1
|
Sheida A, Farshadi M, Mirzaei A, Najjar Khalilabad S, Zarepour F, Taghavi SP, Hosseini Khabr MS, Ravaei F, Rafiei S, Mosadeghi K, Yazdani MS, Fakhraie A, Ghattan A, Zamani Fard MM, Shahyan M, Rafiei M, Rahimian N, Talaei Zavareh SA, Mirzaei H. Potential of Natural Products in the Treatment of Glioma: Focus on Molecular Mechanisms. Cell Biochem Biophys 2024; 82:3157-3208. [PMID: 39150676 DOI: 10.1007/s12013-024-01447-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] [Accepted: 07/17/2024] [Indexed: 08/17/2024]
Abstract
Despite the waning of traditional treatments for glioma due to possible long-term issues, the healing possibilities of substances derived from nature have been reignited in the scientific community. These natural substances, commonly found in fruits and vegetables, are considered potential alternatives to pharmaceuticals, as they have been shown in prior research to impact pathways surrounding cancer progression, metastases, invasion, and resistance. This review will explore the supposed molecular mechanisms of different natural components, such as berberine, curcumin, coffee, resveratrol, epigallocatechin-3-gallate, quercetin, tanshinone, silymarin, coumarin, and lycopene, concerning glioma treatment. While the benefits of a balanced diet containing these compounds are widely recognized, there is considerable scope for investigating the efficacy of these natural products in treating glioma.
Collapse
Affiliation(s)
- Amirhossein Sheida
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | | | - Amirhossein Mirzaei
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Shakiba Najjar Khalilabad
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Zarepour
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Seyed Pouya Taghavi
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Maryam Sadat Hosseini Khabr
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Fatemeh Ravaei
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Sara Rafiei
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Department of Internal Medicine, School of Medicine, Firoozgar Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Kimia Mosadeghi
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Sepehr Yazdani
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Ali Fakhraie
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Alireza Ghattan
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Masoud Zamani Fard
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Maryam Shahyan
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Moein Rafiei
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Neda Rahimian
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences (IUMS), Tehran, Iran.
- Department of Internal Medicine, School of Medicine, Firoozgar Hospital, Iran University of Medical Sciences, Tehran, Iran.
| | | | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
2
|
Manica D, da Silva GB, de Lima J, Cassol J, Dallagnol P, Narzetti RA, Moreno M, Bagatini MD. Caffeine reduces viability, induces apoptosis, inhibits migration and modulates the CD39/CD73 axis in metastatic cutaneous melanoma cells. Purinergic Signal 2024; 20:385-397. [PMID: 37768408 PMCID: PMC11303616 DOI: 10.1007/s11302-023-09967-6] [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: 06/26/2023] [Accepted: 09/10/2023] [Indexed: 09/29/2023] Open
Abstract
We aimed to evaluate the effect of caffeine on viability, apoptosis, migration, redox profile and modulatory effect of the purinergic system of cutaneous melanoma cells. The melanoma cells SK-MEL-28 and non-tumoural CCD-1059sk cells were treated for 24 h with different concentrations of caffeine. Cell viability was evaluated by a biochemical assay and fluorescence microscopy, and flow cytometry assessed apoptosis induction. A wound-healing assay assessed cell migration. The redox profile was evaluated by the levels of markers of reactive oxygen species (ROS), nitric oxide (NOx), total thiols (PSH) and non-protein thiols (NPSH). RT-qPCR and flow cytometry assessed the expression of CD39 and CD73. ATPase/ADPase and AMPase enzyme activities were evaluated by hydrolysis of ATP, ADP and AMP nucleotides. A bioluminescent assay assessed extracellular ATP levels. Caffeine significantly reduced melanoma cell viability and migration and did not affect non-tumoural cells. Caffeine increased ROS levels and improved PSH levels in melanoma cells. Furthermore, caffeine reduced CD39 and CD73 expression, decreased ATP, ADP and AMP nucleotide hydrolysis and increased extracellular ATP levels. We have shown that caffeine reduces metastatic cutaneous melanoma cell viability and migration, induces ROS generation and improves PSH levels. In an unprecedented manner, we also showed that caffeine reduces the expression of CD39 and CD73 and, consequently, ATPase/ADPase/AMPase hydrolytic activity of ectonucleotidases, thus displacing the CD39/CD73 axis and increasing extracellular ATP levels. Therefore, caffeine may be an interesting compound for clinical trials with the CD39/CD73 axis as a therapeutic target.
