological Sciences Research Lab, Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Majid Khan
- Faculty of Pharmaceutical Sciences, Abasyn University, Peshawar, Pakistan
| | - Rizwan Ahmad
- Natural Products & Alternative Medicines College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, Dammam, Kingdom of Saudi Arabia
| | - Muhammad Riaz
- Department of Pharmacy, Shaheed Benazir Bhutto University Sheringal, Sheringal, Pakistan
| |
Collapse
|
15
|
Ma FY, Zhou XH, Liang Q. Advances in understanding of role and mechanism of Hippo signaling pathway in colorectal cancer. Shijie Huaren Xiaohua Zazhi 2023; 31:14-19. [DOI: 10.11569/wcjd.v31.i1.14] [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: 01/07/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most common malignant tumors, and most patients have a poor prognosis. Many studies have shown that the Hippo signaling pathway plays a key role in the occurrence and development of CRC by regulating CRC cell proliferation and apoptosis, tumor invasion and metastasis, autophagy, metabolic reprogramming, drug resistance, and other processes. This article reviews the latest progress in research of the expression of key molecules of the Hippo signaling pathway in CRC as well as the understanding of the mechanism by which this pathway regulates the occurrence and development of CRC.
Collapse
Affiliation(s)
- Fu-Yan Ma
- Graduate School of Youjiang Medical College for Nationalities, Baise 533000, Guangxi Zhuang Autonomous Region, China
| | - Xi-Han Zhou
- Department of Gastroenterology, Affiliated Hospital of Youjiang Medical College Nationalities, Baise 533000, Guangxi Zhuang Autonomous Region, China
| | - Qiao Liang
- Graduate School of Youjiang Medical College for Nationalities, Baise 533000, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
16
|
Naktubtim C, Payuhakrit W, Uttarawichien T, Hassametto A, Suwannalert P. YAP, a novel target regulates F-actin rearrangement-associated CAFs transformation and promotes colorectal cancer cell progression. Biomed Pharmacother 2022; 155:113757. [DOI: 10.1016/j.biopha.2022.113757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/16/2022] [Accepted: 09/26/2022] [Indexed: 11/02/2022] Open
|
17
|
Wang M, Liu X, Chen T, Cheng X, Xiao H, Meng X, Jiang Y. Inhibition and potential treatment of colorectal cancer by natural compounds via various signaling pathways. Front Oncol 2022; 12:956793. [PMID: 36158694 PMCID: PMC9496650 DOI: 10.3389/fonc.2022.956793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 07/15/2022] [Indexed: 11/13/2022] Open
Abstract
Colorectal cancer (CRC) is a common type of malignant digestive tract tumor with a high incidence rate worldwide. Currently, the clinical treatment of CRC predominantly include surgical resection, postoperative chemotherapy, and radiotherapy. However, these treatments contain severe limitations such as drug side effects, the risk of recurrence and drug resistance. Some natural compounds found in plants, fungi, marine animals, and bacteria have been shown to inhibit the occurrence and development of CRC. Although the explicit molecular mechanisms underlying the therapeutic effects of these compounds on CRC are not clear, classical signaling transduction pathways such as NF-kB and Wnt/β-catenin are extensively regulated. In this review, we have summarized the specific mechanisms regulating the inhibition and development of CRC by various types of natural compounds through nine signaling pathways, and explored the potential therapeutic values of these natural compounds in the clinical treatment of CRC.
Collapse
Affiliation(s)
- Mingchuan Wang
- Department of Gastrointestinal Colorectal and Anal Surgery, The China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xianjun Liu
- College of Food Engineering, Jilin Engineering Normal University, Changchun, China
| | - Tong Chen
- Department of Gastrointestinal Colorectal and Anal Surgery, The China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xianbin Cheng
- Department of Thyroid Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Huijie Xiao
- Department of Gastrointestinal Colorectal and Anal Surgery, The China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xianglong Meng
- Department of Burns Surgery, The First Hospital of Jilin University, Changchun, China
| | - Yang Jiang
- Department of Gastrointestinal Colorectal and Anal Surgery, The China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
18
|
YAP and TAZ: Monocorial and bicorial transcriptional co-activators in human cancers. Biochim Biophys Acta Rev Cancer 2022; 1877:188756. [PMID: 35777600 DOI: 10.1016/j.bbcan.2022.188756] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 06/09/2022] [Accepted: 06/23/2022] [Indexed: 12/17/2022]
Abstract
The transcriptional regulators YAP and TAZ are involved in numerous physiological processes including organ development, growth, immunity and tissue regeneration. YAP and TAZ dysregulation also contribute to tumorigenesis, thereby making them attractive cancer therapeutic targets. Arbitrarily, YAP and TAZ are often considered as a single protein, and are referred to as YAP/TAZ in most studies. However, increasing experimental evidences documented that YAP and TAZ perform both overlapping and distinct functions in several physiological and pathological processes. In addition to regulating distinct processes, YAP and TAZ are also regulated by distinct upstream cues. The aim of the review is to describe the distinct roles of YAP and TAZ focusing particularly on cancer. Therapeutic strategies targeting either YAP and TAZ proteins or only one of them should be carefully evaluated. Selective targeting of YAP or TAZ may in fact impair different pathways and determine diverse clinical outputs.
Collapse
|
19
|
Meira CS, Soares JWC, Dos Reis BPZC, Pacheco LV, Santos IP, Silva DKC, de Lacerda JC, Daltro SRT, Guimarães ET, Soares MBP. Therapeutic Applications of Physalins: Powerful Natural Weapons. Front Pharmacol 2022; 13:864714. [PMID: 35450054 PMCID: PMC9016203 DOI: 10.3389/fphar.2022.864714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/18/2022] [Indexed: 11/28/2022] Open
Abstract
Physalins, or 16,24-cyclo-13,14-seco steroids, are compounds belonging to the class of withanolides that can be found in plants of Solanaceae family, mainly in species belonging to the genus Physalis spp., which are annual herbaceous plants widely distributed in tropical and subtropical regions of the world. Physalins are versatile molecules that act in several cell signaling pathways and activate different mechanisms of cell death or immunomodulation. A number of studies have shown a variety of actions of these compounds, including anticancer, anti-inflammatory, antiparasitic, antimicrobial, antinociceptive, and antiviral activities. Here we reviewed the main findings related to the anticancer, immunomodulatory, and antiparasitic activities of physalins and its mechanisms of action, highlighting the \challenges and future directions in the pharmacological application of physalins.
