1
|
Liu L, Li Y, Jian C, Guo R, Wang Q. Regulation of apocarotenoids for quality improvement and biofortification of horticultural crops. J Adv Res 2025:S2090-1232(25)00281-4. [PMID: 40320168 DOI: 10.1016/j.jare.2025.04.037] [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: 01/04/2025] [Revised: 04/16/2025] [Accepted: 04/23/2025] [Indexed: 05/10/2025] Open
Abstract
BACKGROUND Agro-food production and consumption impact climate change and human health. Bioactive secondary metabolites in horticulture crops make them an indispensable part of environmentally sustainable and healthy diet. Among them, apocarotenoids from carotenoid degradation are promising in promoting a preference for plant-based foods over other metabolites. AIM OF REVIEW In horticulture crops, carotenoids are vital for photosynthesis and antioxidant defense, but their enzymatic or oxidative metabolites, apocarotenoids, offer greater structural diversity and biological functions. They serve as pigments, scents, signaling molecules, and growth regulators in crop growth and development and provide antioxidant, nutraceutical, and pharmaceutical benefits to human health. The carotenoids as bioactive compounds are well understood. By contrast, much less is explored and reviewed about apocarotenoids. KEY SCIENTIFIC CONCEPTS OF REVIEW Recently identified metabolic pathways and components of apocarotenoids are reviewed. Their significance for quality formation in horticulture crops, including the regulation of pigmentation, aroma, flavor, architecture, nutrition value, and broader ecological interactions is discussed. Additionally, this review specifically highlights two representative apocarotenoids, retinal and abscisic acid (ABA), that exhibit conserved yet distinct regulatory functions across plant and animal kingdoms. Comprehensive dissection of apocarotenoid metabolism and their regulatory mechanisms will enhance apocarotenoid biofortification and subsequent biotechnological exploitation in horticultural commodities. We put forward the perspective that apocarotenoids could enhance horticultural crop quality and then promote sensory- and health-driven dietary choices which will in turn increase consumption and production of horticultural plants and promote both human and ecosystem health.
Collapse
Affiliation(s)
- Lihong Liu
- Key Laboratory of Horticultural Plant Growth, Development and Quality Improvement, Ministry of Agriculture, Department of Horticulture, Zhejiang University, Hangzhou 310058, PR China
| | - Yuening Li
- Key Laboratory of Horticultural Plant Growth, Development and Quality Improvement, Ministry of Agriculture, Department of Horticulture, Zhejiang University, Hangzhou 310058, PR China
| | - Chunxia Jian
- Key Laboratory of Horticultural Plant Growth, Development and Quality Improvement, Ministry of Agriculture, Department of Horticulture, Zhejiang University, Hangzhou 310058, PR China
| | - Rongfang Guo
- Joint FAFU-Dalhousie Lab, College of Horticulture, Fujian Agriculture and Forestry University, Fuzhou 350002, PR China; Institute of Horticultural Biotechnology, College of Horticulture, Fujian Agriculture and Forestry University, Fuzhou 350002, PR China; Key Laboratory of Ministry of Education for Genetics, Breeding and Multiple Utilization of Crops, Fujian Agriculture and Forestry University, Fuzhou 350002, PR China.
| | - Qiaomei Wang
- Key Laboratory of Horticultural Plant Growth, Development and Quality Improvement, Ministry of Agriculture, Department of Horticulture, Zhejiang University, Hangzhou 310058, PR China.
| |
Collapse
|
2
|
Gharib A, Marquez C, Meseguer-Beltran M, Sanchez-Sarasua S, Sanchez-Perez AM. Abscisic acid, an evolutionary conserved hormone: Biosynthesis, therapeutic and diagnostic applications in mammals. Biochem Pharmacol 2024; 229:116521. [PMID: 39251140 DOI: 10.1016/j.bcp.2024.116521] [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: 07/09/2024] [Revised: 08/26/2024] [Accepted: 09/05/2024] [Indexed: 09/11/2024]
Abstract
Abscisic acid (ABA), a phytohormone traditionally recognized for its role in plant stress responses, has recently emerged as a significant player in mammalian defense mechanisms. Like plants, various mammalian cell types synthesize ABA in response to specific health challenges, although the precise pathways remain not fully elucidated. ABA is associated with the regulation of inflammation and insulin signaling, prompting extensive research into its potential as a therapeutic agent for various diseases. ABA exerts its effects through its receptors, particularly PPAR-γ and LANCL-2, which serve as signaling hubs regulating numerous pathways. Through these interactions, ABA profoundly impacts mammalian health, and new ABA targets continue to be identified. Numerous studies in animal models demonstrate ABA's benefit in managing conditions such as neurological and psychiatric disorders, cancer, and malaria infections, all of which involve significant inflammatory dysregulation. In this manuscript we review the studies covering ABA synthesis and release in cell cultures, the signaling pathways regulated by ABA, and how these impact health in preclinical models. Furthermore, we highlight recent research suggesting that measuring ABA levels in human body fluids could serve as a useful biomarker for pathological conditions, providing insights into disease progression and treatment efficacy. This comprehensive review outlines the current understanding of ABA in mammalian pathophysiology, identifying gaps in knowledge, particularly concerning ABA biosynthesis and metabolism in mammals. In addition, this study emphasizes the need for clinical trials to validate the effectiveness of ABA-based therapies and its reliability as a biomarker for various diseases.