Collapse
Affiliation(s)
- Daiane Manica
- Postgraduate Program in Biochemistry, Federal University of Santa Catarina, Florianópolis, SC, Brazil
| | - Gilnei Bruno da Silva
- Multicentric Postgraduate Program in Biochemistry and Molecular Biology, State University of Santa Catarina, Lages, SC, Brazil
| | - Jussara de Lima
- Postgraduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, Brazil
| | - Joana Cassol
- Postgraduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, Brazil
| | - Paula Dallagnol
- Postgraduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, Brazil
| | - Rafael Antônio Narzetti
- Postgraduate Program in Biochemistry, Federal University of Santa Catarina, Florianópolis, SC, Brazil
| | - Marcelo Moreno
- Postgraduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, Brazil.
| | - Margarete Dulce Bagatini
- Postgraduate Program in Biochemistry, Federal University of Santa Catarina, Florianópolis, SC, Brazil.
- Postgraduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, Brazil.
| |
Collapse
|
3
|
Makiso MU, Tola YB, Ogah O, Endale FL. Bioactive compounds in coffee and their role in lowering the risk of major public health consequences: A review. Food Sci Nutr 2024; 12:734-764. [PMID: 38370073 PMCID: PMC10867520 DOI: 10.1002/fsn3.3848] [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: 12/09/2022] [Revised: 11/04/2023] [Accepted: 11/07/2023] [Indexed: 02/20/2024] Open
Abstract
This article addresses the bioactive components in coffee aroma, their metabolism, and the mechanism of action in lowering the risk of various potential health problems. The main bioactive components involved in the perceived aroma of coffee and its related health benefits are caffeine, chlorogenic acid (CGA), trigonelline, diterpenes, and melanoids. These compounds are involved in various physiological activities. Caffeine has been shown to have anticancer properties, as well as the ability to prevent the onset and progression of hepatocellular carcinoma and to be anti-inflammatory. CGA exhibits antioxidant action and is implicated in gut health, neurodegenerative disease protection, type 2 diabetes, and cardiovascular disease prevention. Furthermore, together with diterpenes, CGA has been linked to anticancer activity. Trigonelline, on the other side, has been found to lower oxidative stress by increasing antioxidant enzyme activity and scavenging reactive oxygen species. It also prevents the formation of kidney stones. Diterpenes and melanoids possess anti-inflammatory and antioxidant properties, respectively. Consuming three to four cups of filtered coffee per day, depending on an individual's physiological condition and health status, has been linked to a lower risk of several degenerative diseases. Despite their health benefits, excessive coffee intake above the recommended daily dosage, calcium and vitamin D deficiency, and unfiltered coffee consumption all increase the risk of potential health concerns. In conclusion, moderate coffee consumption lowers the risk of different noncommunicable diseases.
Collapse
Affiliation(s)
- Markos Urugo Makiso
- Department of Food Science and Postharvest TechnologyCollege of Agricultural SciencesWachemo UniversityHossanaEthiopia
- Department of Postharvest ManagementCollege of Agriculture and Veterinary MedicineJimma UniversityJimmaEthiopia
| | - Yetenayet Bekele Tola
- Department of Postharvest ManagementCollege of Agriculture and Veterinary MedicineJimma UniversityJimmaEthiopia
| | - Onwuchekwa Ogah
- Department of Applied BiologyEbonyi State UniversityIsiekeNigeria
| | - Fitsum Liben Endale
- Department of Public HealthCollege of Medicine and Health SciencesWachemo UniversityHossanaEthiopia
| |
Collapse
|
4
|
Song X, Kirtipal N, Lee S, Malý P, Bharadwaj S. Current therapeutic targets and multifaceted physiological impacts of caffeine. Phytother Res 2023; 37:5558-5598. [PMID: 37679309 DOI: 10.1002/ptr.8000] [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: 04/13/2023] [Revised: 08/04/2023] [Accepted: 08/17/2023] [Indexed: 09/09/2023]
Abstract
Caffeine, which shares consubstantial structural similarity with purine adenosine, has been demonstrated as a nonselective adenosine receptor antagonist for eliciting most of the biological functions at physiologically relevant dosages. Accumulating evidence supports caffeine's beneficial effects against different disorders, such as total cardiovascular diseases and type 2 diabetes. Conversely, paradoxical effects are also linked to caffeine ingestion in humans including hypertension-hypotension and tachycardia-bradycardia. These observations suggest the association of caffeine action with its ingested concentration and/or concurrent interaction with preferential molecular targets to direct explicit events in the human body. Thus, a coherent analysis of the functional targets of caffeine, relevant to normal physiology, and disease pathophysiology, is required to understand the pharmacology of caffeine. This review provides a broad overview of the experimentally validated targets of caffeine, particularly those of therapeutic interest, and the impacts of caffeine on organ-specific physiology and pathophysiology. Overall, the available empirical and epidemiological evidence supports the dose-dependent functional activities of caffeine and advocates for further studies to get insights into the caffeine-induced changes under specific conditions, such as asthma, DNA repair, and cancer, in view of its therapeutic applications.