Collapse
Affiliation(s)
- Cássio Santana Meira
- SENAI Institute of Innovation in Health Advanced Systems (CIMATEC ISI SAS), University Center SENAI/CIMATEC, Salvador, Brazil.,Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Brazil.,Department of Life Sciences, State University of Bahia (UNEB), Salvador, Brazil
| | | | | | | | | | | | - Julia Costa de Lacerda
- Bahiana School of Medicine and Public Health, Bahiana Foundation for the Development of Sciences, Salvador, Brazil
| | | | - Elisalva Teixeira Guimarães
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Brazil.,Department of Life Sciences, State University of Bahia (UNEB), Salvador, Brazil
| | - Milena Botelho Pereira Soares
- SENAI Institute of Innovation in Health Advanced Systems (CIMATEC ISI SAS), University Center SENAI/CIMATEC, Salvador, Brazil.,Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Brazil
| |
Collapse
|
20
|
Yang J, Sun Y, Cao F, Yang B, Kuang H. Natural Products from Physalis alkekengi L. var. franchetii (Mast.) Makino: A Review on Their Structural Analysis, Quality Control, Pharmacology, and Pharmacokinetics. Molecules 2022; 27:molecules27030695. [PMID: 35163960 PMCID: PMC8840080 DOI: 10.3390/molecules27030695] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/15/2022] [Accepted: 01/17/2022] [Indexed: 12/21/2022] Open
Abstract
The calyxes and fruits of Physalis alkekengi L. var. franchetii (Mast.) Makino (P. alkekengi), a medicinal and edible plant, are frequently used as heat-clearing and detoxifying agents in thousands of Chinese medicine prescriptions. For thousands of years in China, they have been widely used in clinical practice to treat throat disease, hepatitis, and bacillary dysentery. This systematic review summarizes their structural analysis, quality control, pharmacology, and pharmacokinetics. Furthermore, the possible development trends and perspectives for future research studies on this medicinal plant are discussed. Relevant information on the calyxes and fruits of P. alkekengi was collected from electronic databases, Chinese herbal classics, and Chinese Pharmacopoeia. Moreover, information was collected from ancient documents in China. The components isolated and identified in P. alkekengi include steroids, flavonoids, phenylpropanoids, alkaloids, nucleosides, terpenoids, megastigmane, aliphatic derivatives, organic acids, coumarins, and sucrose esters. Steroids, particularly physalins and flavonoids, are the major characteristic and bioactive ingredients in P. alkekengi. According to the literature, physalins are synthesized by the mevalonate and 2-C-methyl-d-erythritol-4-phosphate pathways, and flavonoids are synthesized by the phenylpropanoid pathway. Since the chemical components and pharmacological effects of P. alkekengi are complex and varied, there are different standards for the evaluation of its quality and efficacy. In most cases, the analysis was performed using high-performance liquid chromatography coupled with ultraviolet detection. A pharmacological study showed that the crude extracts and isolated compounds from P. alkekengi had extensive in vitro and in vivo biological activities (e.g., anti-inflammatory, anti-tumor, immunosuppressive, antibacterial, anti-leishmanial, anti-asthmatic, anti-diabetic, anti-oxidative, anti-malarial, anti-Alzheimer's disease, and vasodilatory). Moreover, the relevant anti-inflammatory and anti-tumor mechanisms were elucidated. The reported activities indicate the great pharmacological potential of P. alkekengi. Similarly, studies on the pharmacokinetics of specific compounds will also contribute to the progress of clinical research in this setting.
Collapse
Affiliation(s)
- Jing Yang
- Key Laboratory of Basic and Application Research of Beiyao, Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin 150040, China; (J.Y.); (Y.S.); (B.Y.)
| | - Yanping Sun
- Key Laboratory of Basic and Application Research of Beiyao, Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin 150040, China; (J.Y.); (Y.S.); (B.Y.)
| | - Feng Cao
- Ganjiang Chinese Medicine Innovation Center, Nanchang 330000, China;
| | - Bingyou Yang
- Key Laboratory of Basic and Application Research of Beiyao, Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin 150040, China; (J.Y.); (Y.S.); (B.Y.)
| | - Haixue Kuang
- Key Laboratory of Basic and Application Research of Beiyao, Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin 150040, China; (J.Y.); (Y.S.); (B.Y.)
- Correspondence: ; Tel.: +86-0451-82197188
| |
Collapse
|
21
|
Hu Y, Wang Q, Yu J, Zhou Q, Deng Y, Liu J, Zhang L, Xu Y, Xiong W, Wang Y. Tartrate-resistant acid phosphatase 5 promotes pulmonary fibrosis by modulating β-catenin signaling. Nat Commun 2022; 13:114. [PMID: 35013220 PMCID: PMC8748833 DOI: 10.1038/s41467-021-27684-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 12/02/2021] [Indexed: 02/07/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal interstitial lung disease with limited therapeutic options. Tartrate-resistant acid phosphatase 5 (ACP5) performs a variety of functions. However, its role in IPF remains unclear. Here, we demonstrate that the levels of ACP5 are increased in IPF patient samples and mice with bleomycin (BLM)-induced pulmonary fibrosis. In particular, higher levels of ACP5 are present in the sera of IPF patients with a diffusing capacity of the lungs for carbonmonoxide (DLCO) less than 40% of the predicted value. Additionally, Acp5 deficiency protects mice from BLM-induced lung injury and fibrosis coupled with a significant reduction of fibroblast differentiation and proliferation. Mechanistic studies reveal that Acp5 is upregulated by transforming growth factor-β1 (TGF-β1) in a TGF-β receptor 1 (TGFβR1)/Smad family member 3 (Smad3)-dependent manner, after which Acp5 dephosphorylates p-β-catenin at serine 33 and threonine 41, inhibiting the degradation of β-catenin and subsequently enhancing β-catenin signaling in the nucleus, which promotes the differentiation, proliferation and migration of fibroblast. More importantly, the treatment of mice with Acp5 siRNA-loaded liposomes or Acp5 inhibitor reverses established lung fibrosis. In conclusions, Acp5 is involved in the initiation and progression of pulmonary fibrosis and strategies aimed at silencing or suppressing Acp5 could be considered as potential therapeutic approaches against pulmonary fibrosis. Idiopathic pulmonary fibrosis is a fatal lung disease with limited treatment options. Here the authors show that tartrate-resistant acid phosphatase 5 (Acp5) promotes lung fibrosis by enhancing beta-catenin signaling and that inhibition of Acp5 can reverse stablished pulmonary fibrosis.