Collapse
Affiliation(s)
- Amir Gharib
- Neurobiotecnologia Group, Institute of Advanced Materiales (INAM), Universitat Jaume I, Avda. de Vicent Sos Baynat, S/n, 12071 Castelló de La Plana, Spain; Department of Laboratory Sciences, Borujerd Branch, Islamic Azad University, Borujerd, Iran
| | - Carlee Marquez
- Neurobiotecnologia Group, Institute of Advanced Materiales (INAM), Universitat Jaume I, Avda. de Vicent Sos Baynat, S/n, 12071 Castelló de La Plana, Spain
| | - Maria Meseguer-Beltran
- Neurobiotecnologia Group, Institute of Advanced Materiales (INAM), Universitat Jaume I, Avda. de Vicent Sos Baynat, S/n, 12071 Castelló de La Plana, Spain
| | - Sandra Sanchez-Sarasua
- Neurobiotecnologia Group, Institute of Advanced Materiales (INAM), Universitat Jaume I, Avda. de Vicent Sos Baynat, S/n, 12071 Castelló de La Plana, Spain; CNRS UMR 5293, Institut Des Maladies Neurodégénératives, Centre Paul Broca-Nouvelle Aquitaine, University of Bordeaux, Bordeaux, France.
| | - Ana M Sanchez-Perez
- Neurobiotecnologia Group, Institute of Advanced Materiales (INAM), Universitat Jaume I, Avda. de Vicent Sos Baynat, S/n, 12071 Castelló de La Plana, Spain.
| |
Collapse
|
3
|
Iranmanesh Z, Dehestani M, Esmaeili-Mahani S. Discovering novel targets of abscisic acid using computational approaches. Comput Biol Chem 2024; 112:108157. [PMID: 39047594 DOI: 10.1016/j.compbiolchem.2024.108157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 07/16/2024] [Accepted: 07/17/2024] [Indexed: 07/27/2024]
Abstract
Abscisic acid (ABA) is a crucial plant hormone that is naturally produced in various mammalian tissues and holds significant potential as a therapeutic molecule in humans. ABA is selected for this study due to its known roles in essential human metabolic processes, such as glucose homeostasis, immune responses, cardiovascular system, and inflammation regulation. Despite its known importance, the molecular mechanism underlying ABA's action remain largely unexplored. This study employed computational techniques to identify potential human ABA receptors. We screened 64 candidate molecules using online servers and performed molecular docking to assess binding affinity and interaction types with ABA. The stability and dynamics of the best complexes were investigated using molecular dynamics simulation over a 100 ns time period. Root mean square fluctuations (RMSF), root mean square deviation (RMSD), solvent-accessible surface area (SASA), radius of gyration (Rg), free energy landscape (FEL), and principal component analysis (PCA) were analyzed. Next, the molecular mechanics Poisson-Boltzmann surface area (MM-PBSA) method was employed to calculate the binding energies of the complexes based on the simulated data. Our study successfully pinpointed four key receptors responsible for ABA signaling (androgen receptor, glucocorticoid receptor, mineralocorticoid receptor, and retinoic acid receptor beta) that have a strong affinity for binding with ABA and remained structurally stable throughout the simulations. The simulations with Hydralazine as an unrelated ligand were conducted to validate the specificity of the identified receptors for ABA. The findings of this study can contribute to further experimental validation and a better understanding of how ABA functions in humans.
Collapse
Affiliation(s)
- Zahra Iranmanesh
- Department of Chemistry, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Maryam Dehestani
- Department of Chemistry, Shahid Bahonar University of Kerman, Kerman, Iran.
| | | |
Collapse
|
4
|
Shen Z, Yu N, Zhang Y, Jia M, Sun Y, Li Y, Zhao L. The potential roles of HIF-1α in epithelial-mesenchymal transition and ferroptosis in tumor cells. Cell Signal 2024; 122:111345. [PMID: 39134249 DOI: 10.1016/j.cellsig.2024.111345] [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: 06/26/2024] [Revised: 08/03/2024] [Accepted: 08/09/2024] [Indexed: 08/15/2024]
Abstract
In tumors, the rapid proliferation of cells and the imperfect blood supply system lead to hypoxia, which can regulate the adaptation of tumor cells to the hypoxic environment through hypoxia-inducible factor-1α (HIF-1α) and promote tumor development in multiple ways. Recent studies have found that epithelial-mesenchymal transition (EMT) and ferroptosis play important roles in the progression of tumor cells. The activation of HIF-1α is considered a key factor in inducing EMT in tumor cells. When HIF-1α is activated, it can regulate EMT-related genes, causing tumor cells to gradually lose their epithelial characteristics and acquire more invasive mesenchymal traits. The occurrence of EMT allows tumor cells to better adapt to changes in the surrounding tissue, enhancing their migratory and invasive capabilities, thus promoting tumor progression. At the same time, HIF-1α also plays a crucial regulatory role in ferroptosis in tumor cells. In a hypoxic environment, HIF-1α may affect processes such as iron metabolism and oxidative stress responses, inducing ferroptosis in tumor cells. This article briefly reviews the dual role of HIF-1α in EMT and ferroptosis in tumor cells, helping to gain a deeper understanding of the regulatory pathways of HIF-1α in the development of tumor cells, providing a new perspective for understanding the pathogenesis of tumors. The regulation of HIF-1α may become an important strategy for future tumor therapy.