Collapse
Affiliation(s)
- Xinjie Song
- Zhejiang Provincial Key Lab for Chemical and Biological Processing Technology of Farm Product, School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou, China
| | - Nikhil Kirtipal
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Sunjae Lee
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Petr Malý
- Laboratory of Ligand Engineering, Institute of Biotechnology of the Czech Academy of Sciences v.v.i, BIOCEV Research Center, Vestec, Czech Republic
| | - Shiv Bharadwaj
- Laboratory of Ligand Engineering, Institute of Biotechnology of the Czech Academy of Sciences v.v.i, BIOCEV Research Center, Vestec, Czech Republic
| |
Collapse
|
5
|
Lin Z, Wei J, Hu Y, Pi D, Jiang M, Lang T. Caffeine Synthesis and Its Mechanism and Application by Microbial Degradation, A Review. Foods 2023; 12:2721. [PMID: 37509813 PMCID: PMC10380055 DOI: 10.3390/foods12142721] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/10/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
Caffeine is a metabolite derived from purine nucleotides, typically accounting for 2-5% of the dry weight of tea and 1-2% of the dry weight of coffee. In the tea and coffee plants, the main synthesis pathway of caffeine is a four-step sequence consisting of three methylation reactions and one nucleosidase reaction using xanthine as a precursor. In bacteria, caffeine degradation occurs mainly through the pathways of N-demethylation and C-8 oxidation. However, a study fully and systematically summarizing the metabolism and application of caffeine in microorganisms has not been established elsewhere. In the present study, we provide a review of the biosynthesis, microbial degradation, gene expression, and application of caffeine microbial degradation. The present review aims to further elaborate the mechanism of caffeine metabolism by microorganisms and explore the development prospects in this field.
Collapse
Affiliation(s)
- Zhipeng Lin
- School of Chemistry and Chemical Engineering, Guangxi Minzu University, Nanning 530008, China
- Guangxi Key Laboratory for Polysaccharide Materials and Modifications, School of Marine Sciences and Biotechnology, Guangxi Minzu University, Nanning 530008, China
| | - Jian Wei
- Institute of Ecology, College of Urban and Environmental Sciences and Key Laboratory for Earth Surface Processes of Ministry of Education, Peking University, Beijing 100091, China
| | - Yongqiang Hu
- Guangxi Key Laboratory for Polysaccharide Materials and Modifications, School of Marine Sciences and Biotechnology, Guangxi Minzu University, Nanning 530008, China
| | - Dujuan Pi
- Guangxi Key Laboratory for Polysaccharide Materials and Modifications, School of Marine Sciences and Biotechnology, Guangxi Minzu University, Nanning 530008, China
| | - Mingguo Jiang
- School of Chemistry and Chemical Engineering, Guangxi Minzu University, Nanning 530008, China
- Guangxi Key Laboratory for Polysaccharide Materials and Modifications, School of Marine Sciences and Biotechnology, Guangxi Minzu University, Nanning 530008, China
| | - Tao Lang
- MNR Key Laboratory for Geo-Environmental Monitoring of Great Bay Area & Shenzhen Key Laboratory of Marine Bioresource and Eco-Environmental Science, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518071, China
| |
Collapse
|
6
|
Liu H, Peng J, Huang L, Ruan D, Li Y, Yuan F, Tu Z, Huang K, Zhu X. The role of lysosomal peptidases in glioma immune escape: underlying mechanisms and therapeutic strategies. Front Immunol 2023; 14:1154146. [PMID: 37398678 PMCID: PMC10311646 DOI: 10.3389/fimmu.2023.1154146] [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: 01/30/2023] [Accepted: 06/02/2023] [Indexed: 07/04/2023] Open
Abstract
Glioblastoma is the most common primary malignant tumor of the central nervous system, which has the characteristics of strong invasion, frequent recurrence, and rapid progression. These characteristics are inseparable from the evasion of glioma cells from immune killing, which makes immune escape a great obstacle to the treatment of glioma, and studies have confirmed that glioma patients with immune escape tend to have poor prognosis. The lysosomal peptidase lysosome family plays an important role in the immune escape process of glioma, which mainly includes aspartic acid cathepsin, serine cathepsin, asparagine endopeptidases, and cysteine cathepsins. Among them, the cysteine cathepsin family plays a prominent role in the immune escape of glioma. Numerous studies have confirmed that glioma immune escape mediated by lysosomal peptidases has something to do with autophagy, cell signaling pathways, immune cells, cytokines, and other mechanisms, especially lysosome organization. The relationship between protease and autophagy is more complicated, and the current research is neither complete nor in-depth. Therefore, this article reviews how lysosomal peptidases mediate the immune escape of glioma through the above mechanisms and explores the possibility of lysosomal peptidases as a target of glioma immunotherapy.