Collapse
Affiliation(s)
- Yinan Hu
- Department of Pulmonary and Critical Care Medicine, NHC Key Laboratory of Respiratory Diseases, Key Site of National Clinical Research Center for Respiratory Disease, Wuhan Clinical Medical Research Center for Chronic Airway Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China.,Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, National Center for Respiratory Medicine, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, National Clinical Research Center for Respiratory Diseases, China-Japan Friendship Hospital, 100029, Beijing, China
| | - Qi Wang
- Department of Pulmonary and Critical Care Medicine, NHC Key Laboratory of Respiratory Diseases, Key Site of National Clinical Research Center for Respiratory Disease, Wuhan Clinical Medical Research Center for Chronic Airway Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China
| | - Jun Yu
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China
| | - Qing Zhou
- The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China
| | - Yanhan Deng
- Department of Pulmonary and Critical Care Medicine, NHC Key Laboratory of Respiratory Diseases, Key Site of National Clinical Research Center for Respiratory Disease, Wuhan Clinical Medical Research Center for Chronic Airway Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China
| | - Juan Liu
- Department of Pulmonary and Critical Care Medicine, NHC Key Laboratory of Respiratory Diseases, Key Site of National Clinical Research Center for Respiratory Disease, Wuhan Clinical Medical Research Center for Chronic Airway Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China
| | - Lei Zhang
- Department of Pulmonary and Critical Care Medicine, NHC Key Laboratory of Respiratory Diseases, Key Site of National Clinical Research Center for Respiratory Disease, Wuhan Clinical Medical Research Center for Chronic Airway Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China
| | - Yongjian Xu
- Department of Pulmonary and Critical Care Medicine, NHC Key Laboratory of Respiratory Diseases, Key Site of National Clinical Research Center for Respiratory Disease, Wuhan Clinical Medical Research Center for Chronic Airway Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China
| | - Weining Xiong
- Department of Pulmonary and Critical Care Medicine, NHC Key Laboratory of Respiratory Diseases, Key Site of National Clinical Research Center for Respiratory Disease, Wuhan Clinical Medical Research Center for Chronic Airway Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China. .,Department of Pulmonary and Critical Care Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, 639 Zhizaoju Lu, Shanghai, 200011, China.
| | - Yi Wang
- Department of Pulmonary and Critical Care Medicine, NHC Key Laboratory of Respiratory Diseases, Key Site of National Clinical Research Center for Respiratory Disease, Wuhan Clinical Medical Research Center for Chronic Airway Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China.
| |
Collapse
|
22
|
Niu JB, Hua CQ, Liu Y, Yu GX, Yang JJ, Li YR, Zhang YB, Qi YQ, Song J, Jin CY, Zhang SY. Discovery of N-aryl sulphonamide-quinazoline derivatives as anti-gastric cancer agents in vitro and in vivo via activating the Hippo signalling pathway. J Enzyme Inhib Med Chem 2021; 36:1715-1731. [PMID: 34425716 PMCID: PMC8386742 DOI: 10.1080/14756366.2021.1958211] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 06/17/2021] [Accepted: 07/16/2021] [Indexed: 01/07/2023] Open
Abstract
Hippo signalling pathway plays a crucial role in tumorigenesis and cancer progression. In this work, we identified an N-aryl sulphonamide-quinazoline derivative, compound 9i as an anti-gastric cancer agent, which exhibited potent antiproliferative ability with IC50 values of 0.36 μM (MGC-803 cells), 0.70 μM (HCT-116 cells), 1.04 μM (PC-3 cells), and 0.81 μM (MCF-7 cells), respectively and inhibited YAP activity by the activation of p-LATS. Compound 9i was effective in suppressing MGC-803 xenograft tumour growth in nude mice without obvious toxicity and significantly down-regulated the expression of YAP in vivo. Compound 9i arrested cells in the G2/M phase, induced intrinsic apoptosis, and inhibited cell colony formation in MGC-803 and SGC-7901 cells. Therefore, compound 9i is to be reported as an anti-gastric cancer agent via activating the Hippo signalling pathway and might help foster a new strategy for the cancer treatment by activating the Hippo signalling pathway regulatory function to inhibit the activity of YAP.