Collapse
Affiliation(s)
- Zhongjun Shen
- Department of Blood Transfusion, Second Hospital of Jilin University, Changchun, 130041 Jilin, China
| | - Na Yu
- Department of Blood Transfusion, Second Hospital of Jilin University, Changchun, 130041 Jilin, China
| | - Yanfeng Zhang
- Department of Blood Transfusion, Second Hospital of Jilin University, Changchun, 130041 Jilin, China
| | - Mingbo Jia
- Department of Blood Transfusion, Second Hospital of Jilin University, Changchun, 130041 Jilin, China
| | - Ying Sun
- Department of Blood Transfusion, Second Hospital of Jilin University, Changchun, 130041 Jilin, China
| | - Yao Li
- Department of Blood Transfusion, Second Hospital of Jilin University, Changchun, 130041 Jilin, China
| | - Liyan Zhao
- Department of Blood Transfusion, Second Hospital of Jilin University, Changchun, 130041 Jilin, China.
| |
Collapse
|
5
|
Horváth L, Biri-Kovács B, Baranyai Z, Stipsicz B, Méhes E, Jezsó B, Krátký M, Vinšová J, Bősze S. New Salicylanilide Derivatives and Their Peptide Conjugates as Anticancer Compounds: Synthesis, Characterization, and In Vitro Effect on Glioblastoma. ACS OMEGA 2024; 9:16927-16948. [PMID: 38645331 PMCID: PMC11024950 DOI: 10.1021/acsomega.3c05727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 12/28/2023] [Accepted: 01/03/2024] [Indexed: 04/23/2024]
Abstract
Pharmacologically active salicylanilides (2-hydroxy-N-phenylbenzamides) have been a promising area of interest in medicinal chemistry-related research for quite some time. This group of compounds has shown a wide spectrum of biological activities, including but not limited to anticancer effects. In this study, substituted salicylanilides were chosen to evaluate the in vitro activity on U87 human glioblastoma (GBM) cells. The parent salicylanilide, salicylanilide 5-chloropyrazinoates, a 4-aminosalicylic acid derivative, and the new salicylanilide 4-formylbenzoates were chemically and in vitro characterized. To enhance the internalization of the compounds, they were conjugated to delivery peptides with the formation of oxime bonds. Oligotuftsins ([TKPKG]n, n = 1-4), the ligands of neuropilin receptors, were used as GBM-targeting carrier peptides. The in vitro cellular uptake, intracellular localization, and penetration ability on tissue-mimicking models of the fluorescent peptide derivatives were determined. The compounds and their peptide conjugates significantly decreased the viability of U87 glioma cells. Salicylanilide compound-induced GBM cell death was associated with activation of autophagy, as characterized by immunodetection of autophagy-related processing of light chain 3 protein.
Collapse
Affiliation(s)
- Lilla Horváth
- ELKH-ELTE
Research Group of Peptide Chemistry, Eötvös Loránd
Research Network, Eötvös Loránd
University, Budapest 1117, Hungary
| | - Beáta Biri-Kovács
- ELKH-ELTE
Research Group of Peptide Chemistry, Eötvös Loránd
Research Network, Eötvös Loránd
University, Budapest 1117, Hungary
| | - Zsuzsa Baranyai
- ELKH-ELTE
Research Group of Peptide Chemistry, Eötvös Loránd
Research Network, Eötvös Loránd
University, Budapest 1117, Hungary
| | - Bence Stipsicz
- ELKH-ELTE
Research Group of Peptide Chemistry, Eötvös Loránd
Research Network, Eötvös Loránd
University, Budapest 1117, Hungary
- Institute
of Biology, Doctoral School of Biology, Eötvös Loránd University, Budapest 1117, Hungary
| | - Előd Méhes
- Institute
of Physics, Department of Biological Physics, Eötvös Loránd University, Budapest 1117, Hungary
| | - Bálint Jezsó
- Research
Centre for Natural Sciences, Institute of
Enzymology, Budapest 1053, Hungary
- ELTE-MTA
“Momentum” Motor Enzymology Research Group, Department
of Biochemistry, Eötvös Loránd
University, Budapest 1117, Hungary
| | - Martin Krátký
- Department
of Organic and Bioorganic Chemistry, Faculty of Pharmacy in Hradec
Králové, Charles University, 500 03 Hradec Králové, Czech Republic
| | - Jarmila Vinšová
- Department
of Organic and Bioorganic Chemistry, Faculty of Pharmacy in Hradec
Králové, Charles University, 500 03 Hradec Králové, Czech Republic
| | - Szilvia Bősze
- ELKH-ELTE
Research Group of Peptide Chemistry, Eötvös Loránd
Research Network, Eötvös Loránd
University, Budapest 1117, Hungary
| |
Collapse
|
6
|
Liu X, Ye Z, Rao D, Chen Q, Zhang Z. DUSP4 maintains the survival and LSD1 protein stability in esophageal squamous cell carcinoma cells by inhibiting JNK signaling-dependent autophagy. In Vitro Cell Dev Biol Anim 2024; 60:115-122. [PMID: 38286920 DOI: 10.1007/s11626-023-00845-4] [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: 05/05/2023] [Accepted: 12/13/2023] [Indexed: 01/31/2024]
Abstract