Collapse
Affiliation(s)
- Hao Liu
- Department of Neurosurgery, The Second Affifiliated Hospital of Nanchang University, Nanchang, China
- The Second Clinical Medical College of Nanchang University, Nanchang, China
| | - Jie Peng
- Department of Neurosurgery, The Second Affifiliated Hospital of Nanchang University, Nanchang, China
- The Second Clinical Medical College of Nanchang University, Nanchang, China
| | - Linzhen Huang
- The Second Clinical Medical College of Nanchang University, Nanchang, China
| | - Dong Ruan
- The Second Clinical Medical College of Nanchang University, Nanchang, China
| | - Yuguang Li
- The Second Clinical Medical College of Nanchang University, Nanchang, China
| | - Fan Yuan
- The Second Clinical Medical College of Nanchang University, Nanchang, China
| | - Zewei Tu
- Department of Neurosurgery, The Second Affifiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, China
- Institute of Neuroscience, Nanchang University, Nanchang, China
- Jiangxi Health Commission (JXHC) Key Laboratory of Neurological Medicine, Nanchang, China
| | - Kai Huang
- Department of Neurosurgery, The Second Affifiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, China
- Institute of Neuroscience, Nanchang University, Nanchang, China
- Jiangxi Health Commission (JXHC) Key Laboratory of Neurological Medicine, Nanchang, China
| | - Xingen Zhu
- Department of Neurosurgery, The Second Affifiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, China
- Institute of Neuroscience, Nanchang University, Nanchang, China
- Jiangxi Health Commission (JXHC) Key Laboratory of Neurological Medicine, Nanchang, China
| |
Collapse
|
7
|
Cathepsins Trigger Cell Death and Regulate Radioresistance in Glioblastoma. Cells 2022; 11:cells11244108. [PMID: 36552871 PMCID: PMC9777369 DOI: 10.3390/cells11244108] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 12/09/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Treatment of glioblastoma (GBM) remains very challenging, and it is particularly important to find sensitive and specific molecular targets. In this work, we reveal the relationship between the expression of cathepsins and radioresistance in GBM. We analyzed cathepsins (cathepsin B, cathepsin D, cathepsin L, and cathepsin Z/X), which are highly associated with the radioresistance of GBM by regulating different types of cell death. Cathepsins could be potential targets for GBM treatment.
Collapse
|
8
|
Mitra S, Dash R, Munni YA, Selsi NJ, Akter N, Uddin MN, Mazumder K, Moon IS. Natural Products Targeting Hsp90 for a Concurrent Strategy in Glioblastoma and Neurodegeneration. Metabolites 2022; 12:1153. [PMID: 36422293 PMCID: PMC9697676 DOI: 10.3390/metabo12111153] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/16/2022] [Accepted: 11/16/2022] [Indexed: 09/16/2023] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most common aggressive, resistant, and invasive primary brain tumors that share neurodegenerative actions, resembling many neurodegenerative diseases. Although multiple conventional approaches, including chemoradiation, are more frequent in GBM therapy, these approaches are ineffective in extending the mean survival rate and are associated with various side effects, including neurodegeneration. This review proposes an alternative strategy for managing GBM and neurodegeneration by targeting heat shock protein 90 (Hsp90). Hsp90 is a well-known molecular chaperone that plays essential roles in maintaining and stabilizing protein folding to degradation in protein homeostasis and modulates signaling in cancer and neurodegeneration by regulating many client protein substrates. The therapeutic benefits of Hsp90 inhibition are well-known for several malignancies, and recent evidence highlights that Hsp90 inhibitors potentially inhibit the aggressiveness of GBM, increasing the sensitivity of conventional treatment and providing neuroprotection in various neurodegenerative diseases. Herein, the overview of Hsp90 modulation in GBM and neurodegeneration progress has been discussed with a summary of recent outcomes on Hsp90 inhibition in various GBM models and neurodegeneration. Particular emphasis is also given to natural Hsp90 inhibitors that have been evidenced to show dual protection in both GBM and neurodegeneration.