Collapse
Affiliation(s)
- Jin-Bo Niu
- The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chun-Quan Hua
- School of Pharmaceutical Sciences, Institute of Drug Discovery & Development, Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou, China
| | - Yuan Liu
- School of Pharmaceutical Sciences, Institute of Drug Discovery & Development, Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou, China
| | - Guang-Xi Yu
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jia-Jia Yang
- Department of Pharmacy, Zhengzhou People's Hospital, Zhengzhou, China
| | - Yin-Ru Li
- The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yan-Bing Zhang
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Ying-Qiu Qi
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jian Song
- School of Pharmaceutical Sciences, Institute of Drug Discovery & Development, Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou, China
| | - Cheng-Yun Jin
- The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- School of Pharmaceutical Sciences, Institute of Drug Discovery & Development, Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou, China
| | - Sai-Yang Zhang
- The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
23
|
Pyrvinium pamoate regulates MGMT expression through suppressing the Wnt/β-catenin signaling pathway to enhance the glioblastoma sensitivity to temozolomide. Cell Death Discov 2021; 7:288. [PMID: 34642308 PMCID: PMC8511032 DOI: 10.1038/s41420-021-00654-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 08/23/2021] [Accepted: 09/13/2021] [Indexed: 12/16/2022] Open
Abstract
Temozolomide (TMZ) is the mainstream chemotherapeutic drug for treating glioblastoma multiforme (GBM), but the intrinsic or acquired chemoresistance to TMZ has become the leading clinical concern, which is related to the repair of DNA alkylation sites by O6-methylguanine-DNA methyltransferase (MGMT). Pyrvinium pamoate (PP), the FDA-approved anthelminthic drug, has been reported to inhibit the Wnt/β-catenin pathway within numerous cancer types, and Wnt/β-catenin signaling pathway can modulate the expression of MGMT gene. However, whether PP affects the expression of MGMT and enhances TMZ sensitivity in GBM cells remains unclear. In the present study, we found that PP and TMZ had synergistic effect on inhibiting the viability of GBM cells, and PP induced inhibition of MGMT and enhanced the TMZ chemosensitivity of GBM cells through down-regulating Wnt/β-catenin pathway. Moreover, the overexpression of MGMT or β-catenin weakened the synergy between PP and TMZ. The mechanism of PP in inhibiting the Wnt pathway was indicated that PP resulted in the degradation of β-catenin via the AKT/GSK3β/β-catenin signaling axis. Moreover, Ser552 phosphorylation in β-catenin, which promotes its nuclear accumulation and transcriptional activity, is blocked by PP that also inhibits the Wnt pathway to some extent. The intracranial GBM mouse model also demonstrated that the synergy between PP and TMZ could be achieved through down-regulating β-catenin and MGMT, which prolonged the survival time of tumor-bearing mice. Taken together, our data suggest that PP may serve as the prospect medicine to improve the chemotherapeutic effect on GBM, especially for chemoresistant to TMZ induced by MGMT overexpression.
Collapse
|
24
|
Al-Hakeim HK, Asad HN, Maes M. Wnt/β-catenin pathway proteins in end-stage renal disease. Biomark Med 2021; 15:1423-1434. [PMID: 34554011 DOI: 10.2217/bmm-2021-0177] [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] [Indexed: 02/03/2023] Open
Abstract
Aim: To delineate the association of end-stage renal disease (ESRD) and Wnt-proteins including the agonist R-spondin-1, the transducer β-catenin and the antagonists DKK1 and sclerostin. Materials & methods: Serum Wnt-pathway proteins levels were measured by ELISA in 60 ESRD patients and 30 normal controls. Results: DKK1 and sclerostin were significantly higher in ESRD than in controls, and β-catenin and the catenin + R-spondin-1/DKK1 + sclerostin ratio, reflecting the ratio of agonist and transducer on antagonists (AT/ANTA), were significantly lower in ESRD. Estimated glomerular filtration rate was significantly associated with DKK1 and sclerostin (inversely), β-catenin (positively) and the AT/ANTA ratio (r = 0.468, p < 0.001). Conclusion: Wnt/β-catenin pathway proteins show significant alterations in ESRD, indicating significantly increased levels of antagonists.
Collapse
Affiliation(s)
| | - Halah N Asad
- Al Najaf Health Directorate, Higher Health Institute, Najaf, Iraq
| | - Michael Maes
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.,Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria.,School of Medicine, IMPACT Strategic Research Centre, Deakin University, VIC, 3220, Australia
| |
Collapse
|
25
|
Mäntylä E, Ihalainen TO. Brick Strex: a robust device built of LEGO bricks for mechanical manipulation of cells. Sci Rep 2021; 11:18520. [PMID: 34531455 PMCID: PMC8445989 DOI: 10.1038/s41598-021-97900-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 08/30/2021] [Indexed: 02/08/2023] Open
Abstract
Cellular forces, mechanics and other physical factors are important co-regulators of normal cell and tissue physiology. These cues are often misregulated in diseases such as cancer, where altered tissue mechanics contribute to the disease progression. Furthermore, intercellular tensile and compressive force-related signaling is highlighted in collective cell behavior during development. However, the mechanistic understanding on the role of physical forces in regulation of cellular physiology, including gene expression and signaling, is still lacking. This is partly because studies on the molecular mechanisms of force transmission require easily controllable experimental designs. These approaches should enable both easy mechanical manipulation of cells and, importantly, readouts ranging from microscopy imaging to biochemical assays. To achieve a robust solution for mechanical manipulation of cells, we developed devices built of LEGO bricks allowing manual, motorized and/or cyclic cell stretching and compression studies. By using these devices, we show that [Formula: see text]-catenin responds differentially to epithelial monolayer stretching and lateral compression, either localizing more to the cell nuclei or cell-cell junctions, respectively. In addition, we show that epithelial compression drives cytoplasmic retention and phosphorylation of transcription coregulator YAP1. We provide a complete part listing and video assembly instructions, allowing other researchers to build and use the devices in cellular mechanics-related studies.