DUSP4 is a biomarker of esophageal squamous cell carcinoma (ESCC), which is responsible for the prognosis in ESCC. However, the underlying mechanism of DUSP4-regulated ESCC carcinogenesis is unknown. As a negative regulator of JNK, DUSP4 can inhibit autophagy, which contributes to tumorigenesis. This study aimed to explore the role of autophagy in DUSP4-regulated ESCC carcinogenesis. Our results showed that DUSP4 overexpression inhibited autophagy and promoted LSD1 protein expression in ESCC cells, while DUSP4 silencing showed the opposite effects. However, DUSP4 overexpression and silencing did not affect LSD1 mRNA expression. But the regulatory ability of DUSP4 overexpression on autophagy, death level, and LSD1 protein was reversed by rapamycin. In addition, DUSP4 overexpression inhibited JNK and Bcl2 phosphorylation and the dissociation of Bcl2-Beclin1 complex, while DUSP4 silencing promoted JNK and Bcl2 phosphorylation. Moreover, the regulatory ability of DUSP4 overexpression on autophagy, death, and LSD1 protein was reversed by JNK activator anisomycin. The xenograft assays also showed that DUSP4 overexpression-promoted ESCC tumor growth in vivo and LC3II and LSD1 protein expression in tumor tissues were reversed by rapamycin or anisomycin. Overall, DUSP4 inhibits Bcl2-Beclin1-autophagy signal transduction through the negative regulation of JNK, thus suppressing autophagic death and the autophagic degradation of LSD1 in ESCC, by which DUSP4 promotes ESCC carcinogenesis.
Collapse
Affiliation(s)
- Xinxin Liu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, Jiangxi, China
- Gannan Branch of National Clinical Research Center for Geriatrics Ganzhou 341000, Jiangxi, China
| | - Zhou Ye
- Department of Digestive, The 900Th Hospital of Joint Logistic Support Force, PLA, Fuzhou, 350001, Fujian, China
| | - Dingyu Rao
- Department of Thoracic Surgery, First Affiliated Hospital of Gannan Medical University, No. 3, Outangli, Xingannan Road, Zhanggong District, Ganzhou, 341000, Jiangxi, China
| | - Qianshun Chen
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, Fujian, China.
| | - Zuxiong Zhang
- Department of Thoracic Surgery, First Affiliated Hospital of Gannan Medical University, No. 3, Outangli, Xingannan Road, Zhanggong District, Ganzhou, 341000, Jiangxi, China.
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, Fujian, China.
| |
Collapse
|
7
|
Subodh, Ravina, Priyanka, Narang J, Mohan H. Biosensors for phytohormone Abscisic acid and its role in humans: A review. SENSORS INTERNATIONAL 2023. [DOI: 10.1016/j.sintl.2023.100234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/24/2023] Open
|
8
|
Zang L, Song Y, Tian Y, Hu N. TAT-Beclin 1 represses the carcinogenesis of DUSP4-positive PTC by enhancing autophagy. Mol Biol Rep 2023; 50:1425-1436. [PMID: 36474060 DOI: 10.1007/s11033-022-08109-2] [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: 09/20/2021] [Accepted: 11/09/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND DUSP4 is a pro-tumorigenic molecule of papillary thyroid carcinoma (PTC). DUSP4 also exists as an autophagic regulator. Moreover, DUSP4, as a negative regulator of MAPK, can prevent Beclin 1 from participating in autophagic response. This study aimed to explore whether TAT-Beclin 1, a recombinant protein of Beclin 1, could inhibit the tumorigenesis of DUSP4-positive PTC by regulating autophagy. METHODS First, we divided PTC tissues into three groups according to DUSP4 expression levels by immunohistochemical analyses, and evaluated the relationship between autophagic molecules (Beclin 1 and LC3II) and DUSP4 using Western blotting assays. After overexpression of DUSP4 by lentiviral transduction, the in vitro and in vivo roles of TAT-Beclin 1 on DUSP4-overexpressed PTC cells were assessed (including autophagic activity, cell survival and function, and tumor growth). The roles of TAT-Beclin 1 in the survival of DUSP4-silenced PTC cells were also evaluated. RESULTS Our results showed that the expression levels of autophagic proteins decreased with the increase of DUSP4 expression in PTC tissues. In PTC cells, DUSP4 overexpression-inhibited autophagic activity (including Beclin 1 expression, LC3 conversion rate and LC3-puncta formation) and -promoted cell proliferation and migration were reversed by TAT-Beclin 1 administration. In vivo assays also showed that DUSP4-overexpressed PTC cells had stronger tumorigenic ability and weaker autophagic activity, which was blocked by TAT-Beclin 1 administration. CONCLUSION TAT-Beclin 1, as an autophagic promoter, could repress the carcinogenesis of DUSP4-positive PTC, which implies that the use of TAT-Beclin 1 for the PTC patients' treatment might be determined according to the DUSP4 level in their tumors.