Collapse
Affiliation(s)
- Sarmistha Mitra
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju 38066, Republic of Korea
| | - Raju Dash
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju 38066, Republic of Korea
| | - Yeasmin Akter Munni
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju 38066, Republic of Korea
| | - Nusrat Jahan Selsi
- Product Development Department, Popular Pharmaceuticals Ltd., Dhaka 1207, Bangladesh
| | - Nasrin Akter
- Department of Clinical Pharmacy and Molecular Pharmacology, East West University Bangladesh, Dhaka 1212, Bangladesh
| | - Md Nazim Uddin
- Department of Pharmacy, Southern University Bangladesh, Chittagong 4000, Bangladesh
| | - Kishor Mazumder
- Department of Pharmacy, Jashore University of Science and Technology, Jashore 7408, Bangladesh
- School of Optometry and Vision Science, UNSW Medicine, University of New South Wales (UNSW), Sydney, NSW 2052, Australia
| | - Il Soo Moon
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju 38066, Republic of Korea
| |
Collapse
|
9
|
Caffeine Inhibits NLRP3 Inflammasome Activation by Downregulating TLR4/MAPK/NF-κB Signaling Pathway in an Experimental NASH Model. Int J Mol Sci 2022; 23:ijms23179954. [PMID: 36077357 PMCID: PMC9456282 DOI: 10.3390/ijms23179954] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 08/26/2022] [Accepted: 08/29/2022] [Indexed: 12/13/2022] Open
Abstract
Caffeine elicits protective effects against liver diseases, such as NASH; however, its mechanism of action involving the pyrin domain-containing-3 (NLRP3) inflammasome signaling pathway remains to be elucidated. This study aimed to evaluate the effect of caffeine on the NLRP3 inflammasome signaling pathway in a rat model of NASH. NASH was induced by feeding rats a high-fat, -sucrose, and -cholesterol diet (HFSCD) for 15 weeks along with a weekly low dose (400 mg/kg, i.p.) of CCl4. Caffeine was administered at 50 mg/kg p.o. The effects of HFSCD+CCl4 and caffeine on the liver were evaluated using biochemical, ultrastructural, histological, and molecular biological approaches. The HFSCD+CCl4-treated rats showed fat accumulation in the liver, elevated levels of inflammatory mediators, NLRP3 inflammasome activation, antioxidant dysregulation, and liver fibrosis. Caffeine reduced necrosis, cholestasis, oxidative stress, and fibrosis. Caffeine exhibited anti-inflammatory effects by attenuating NLRP3 inflammasome activation. Moreover, caffeine prevented increases in toll-like receptor 4 (TLR4) and nuclear factor-κB (NF-κB) protein levels and mitigated the phosphorylation of mitogen-activated protein kinase (MAPK). Importantly, caffeine prevented the activation of hepatic stellate cells. This study is the first to report that caffeine ameliorates NASH by inhibiting NLRP3 inflammasome activation through the suppression of the TLR4/MAPK/NF-κB signaling pathway.