Collapse
Affiliation(s)
- Elina Mäntylä
- grid.502801.e0000 0001 2314 6254BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520 Tampere, Finland
| | - Teemu O. Ihalainen
- grid.502801.e0000 0001 2314 6254BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520 Tampere, Finland
| |
Collapse
|
26
|
Pehlivan M, Caliskan C, Yuce Z, Sercan HO. sFRP1 Expression Regulates Wnt signaling in Chronic Myeloid Leukemia K562 Cells. Anticancer Agents Med Chem 2021; 22:1354-1362. [PMID: 34030621 DOI: 10.2174/1871520621666210524162145] [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: 12/10/2020] [Revised: 03/23/2021] [Accepted: 04/12/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Wnt signaling cascades play important roles in cell fate decisions and their deregulation has been documented in many diseases, including malignant tumors and leukemia. One mechanism of aberrant Wnt signaling is the silencing of Wnt inhibitors through epigenetic mechanisms. The sFRPs are one of the most studied Wnt inhibitors; and the sFRP1 loss is known in many hematological malignancies. Therefore, we aimed to compare the expression of Wnt related genes in the presence and absence of sFRP1 in chronic myeloid leukemia (CML) cell line. OBJECTIVE It is important to understand how sFRP1 and sFRP1 perform on CML to design new agents and strategies for resistant and advanced forms of CML. MATERIALS AND METHODS We used K562 cells, which normally do not express sFRP1 and its sFRP1 expressing subclone K562s. Total RNA was isolated from K562 and K562s cell lines end converted cDNA. PCR Array experiments performed using Human Wnt Signaling Pathway Plus RT2 Profiler™ kit. Wnt signaling pathway activation was studied by western blot for downstream signaling targets. RESULTS The WNT3, LRP6, PRICKLE1 and BTRC expressions were significantly decreased in the presence of sFRP1; while WNT5B increased. The sFRP1 expression inhibited stabilization of total β-catenin protein and downstream effector phosphorylation of noncanonical Wnt/PCP signaling; whereas Ca2+/PKC signaling remained active. CONCLUSION
Collapse
Affiliation(s)
- Melek Pehlivan
- Vocational School of Health Services, Izmir Katip Celebi University, Izmir, Turkey
| | - Ceyda Caliskan
- Department of Biomedical Sciences, University of Sheffield, Sheffield, United Kingdom
| | - Zeynep Yuce
- Dokuz Eylul University, Department of Medical Biology and Genetics, Izmir. Turkey
| | - Hakki O Sercan
- Dokuz Eylul University, Department of Medical Biology and Genetics, Izmir. Turkey
| |
Collapse
|
27
|
Ortíz R, Quiñonero F, García-Pinel B, Fuel M, Mesas C, Cabeza L, Melguizo C, Prados J. Nanomedicine to Overcome Multidrug Resistance Mechanisms in Colon and Pancreatic Cancer: Recent Progress. Cancers (Basel) 2021; 13:2058. [PMID: 33923200 PMCID: PMC8123136 DOI: 10.3390/cancers13092058] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 04/22/2021] [Accepted: 04/22/2021] [Indexed: 12/24/2022] Open
Abstract
The development of drug resistance is one of the main causes of cancer treatment failure. This phenomenon occurs very frequently in different types of cancer, including colon and pancreatic cancers. However, the underlying molecular mechanisms are not fully understood. In recent years, nanomedicine has improved the delivery and efficacy of drugs, and has decreased their side effects. In addition, it has allowed to design drugs capable of avoiding certain resistance mechanisms of tumors. In this article, we review the main resistance mechanisms in colon and pancreatic cancers, along with the most relevant strategies offered by nanodrugs to overcome this obstacle. These strategies include the inhibition of efflux pumps, the use of specific targets, the development of nanomedicines affecting the environment of cancer-specific tissues, the modulation of DNA repair mechanisms or RNA (miRNA), and specific approaches to damage cancer stem cells, among others. This review aims to illustrate how advanced nanoformulations, including polymeric conjugates, micelles, dendrimers, liposomes, metallic and carbon-based nanoparticles, are allowing to overcome one of the main limitations in the treatment of colon and pancreatic cancers. The future development of nanomedicine opens new horizons for cancer treatment.
Collapse
Affiliation(s)
- Raúl Ortíz
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain; (R.O.); (F.Q.); (B.G.-P.); (M.F.); (C.M.); (L.C.); (J.P.)
- Department of Anatomy and Embriology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
- Instituto Biosanitario de Granada (ibs.GRANADA), 18014 Granada, Spain
| | - Francisco Quiñonero
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain; (R.O.); (F.Q.); (B.G.-P.); (M.F.); (C.M.); (L.C.); (J.P.)
- Department of Anatomy and Embriology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
- Instituto Biosanitario de Granada (ibs.GRANADA), 18014 Granada, Spain
| | - Beatriz García-Pinel
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain; (R.O.); (F.Q.); (B.G.-P.); (M.F.); (C.M.); (L.C.); (J.P.)
- Department of Anatomy and Embriology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
- Instituto Biosanitario de Granada (ibs.GRANADA), 18014 Granada, Spain
| | - Marco Fuel
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain; (R.O.); (F.Q.); (B.G.-P.); (M.F.); (C.M.); (L.C.); (J.P.)
- Instituto Biosanitario de Granada (ibs.GRANADA), 18014 Granada, Spain
| | - Cristina Mesas
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain; (R.O.); (F.Q.); (B.G.-P.); (M.F.); (C.M.); (L.C.); (J.P.)
- Department of Anatomy and Embriology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
- Instituto Biosanitario de Granada (ibs.GRANADA), 18014 Granada, Spain
| | - Laura Cabeza
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain; (R.O.); (F.Q.); (B.G.-P.); (M.F.); (C.M.); (L.C.); (J.P.)
- Department of Anatomy and Embriology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
- Instituto Biosanitario de Granada (ibs.GRANADA), 18014 Granada, Spain
| | - Consolación Melguizo
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain; (R.O.); (F.Q.); (B.G.-P.); (M.F.); (C.M.); (L.C.); (J.P.)
- Department of Anatomy and Embriology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
- Instituto Biosanitario de Granada (ibs.GRANADA), 18014 Granada, Spain
| | - Jose Prados
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain; (R.O.); (F.Q.); (B.G.-P.); (M.F.); (C.M.); (L.C.); (J.P.)
- Department of Anatomy and Embriology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
- Instituto Biosanitario de Granada (ibs.GRANADA), 18014 Granada, Spain
| |
Collapse
|
28
|
Yang Y, Xiang K, Sun D, Zheng M, Song Z, Li M, Wang X, Li H, Chen L. Withanolides from dietary tomatillo suppress HT1080 cancer cell growth by targeting mutant IDH1. Bioorg Med Chem 2021; 36:116095. [PMID: 33735687 DOI: 10.1016/j.bmc.2021.116095] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 02/19/2021] [Accepted: 02/20/2021] [Indexed: 11/16/2022]
Abstract
Isocitrate dehydrogenase (IDH) is one key rate-limiting enzyme in the tricarboxylic acid cycle, which is related to various cancers. Tomatillo (Physalis ixocarpa), a special tomato, is widely consumed as nutritious vegetable in Mexico, USA, etc. As a rich source for withanolides, the fruits of P. ixocarpa were investigated, leading to the isolation of 11 type-A withanolides including 4 new ones (1 is an artificial withanolide). All these withanolides were evaluated for their inhibition on mutant IDH1 enzyme activity. Among them, physalin F (11) exhibited potent enzyme inhibitory activity and binding affinity with mutant IDH1. It inhibits the proliferation of HT1080 cells by selectively inhibiting the activity of mutant IDH1. Since Ixocarpalactone A, another major type-B withanolide in this plant, could act on another energy metabolism target PHGDH, the presence of different types of withanolides in tomatillo and their synergistic effect could make it a potential antitumor functional food or drug.