Collapse
Affiliation(s)
- Leilei Zang
- Department 5 of General Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, 050005, Hebei, China
| | - Yanmei Song
- Department of Infection Management/Public Health, Hebei People's Hospital, Shijiazhuang, 050057, Hebei, China
| | - Yanhua Tian
- Department 2 of Oncology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050005, Hebei, China
| | - Ning Hu
- Department 4 of General Surgery, The Second Hospital of Hebei Medical University, No.215 Heping West Road, Xinhua District, Shijiazhuang, 050005, Hebei, China.
| |
Collapse
|
9
|
Xu W, Nie C, Chen X. DUSP4 inhibits autophagic cell death and apoptosis in colorectal cancer by regulating BCL2-Beclin1/Bax signaling. Mol Biol Rep 2023; 50:3229-3239. [PMID: 36705792 DOI: 10.1007/s11033-023-08270-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 01/10/2023] [Indexed: 01/28/2023]
Abstract
BACKGROUND The DUSP4 gene plays an important role in the carcinogenesis of colorectal cancer (CRC). However, the underlying mechanism of DUSP4-regulated colorectal carcinogenesis is unknown. DUSP4 is a negative regulator of the MAP kinase (MAPK) JNK, and JNK-mediated BCL2 phosphorylation is associated with apoptosis and autophagic cell death. Our study aimed to explore the significance of BCL2 phosphorylation-dependent autophagy and apoptosis in DUSP4-promoted colorectal carcinogenesis. METHODS We first investigated the roles of DUSP4 in the survival of HCT116 and SW480 CRC cell lines using gene-silencing and -overexpression techniques. Next, we explored the effects of DUSP4 on the BCL2 phosphorylation, autophagy and apoptosis of HCT116 and SW480 cells. Ultimately, with the help of pharmacological inhibitors of Beclin1 and BCL2 (spautin-1 and ABT-737), the relationship between BCL2-Beclin1/Bax signaling and DUSP4-regulated autophagy, apoptosis, survival and migration in HCT116 cells was clarified. RESULTS Our results first confirmed the contribution of DUSP4 to the survival of HCT116 and SW480 cells. In addition, DUSP4 silencing resulted in BCL2 phosphorylation and the enhancement in autophagy and apoptosis in HCT116 and SW480 cells, while DUSP4 overexpression showed the opposite effect. Moreover, DUSP4 silencing inhibited the protein interaction between BCL2 and Beclin1 or Bax in HCT116 cells. Moreover, the survival and migration of HCT116 cells inhibited by DUSP4 silencing were blocked by autophagy inhibition with spautin-1. Notably, the survival and migration of HCT116 cells promoted by DUSP4 overexpression were reversed by ABT-737. CONCLUSIONS It was indicated that DUSP4 can maintain the survival and function of CRC cells by inhibiting BCL2 phosphorylation-dependent autophagic cell death and apoptosis.
Collapse
Affiliation(s)
- Weifeng Xu
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University, No.127 Dong Ming Road, Zhengzhou, 450008, Henan, People's Republic of China
| | - Caiyun Nie
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University, No.127 Dong Ming Road, Zhengzhou, 450008, Henan, People's Republic of China
| | - Xiaobing Chen
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University, No.127 Dong Ming Road, Zhengzhou, 450008, Henan, People's Republic of China.
| |
Collapse
|
10
|
Del Mondo A, Vinaccia A, Pistelli L, Brunet C, Sansone C. On the human health benefits of microalgal phytohormones: An explorative in silico analysis. Comput Struct Biotechnol J 2023; 21:1092-1101. [PMID: 36789263 PMCID: PMC9900276 DOI: 10.1016/j.csbj.2023.01.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 01/23/2023] [Accepted: 01/23/2023] [Indexed: 01/26/2023] Open
Abstract
Phytohormones represent a group of secondary metabolites with different chemical structures, in which belong auxins, cytokinins, gibberellins, or brassinosteroids. In higher plants, they cover active roles in growth or defense function, while their potential benefits for human health protection were noted for some phytohormones and little explored for many others. In this study, we developed a target fishing strategy on fifty-three selected naturally occurring phytohormones covering different families towards proteins involved in key cellular functions related to human metabolism and health protection/disease. This in silico analysis strategy aims to screen the potential human health-driven bioactivity of more than fifty phytohormones through the analysis of their interactions with specific targets. From this analysis, twenty-eight human targets were recovered. Some targets e.g., the proteins mitochondrial glutamate dehydrogenase (GLUD1) or nerve growth factor (NGF) bound many phytohormones, highlighting their involvement in amino acid metabolism and/or in the maintenance or survival of neurons. Conversely, some phytohormones specifically interacted with some proteins, e.g., SPRY domain-containing SOCS box protein 2 (SPSB2) or Inosine-5'-monophosphate dehydrogenase 1 (IMPDH1), both involved in human immune response. They were then investigated with a molecular docking analysis approach. Our bioprospecting study indicated that many phytohormones may endow human health benefits, with potential functional role in multiple cellular processes including immune response and cell cycle progression.
Collapse
|
11
|
Jabbar ZR, Sahib HB. The Effects of Abscisic Acid on Angiogenesis in Both ex vivo and in vivo Assays. Asian Pac J Cancer Prev 2022; 23:4193-4203. [PMID: 36580002 PMCID: PMC9971465 DOI: 10.31557/apjcp.2022.23.12.4193] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Angiogenesis is a complex biological process wherein novel capillary blood vessels mature from pre-existing vasculature for delivering tissues with oxygen and nutrients. Natural molecules that have anti-angiogenic activity and toxicity can raise the focus on using plant sources as essential therapeutic agent. OBJECTIVE The current research was intended to estimate the probable anti-angiogenic activity of abscisic acid alone and in combination with prednisolone, a well-known angiostatic glucocorticoid. METHODS two months old albino rats were used in this study. ABA and prednisolone stock solutions were prepared and added after embedding aortic rings in growth media. The ex vivo rat aorta ring assay (RAR) was applied to explore the anti-angiogenic effect of ABA. The in vivo chorioallantoic membrane assay (CAM) was applied to quantify the blood vessels inhibition zone by ABA effect. That zone was calculated as the mean inhibition region on eggs in mm ± SD. RESULTS Abscisic acid screened byex vivo and in vivo assays, revealed a significant dose-dependent blood vessels inhibition in comparison to the negative control with IC50= 7.5µg/ml and a synergism effect when combined with prednisolone. CONCLUSION The synergism activity of ABA with prednisolone can significantly inhibit blood vessels growth in RAR and CAM assays. These results shed the light on the potential clinic values of ABA, and prednisolone combination in angiogenesis-dependent tumors.
Collapse
Affiliation(s)
| | - Hayder B Sahib
- Department of Pharmacology, College of Medicine, Al-Nahrain University, Baghdad, Iraq.
| |
Collapse
|
12
|
Lin S, Qin HZ, Li ZY, Zhu H, Long L, Xu LB. Gallic acid suppresses the progression of triple-negative breast cancer HCC1806 cells via modulating PI3K/AKT/EGFR and MAPK signaling pathways. Front Pharmacol 2022; 13:1049117. [PMID: 36523491 PMCID: PMC9744937 DOI: 10.3389/fphar.2022.1049117] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 11/15/2022] [Indexed: 11/04/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is a severe threat to women's health because of its aggressive nature, early age of onset, and high recurrence rate. Therefore, in this study, we aimed to evaluate the anti-tumor effects of Gallic acid (GA) on the TNBC HCC1806 cells in vitro. The cell proliferation was detected by MTT and plate clone formation assays, cell apoptosis, cell cycle, and mitochondrial membrane potential (MMP) were analyzed by flow cytometry and Hoechst 33258 staining assays, and the intracellular reactive oxygen species (ROS) accumulation were also investigated. Real-Time PCR and western blot were examined to explore the mechanism of action. The results indicated that GA suppressed HCC1806 cells proliferation and promoted HCC1806 cells apoptosis. Meanwhile, GA treatment changed the morphology of the HCC1806 cells. In addition, GA blocked the HCC1806 cells cycle in the S phase, and it induced cells apoptosis accompanied by ROS accumulation and MMP depolarization. Real-Time PCR results suggested that GA increased Bax, Caspase-3, Caspase-9, P53, JINK and P38 mRNA expression, and decreased Bcl-2, PI3K, AKT and EGFR mRNA expression. Western blotting results suggested that GA increased Bax, cleaved-Caspase-3, cleaved-Caspase-9, P53, P-ERK1/2, P-JNK, P-P38 proteins expression, and decreased Bcl-2, P-PI3K, P-AKT, P-EGFR proteins expression. Furthermore, molecular docking suggested that GA has the high affinity for PI3K, AKT, EGFR, ERK1/2, JNK, and P38. In conclusion, GA could suppress HCC1806 cells proliferation and promote HCC1806 cells apoptosis through the mitochondrial apoptosis pathway and induces ROS generation which further inhibits PI3K/AKT/EGFR and activates MAPK signaling pathways. Our study will provide some new references for using GA in the treatment of TNBC.