Collapse
Affiliation(s)
- Yueying Yang
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Ke Xiang
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Dejuan Sun
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Mengzhu Zheng
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhuorui Song
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Mingxue Li
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xuanbin Wang
- Laboratory of Chinese Herbal Pharmacology, Oncology Center, Renmin Hospital, Biomedical Research Institute, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan 442000, China.
| | - Hua Li
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China; Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Lixia Chen
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China.
| |
Collapse
|
29
|
Mesas C, Fuel M, Martínez R, Prados J, Melguizo C, Porres JM. In vitro evidence of the antitumor capacity of Solanaceae and Cucurbitaceae in colon cancer: A systematic review. Crit Rev Food Sci Nutr 2021; 62:6293-6314. [PMID: 33739207 DOI: 10.1080/10408398.2021.1900058] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Colon cancer is the fourth leading cause of cancer deaths around the world. Despite advances in understanding its etiology and in diagnosis and treatment, new therapeutic strategies are still required. In this sense, the Solanaceae and Cucurbitaceae families have been widely used to treat various pathologies, including cancer, for their bioactive components. The objective of this systematic review was to analyze the antitumor activity of the bioactive components present in extracts from Solanaceae and Cucurbitaceae families using different in in vitro models of colon cancer. 241 publications have been identified (published from January 2008 to January 2020) from different electronic data base. 44 articles were included, 26 of which examined the Solanaceae family. The antitumor activity exhibited by this family was due to the withanolide-type steroid compounds they harbor. 18 articles were related to the Cucurbitaceae family. This family is characterized by their production of cucurbitacin-type triterpenoid compounds and their derivatives, which confer antitumor activity. In conclusion, the different genera belonging to both families are an important source of bioactive compounds with relevant activity against colon cancer. More experimental and in vivo studies will be required to corroborate their antitumor activity and to leverage them in future clinical practice.
Collapse
Affiliation(s)
- Cristina Mesas
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, Spain.,Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Marco Fuel
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Rosario Martínez
- Department of Physiology, Institute of Nutrition and Food Technology (INyTA), Center of Biomedical Research (CIBM), University of Granada, Granada, Spain
| | - Jose Prados
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, Spain.,Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Consolación Melguizo
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, Spain.,Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Jesús M Porres
- Department of Physiology, Institute of Nutrition and Food Technology (INyTA), Center of Biomedical Research (CIBM), University of Granada, Granada, Spain
| |
Collapse
|
30
|
Jiang L, Zhang J, Xu Q, Wang B, Yao Y, Sun L, Wang X, Zhou D, Gao L, Song S, Zhu X. YAP promotes the proliferation and migration of colorectal cancer cells through the Glut3/AMPK signaling pathway. Oncol Lett 2021; 21:312. [PMID: 33692844 PMCID: PMC7933749 DOI: 10.3892/ol.2021.12573] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 02/02/2021] [Indexed: 12/16/2022] Open
Abstract
Yes-associated protein (YAP), as a major downstream effector in the Hippo signaling pathway, is considered as an oncogene in cancer. The present study aimed to investigate the potential role of YAP in the development and progression of colorectal cancer (CRC). The mRNA and protein expression levels of YAP in human CRC tissue samples and adjacent normal tissue were analyzed using public databases, as well as clinical samples. The potential roles of YAP and the underlying mechanism regulating the proliferation and migration of CRC cells were examined using genetic manipulation in vitro. The correlation between the expression of the YAP gene and epithelial-to-mesenchymal transition (EMT) markers was investigated in order to determine the mechanism underlying the observed effects of YAP. YAP mRNA expression levels were significantly upregulated in CRC tissue compared with in normal tissue, as determined using datasets obtained from Oncomine. Similarly, in clinical samples, the protein expression levels of YAP were significantly upregulated in CRC tissue samples compared with in normal tissue samples. YAP knockdown inhibited the proliferation and migration of CRC cells in vitro, whereas its overexpression resulted in the opposite effect. The expression levels of the YAP gene were positively correlated with those of EMT markers (such as vimentin and N-cadherin) and EMT-inducing transcription factors (such as Snail1, Slug and zinc finger E-box binding homeobox 1 and 2) in CRC samples from Gene Expression Profiling Interactive Analysis. Furthermore, YAP silencing increased the protein expression of E-cadherin and decreased that of vimentin in CRC cells. By contrast, the overexpression of YAP had the opposite effect. YAP promoted the glucose transporter 3 (Glut3)/AMP-activated protein kinase (AMPK) signaling pathway in CRC cells. In conclusion, YAP promoted the proliferation and migration of CRC cells, as well as the expression of EMT markers, possibly by regulating the Glut3/AMPK signaling pathway.
Collapse
Affiliation(s)
- Linhua Jiang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Jiawen Zhang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Qixuan Xu
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Bin Wang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Yizhou Yao
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Liang Sun
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Xuchao Wang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Diyuan Zhou
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Ling Gao
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Shiduo Song
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Xinguo Zhu
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| |
Collapse
|
31
|
Yang H, Wang X, Wang C, Yin F, Qu L, Shi C, Zhao J, Li S, Ji L, Peng W, Luo H, Cheng M, Kong L. Optimization of WZ4003 as NUAK inhibitors against human colorectal cancer. Eur J Med Chem 2020; 210:113080. [PMID: 33310286 DOI: 10.1016/j.ejmech.2020.113080] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/16/2020] [Accepted: 12/01/2020] [Indexed: 10/22/2022]
Abstract
NUAK, the member of AMPK (AMP-activated protein kinase) family of protein kinases, is phosphorylated and activated by the LKB1 (liver kinase B1) tumor suppressor protein kinase. Recent work has indicated that NUAK1 is a key component of the antioxidant stress response pathway, and the inhibition of NUAK1 will suppress the growth and survival of colorectal tumors. As a promising target for anticancer drugs, few inhibitors of NUAK were developed. With this goal in mind, based on NUAK inhibitor WZ4003, a series of derivatives has been synthesized and evaluated for anticancer activity. Compound 9q, a derivative of WZ4003 by removing a methoxy group, was found to be the most potential one with stronger inhibitory against NUAK1/2 enzyme activity, tumor cell proliferation and inducing apoptosis of tumor cells. By in vivo efficacy evaluations of colorectal SW480 xenografts, 9q suppresses tumor growth more effectively with an excellent safety profile in vivo and is therefore seen as a suitable candidate for further investigation.
Collapse
Affiliation(s)
- Huali Yang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, China; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Xiaobing Wang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, China
| | - Cheng Wang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, China
| | - Fucheng Yin
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, China
| | - Lailiang Qu
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, China
| | - Cunjian Shi
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, China
| | - Jinhua Zhao
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, China
| | - Shang Li
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, China
| | - Limei Ji
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, China
| | - Wan Peng
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, China
| | - Heng Luo
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, China
| | - Maosheng Cheng
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Lingyi Kong
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, China; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| |
Collapse
|
32
|
Chung HH, Lee CT, Hu JM, Chou YC, Lin YW, Shih YL. NKX6.1 Represses Tumorigenesis, Metastasis, and Chemoresistance in Colorectal Cancer. Int J Mol Sci 2020; 21:ijms21145106. [PMID: 32707737 PMCID: PMC7404324 DOI: 10.3390/ijms21145106] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 07/14/2020] [Accepted: 07/16/2020] [Indexed: 12/28/2022] Open
Abstract
Accumulating evidence suggests that NKX6.1 (NK homeobox 1) plays a role in various types of cancer. In our previous studies, we identified NKX6.1 hypermethylation as a promising marker and demonstrated that the NKX6.1 gene functions as a metastasis suppressor through the epigenetic regulation of the epithelial-to-mesenchymal transition (EMT) in cervical cancer. More recently, we have demonstrated that NKX6.1 methylation is related to the chemotherapy response in colorectal cancer (CRC). Nevertheless, the biological function of NKX6.1 in the tumorigenesis of CRC remains unclear. In this study, we showed that NKX6.1 suppresses tumorigenic and metastatic ability both in vitro and in vivo. NKX6.1 represses cell invasion partly through the modulation of EMT. The overexpression of NKX6.1 enhances chemosensitivity in CRC cells. To further explore how NKX6.1 exerts its tumor-suppressive function, we used RNA sequencing technology for comprehensive analysis. The results showed that differentially expressed genes (DEGs) were mainly related to cell migration, response to drug, transcription factor activity, and growth factor activity, suggesting that these DEGs are involved in the function of NKX6.1 suppressing cancer invasion and metastasis. Our results demonstrated that NKX6.1 functions as a tumor suppressor partly by repressing EMT and enhancing chemosensitivity in CRC, making it a potential therapeutic target.
Collapse
Affiliation(s)
- Hsin-Hua Chung
- Graduate Institute of Medical Sciences, National Defense Medical Center, No.161, Sec.6, Minquan East Rd., Neihu District, Taipei 11490, Taiwan; (H.-H.C.); (J.-M.H.); (Y.-W.L.)
| | - Chun-Te Lee
- Division of Urological Surgery, Department of Surgery, Tri-Service General Hospital Songshan Branch, No.131, Jiankang Rd., Songshan District, Taipei 10581, Taiwan;
| | - Je-Ming Hu
- Graduate Institute of Medical Sciences, National Defense Medical Center, No.161, Sec.6, Minquan East Rd., Neihu District, Taipei 11490, Taiwan; (H.-H.C.); (J.-M.H.); (Y.-W.L.)
- Division of Colorectal Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, No.325, Sec.2, Chenggong Rd., Neihu District, Taipei 11490, Taiwan
| | - Yu-Ching Chou
- School of Public Health, National Defense Medical Center, No.161, Sec.6, Minquan East Rd., Neihu District, Taipei 11490, Taiwan;
| | - Ya-Wen Lin
- Graduate Institute of Medical Sciences, National Defense Medical Center, No.161, Sec.6, Minquan East Rd., Neihu District, Taipei 11490, Taiwan; (H.-H.C.); (J.-M.H.); (Y.-W.L.)
- Department and Graduate Institute of Microbiology and Immunology, National Defense Medical Center, No.161, Sec.6, Minquan East Rd., Neihu District, Taipei 11490, Taiwan
- Graduate Institute of Life Sciences, National Defense Medical Center, No.161, Sec.6, Minquan East Rd., Neihu District, Taipei 11490, Taiwan
| | - Yu-Lueng Shih
- Graduate Institute of Medical Sciences, National Defense Medical Center, No.161, Sec.6, Minquan East Rd., Neihu District, Taipei 11490, Taiwan; (H.-H.C.); (J.-M.H.); (Y.-W.L.)
- Division of Gastroenterology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, No. 325, Sec. 2, Chenggong Rd., Neihu District, Taipei 11490, Taiwan
- Correspondence: ; Tel./Fax: +886-2-87917654
| |
Collapse
|
33
|
Zhou JW, Wang H, Sun W, Han NN, Chen L. ASPM is a predictor of overall survival and has therapeutic potential in endometrial cancer. Am J Transl Res 2020; 12:1942-1953. [PMID: 32509189 PMCID: PMC7270042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 04/11/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Endometrial carcinoma (EC) is one of the most common cancers in women, and its pathogenesis is complex. Abnormal spindle microtubule assembly (ASPM) is highly expressed in a variety of cancers and is related to poor clinical prognosis and recurrence. However, the role of ASPM in EC is still unclear. Our study was conducted to investigate the association of ASPM with tumour progression and prognosis in EC. METHODS The expression level of ASPM in EC patients was analysed by using the TCGA database and by using immunohistochemistry (IHC) to analyse EC patient samples. The relationship between ASPM expression and clinicopathological variables was analysed by the chi-square test. Survival curves were analysed by Kaplan-Meier survival analysis and log-rank test. Univariate and multivariate Cox regression analyses were performed to measure the prognosis of EC. The effects of ASPM on the proliferation, invasion and metastasis of EC cells (HEC-1A and Ishikawa) were analysed by MTT and Transwell assays. The effect of ASPM on the Wnt/β-catenin signalling pathway was detected by Western blotting. RESULTS ASPM was highly overexpressed in EC. Overexpression of ASPM was related to significantly worse overall survival (P<0.05) in EC patients. Univariate and multivariate Cox regression analyses suggested that upregulation of ASPM was related to poor prognosis in EC. Knockdown of ASPM inhibited the proliferation, migration and invasion of EC cells. ASPM knockdown suppressed the Wnt/β-catenin signalling pathway, while β-catenin overexpression reversed the effect of shASPM on cell activity. CONCLUSIONS ASPM acts as an independent predictor of clinical prognosis and serves as a potential target gene for EC therapy.