Collapse
Affiliation(s)
- Si Lin
- Guangxi Key Laboratory of Zhuang and Yao Ethnic Medicine, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Scientific Research Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, China
| | - Hui-Zhen Qin
- Guangxi Key Laboratory of Zhuang and Yao Ethnic Medicine, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Scientific Research Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, China
| | - Ze-Yu Li
- Guangxi Scientific Research Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, China
| | - Hua Zhu
- Guangxi Key Laboratory of Zhuang and Yao Ethnic Medicine, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Scientific Research Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, China
| | - Li Long
- Guangxi International Zhuang Medicine Hospital, Guangxi University of Chinese Medicine, Nanning, China
| | - Li-Ba Xu
- Guangxi Key Laboratory of Zhuang and Yao Ethnic Medicine, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Scientific Research Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, China
| |
Collapse
|
13
|
Overexpression of PpSnRK1α in Tomato Increased Autophagy Activity under Low Nutrient Stress. Int J Mol Sci 2022; 23:ijms23105464. [PMID: 35628273 PMCID: PMC9141306 DOI: 10.3390/ijms23105464] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/11/2022] [Accepted: 05/11/2022] [Indexed: 11/26/2022] Open
Abstract
Plants suffer from a variety of environmental stresses during their growth and development. The evolutionarily conserved sucrose nonfermenting kinase 1-related protein kinase 1 (SnRK1) plays a central role in the regulation of energy homeostasis in response to stresses. In plant cells, autophagy is a degradation process occurring during development or under stress, such as nutrient starvation. In recent years, SnRK1 signaling has been reported to be an upstream activator of autophagy. However, these studies all focused on the regulatory effect of SnRK1 on TOR signaling and the autophagy-related gene 1 (ATG1) complex. In this study, overexpression of the gene encoding the Prunus persica SnRK1 α subunit (PpSnRK1α) in tomato improved the photosynthetic rates and enhanced the resistance to low nutrient stress (LNS). Overexpression of PpSnRK1α increased autophagy activity and upregulated the expression of seven autophagy-related genes (ATGs). The transcriptional levels of SlSnRK2 family genes were altered significantly by PpSnRK1α, signifying that PpSnRK1α may be involved in the ABA signaling pathway. Further analysis showed that PpSnRK1α not only activated autophagy by inhibiting target of rapamycin (TOR) signaling but also enhanced ABA-induced autophagy. This indicates that PpSnRK1α regulates the photosynthetic rate and induces autophagy, and then responds to low nutrient stress.
Collapse
|
14
|
Targeting Protein Kinase C for Cancer Therapy. Cancers (Basel) 2022; 14:cancers14051104. [PMID: 35267413 PMCID: PMC8909172 DOI: 10.3390/cancers14051104] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/15/2022] [Accepted: 02/17/2022] [Indexed: 12/18/2022] Open
Abstract
Simple Summary The protein kinase C (PKC) family belongs to serine-threonine kinases and consists of several subtypes. Increasing evidence suggests that PKCs are critical players in carcinogenesis. Interestingly, PKCs exert both promotive and suppressive effects on tumor cell growth and metastasis, which have attracted immense attention. Herein, we systematically review the current advances in the structure, regulation and biological functions of PKCs, especially the relationship of PKCs with anti-cancer therapy-induced cell death, including the current knowledge of PKCs function in tumor metabolism and microenvironment. Moreover, we discuss the potential role of PKCs as a target for therapeutic intervention in cancer from basic research and clinical trials. Abstract Protein kinase C (PKC) isoforms, a group of serine-threonine kinases, are important regulators in carcinogenesis. Numerous studies have demonstrated that PKC isoforms exert both positive and negative effects on cancer cell demise. In this review, we systematically summarize the current findings on the architecture, activity regulation and biological functions of PKCs, especially their relationship with anti-cancer therapy-induced cell death. Additionally, we elaborate on current knowledge of the effects of PKCs on tumor metabolism and microenvironment, which have gained increasing attention in oncology-related areas. Furthermore, we underscore the basic experimental and clinical implications of PKCs as a target for cancer therapy to evaluate their therapeutic benefits and potential applications.
Collapse
|
15
|
He H, Du Z, Lin J, Wu W, Yu Y. DUSP4 inhibits autophagic cell death in PTC by inhibiting JNK-BCL2-Beclin1 signaling. Biochem Cell Biol 2021; 99:554-561. [PMID: 33621155 DOI: 10.1139/bcb-2020-0636] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Dual specificity phosphatase 4 (DUSP4) is a prognostic marker and potential target of papillary thyroid carcinoma (PTC); however, the molecular mechanism underlying DUSP4-regulated PTC carcinogenesis is unknown. DUSP4 is a negative regulator of the autophagy promoter, JNK. This study explored the relationship between DUSP4 and JNK-mediated autophagic cell death in PTC, and the roles of DUSP4 in PTC using gain-of-function and loss-of-function assays. In addition, we further identified the significance of the JNK-BCL2-Beclin1-autophagy signaling pathway on DUSP4-regulated PTC carcinogenesis by combining knockdown of DUSP4 with a JNK-specific inhibitor (SP600125). We found that knockdown of DUSP4 promoted the phosphorylation of JNK and BCL2 in PTC cells, and enhanced the release of Beclin1 from the BCL2-Beclin1 complex. Knockdown of DUSP4 promoted autophagy and the death of PTC cells. The death and autophagy enhanced by knockdown of DUSP4 was reversed by the JNK inhibitor. We further extended the in-vitro experiments by subcutaneously injecting nude mice with K1 cells transfected with DUSP4-silencing vector. In-vivo assays showed that knockdown of DUSP4 not only inhibited tumor growth, but also promoted the phosphorylation of JNK and BCL2 and the expression of LC3II. In conclusion, DUSP4 inhibits BCL2-Beclin1-autophagy signaling by negatively regulating JNK activity, thus inhibiting PTC oncogenesis. The data from this study contribute to the prevention and cure of PTC.