Collapse
Affiliation(s)
- Jing-Wei Zhou
- Department of Gynaecology, Jiangsu Province Hospital Nanjing 210000, P. R. China
| | - Hui Wang
- Department of Gynaecology, Jiangsu Province Hospital Nanjing 210000, P. R. China
| | - Wei Sun
- Department of Gynaecology, Jiangsu Province Hospital Nanjing 210000, P. R. China
| | - Nan-Nan Han
- Department of Gynaecology, Jiangsu Province Hospital Nanjing 210000, P. R. China
| | - Liang Chen
- Department of Gynaecology, Jiangsu Province Hospital Nanjing 210000, P. R. China
| |
Collapse
|
34
|
Shan Z, Cai S, Yu J, Zhang Z, Vallecillo TGM, Serafini MJ, Thomas AM, Pham NYN, Bellampalli SS, Moutal A, Zhou Y, Xu GB, Xu YM, Luo S, Patek M, Streicher JM, Gunatilaka AAL, Khanna R. Reversal of Peripheral Neuropathic Pain by the Small-Molecule Natural Product Physalin F via Block of CaV2.3 (R-Type) and CaV2.2 (N-Type) Voltage-Gated Calcium Channels. ACS Chem Neurosci 2019; 10:2939-2955. [PMID: 30946560 DOI: 10.1021/acschemneuro.9b00166] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
No universally efficacious therapy exists for chronic pain, a disease affecting one-fifth of the global population. An overreliance on the prescription of opioids for chronic pain despite their poor ability to improve function has led to a national opioid crisis. In 2018, the NIH launched a Helping to End Addiction Long-term plan to spur discovery and validation of novel targets and mechanisms to develop alternative nonaddictive treatment options. Phytochemicals with medicinal properties have long been used for various treatments worldwide. The natural product physalin F, isolated from the Physalis acutifolia (family: Solanaceae) herb, demonstrated antinociceptive effects in models of inflammatory pain, consistent with earlier reports of its anti-inflammatory and immunomodulatory activities. However, the target of action of physalin F remained unknown. Here, using whole-cell and slice electrophysiology, competition binding assays, and experimental models of neuropathic pain, we uncovered a molecular target for physalin F's antinociceptive actions. We found that physalin F (i) blocks CaV2.3 (R-type) and CaV2.2 (N-type) voltage-gated calcium channels in dorsal root ganglion (DRG) neurons, (ii) does not affect CaV3 (T-type) voltage-gated calcium channels or voltage-gated sodium or potassium channels, (iii) does not bind G-protein coupled opioid receptors, (iv) inhibits the frequency of spontaneous excitatory postsynaptic currents (EPSCs) in spinal cord slices, and (v) reverses tactile hypersensitivity in models of paclitaxel-induced peripheral neuropathy and spinal nerve ligation. Identifying CaV2.2 as a molecular target of physalin F may spur its use as a tool for mechanistic studies and position it as a structural template for future synthetic compounds.
Collapse
Affiliation(s)
- Zhiming Shan
- Department of Anesthesiology, Shenzhen People’s Hospital & Second Clinical Medical College of Jinan University, Shenzhen 518020, P.R. China
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, P.R. China
| | | | - Jie Yu
- College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou 310058, P.R. China
| | - Zhongjun Zhang
- Department of Anesthesiology, Shenzhen People’s Hospital & Second Clinical Medical College of Jinan University, Shenzhen 518020, P.R. China
| | | | | | | | | | | | | | - Yuan Zhou
- The First Hospital of Jilin University, 71 Xinmin Street, Changchun 130021, P. R. China
- BrightRock Path Consulting, LLC, Tucson 85721, Arizona, United States
| | | | | | | | - Marcel Patek
- BrightRock Path Consulting, LLC, Tucson 85721, Arizona, United States
| | | | | | - Rajesh Khanna
- The Center for Innovation in Brain Sciences, The University of Arizona Health Sciences, Tucson, Arizona 85724, United States
| |
Collapse
|
35
|
Fang G, Ye BL, Hu BR, Ruan XJ, Shi YX. CircRNA_100290 promotes colorectal cancer progression through miR-516b-induced downregulation of FZD4 expression and Wnt/β-catenin signaling. Biochem Biophys Res Commun 2018; 504:184-189. [PMID: 30173892 DOI: 10.1016/j.bbrc.2018.08.152] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 08/25/2018] [Indexed: 02/06/2023]
Abstract
Accumulating evidence indicates that circular RNAs (circRNA) exert crucial functions in the development and advance of cancers. CircRNA_100290 has been reported to promote proliferation in oral cancer. However, whether it participates in colorectal cancer (CRC) remains unclear. Here, our report showed that circRNA_100290 level was significantly increased in CRC tissues and cell lines. Besides, circRNA_100290 expression was positively correlated with tumor metastasis while inversely correlated with prognosis. Silencing circRNA_100290 markedly reduced cell proliferation rate, inhibited migration and invasion abilities, but promoted apoptosis in vitro. Mechanistically, our data revealed circRNA_100290 was a competing endogenous RNA (ceRNA) of FZD4 by sponging miR-516b, leading to activation of Wnt/β-catenin pathway. Rescue assay indicated that FZD4-induced activation of β-catenin pathway is indispensable for the function of circRNA_100290 in CRC. In summary, our study for the first time revealed a novel regulatory loop of circRNA_100290/miR-516b/FZD4/Wnt/β-catenin implicated in CRC progression.
Collapse
Affiliation(s)
- Guan Fang
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Bai-Liang Ye
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Bing-Ren Hu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Xiao-Jiao Ruan
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Yi-Xiong Shi
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| |
Collapse
|