Collapse
Affiliation(s)
- Huixiang He
- Department of General Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, China
| | - Zhenshuang Du
- Department of General Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, China
| | - Jianqing Lin
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, China
| | - Wenyi Wu
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, China
| | - Yihuang Yu
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, China
| |
Collapse
|
16
|
Wang P, Li X, Xie Y. B4GalT1 Regulates Apoptosis and Autophagy of Glioblastoma In Vitro and In Vivo. Technol Cancer Res Treat 2020; 19:1533033820980104. [PMID: 33287670 PMCID: PMC7727053 DOI: 10.1177/1533033820980104] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Our study was designed to investigate the role of B4GalT1 in glioblastoma, in vitro and in vivo, to detect whether B4GalT1 knockdown could regulate the development of glioblastoma, and further observe the relationship between B4GalT1 knockdown and the apoptosis and autophagy of glioblastoma. To begin, we looked at TCGA and GEPIA systems to predict the potential function of B4GalT1. Western blot and RT-PCR were used to analyze the expression, or mRNA level, of B4GalT1 at different tissue or cell lines. Next, the occurrence and development of glioblastoma, in vitro and in vivo, was observed by using B4GalT1 knocked down by lentivirus. Finally, the apoptosis and autophagy of glioblastoma was observed in vitro and in vivo. Results show that B4GalT1 was a highly variable gene, and GEPIA and TCGA systems show B4GalT1 expression in GBM tumor tissue was higher than in normal tissue. Pair-wise gene correlation analysis revealed a probable relationship between B4GalT1 and autophagy related proteins. The B4GalT1 expression and mRNA level were increased in tumor cells, or U87 cells. B4GalT1 knocked down by lentivirus could inhibit glioblastoma development, in vitro and in vivo, by reducing tumor weight and volume, increasing survival, and weakening tumor cells proliferation, migration, invasion. B4GalT1 knockdown could increase apoptosis and autophagy of glioblastoma in vitro and in vivo. Our study demonstrates that B4GalT1 may be able to regulate apoptosis and autophagy of glioblastoma. Bax, Bcl-2, cleaved caspase-3, Beclin-1, and LC3 s may be the downstream target factors of B4GalT1 in apoptosis and autophagy, which may provide a new strategy to reduce glioblastoma development by regulating apoptosis and autophagy.
Collapse
Affiliation(s)
- Pu Wang
- Department of Neurology, Xiangyang First People's Hospital of Hubei University, Xiangyang, Hubei, China
| | - Xiaolong Li
- Department of Neurology, Xiangyang First People's Hospital of Hubei University, Xiangyang, Hubei, China
| | - Yuan Xie
- Department of Neurology, Xiangyang First People's Hospital of Hubei University, Xiangyang, Hubei, China
| |
Collapse
|
17
|
Arctigenin Inhibits Glioblastoma Proliferation through the AKT/mTOR Pathway and Induces Autophagy. BIOMED RESEARCH INTERNATIONAL 2020; 2020:3542613. [PMID: 33015162 PMCID: PMC7512051 DOI: 10.1155/2020/3542613] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/11/2020] [Accepted: 08/31/2020] [Indexed: 12/21/2022]
Abstract
Purpose Arctigenin (ARG) is a natural lignan compound extracted from Arctium lappa and has displayed anticancer function and therapeutic effect in a variety of cancers. Arctigenin is mainly from Arctium lappa extract. It has been shown to induce autophagy in various cancers. However, as for whether arctigenin induces autophagy in gliomas or not, the specific mechanism is still worth exploring. Methods Using CCK8, the monoclonal experiment was made to detect the proliferation ability. The scratch experiment and the transwell experiment were applied to the migration and invasion ability. PI/RNase and FITC-conjugated anti-annexin V were used to detect the cell cycle and apoptosis. Western blotting was used to determine the specified protein level, and constructed LC3B-GFP plasmid was used for analysis of autophagy. Results Our research showed that ARG inhibited the growth and proliferation and invasion and migration of glioma cells in a dose-dependent manner (U87MG and T98G) and arrested the cell cycle and induced apoptosis. Interestingly, ARG induced autophagy in a dose-dependent manner. We applied Western blotting to measure the increase in the key autophagy protein LC3B, as well as some other autophagy-related proteins (increase in Beclin-1 and decrease in P62). In order to further explore the mechanism that ARG passed initiating autophagy to inhibit cell growth, we further found by Western blotting that AKT and mTOR phosphorylation proteins (P-AKT, P-mTOR) were reduced after ARG treatment, and we used AKT agonists to rescue, and the phosphorylated proteins of AKT and mTOR increased, and we found that the autophagy-related proteins were also reversed. And interestingly, the protein of apoptosis was also reversed along with autophagy. Conclusions We thought ARG inhibited the proliferation of glioma cells by inducing autophagy and apoptosis through the AKT/mTOR pathway.
Collapse
|