Collapse
|
10
|
Du C, Huang Z, Wei B, Li M. Comprehensive metabolomics study on the pathogenesis of anaplastic astrocytoma via UPLC-Q/TOF-MS. Medicine (Baltimore) 2022; 101:e29594. [PMID: 35945752 PMCID: PMC9351860 DOI: 10.1097/md.0000000000029594] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Anaplastic astrocytoma (AA) is a malignant carcinoma whose pathogenesis remains to be fully elucidated. System biology techniques have been widely used to clarify the mechanism of diseases from a systematic perspective. The present study aimed to explore the pathogenesis and novel potential biomarkers for the diagnosis of AA according to metabolic differences. Patients with AA (n = 12) and healthy controls (n = 15) were recruited. Serum was assayed with untargeted ultraperformance liquid chromatography-quadrupole/time-of-flight-mass spectrometry (UPLC-Q/TOF-MS) metabolomic techniques. The data were further evaluated using multivariate analysis and bioinformatic methods based on the KEGG database to determine the distinct metabolites and perturbed pathways. Principal component analysis and orthogonal projections to latent structures-discriminant analysis (OPLS-DA) identified the significance of the distinct metabolic pattern between patients with AA and healthy controls (P < .001) in both ESI modes. Permutation testing confirmed the validity of the OPLS-DA model (permutation = 200, Q2 < 0.5). In total, 24 differentiated metabolites and 5 metabolic pathways, including sphingolipid, glycerophospholipid, caffeine, linoleic acid, and porphyrin metabolism, were identified based on the OPLS-DA model. 3-Methylxanthine, sphinganine, LysoPC(18:1), and lactosylceramide were recognized as potential biomarkers with excellent sensitivity and specificity (area under the curve > 98%). These findings indicate that the perturbed metabolic pattern related to immune regulation and cellular signal transduction is associated with the pathogenesis of AA. 3-Methylxanthine, sphinganine, LysoPC(18:1), and lactosylceramide could be used as biomarkers of AA in future clinical practice. This study provides a therapeutic basis for further studies on the mechanism and precise clinical diagnosis of AA.
Collapse
Affiliation(s)
- Chao Du
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin
| | - Zhehao Huang
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin
| | - Bo Wei
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin
| | - Miao Li
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin
- * Correspondence: Miao Li, MD, Department of Neurosurgery, China-Japan Union Hospital of Jilin University, 126 Xiantai Street, Changchun, Jilin 130033, PR China (e-mail: )
| |
Collapse
|
11
|
Rudzinska-Radecka M, Frolova AS, Balakireva AV, Gorokhovets NV, Pokrovsky VS, Sokolova DV, Korolev DO, Potoldykova NV, Vinarov AZ, Parodi A, Zamyatnin AA. In Silico, In Vitro, and Clinical Investigations of Cathepsin B and Stefin A mRNA Expression and a Correlation Analysis in Kidney Cancer. Cells 2022; 11:1455. [PMID: 35563761 PMCID: PMC9101197 DOI: 10.3390/cells11091455] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 04/18/2022] [Accepted: 04/21/2022] [Indexed: 02/06/2023] Open
Abstract
The cysteine protease Cathepsin B (CtsB) plays a critical role in multiple signaling pathways, intracellular protein degradation, and processing. Endogenous inhibitors regulate its enzymatic activity, including stefins and other cystatins. Recent data proved that CtsB is implicated in tumor extracellular matrix remodeling, cell invasion, and metastasis: a misbalance between cathepsins and their natural inhibitors is often considered a sign of disease progression. In the present study, we investigated CtsB and stefin A (StfA) expression in renal cell carcinoma (RCC). mRNA analysis unveiled a significant CTSB and STFA increase in RCC tissues compared to adjacent non-cancerogenic tissues and a higher CtsB expression in malignant tumors than in benign renal neoplasms. Further analysis highlighted a positive correlation between CtsB and StfA expression as a function of patient sex, age, tumor size, grade, lymph node invasion, metastasis occurrence, and survival. Alternative overexpression and silencing of CtsB and StfA confirmed the correlation expression between these proteins in human RCC-derived cells through protein analysis and fluorescent microscopy. Finally, the ectopic expression of CtsB and StfA increased RCC cell proliferation. Our data strongly indicated that CtsB and StfA expression play an important role in RCC development by mutually stimulating their expression in RCC progression.
Collapse
Affiliation(s)
- Magdalena Rudzinska-Radecka
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (M.R.-R.); (A.S.F.); (A.V.B.); (N.V.G.); (A.P.)
- Institute of Physical Chemistry, Polish Academy of Sciences, 01-224 Warsaw, Poland
| | - Anastasia S. Frolova
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (M.R.-R.); (A.S.F.); (A.V.B.); (N.V.G.); (A.P.)
- Department of Biotechnology, Sirius University of Science and Technology, 1 Olympic Ave., 354340 Sochi, Russia; (V.S.P.); (D.V.S.)
| | - Anastasia V. Balakireva
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (M.R.-R.); (A.S.F.); (A.V.B.); (N.V.G.); (A.P.)
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia
| | - Neonila V. Gorokhovets
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (M.R.-R.); (A.S.F.); (A.V.B.); (N.V.G.); (A.P.)
| | - Vadim S. Pokrovsky
- Department of Biotechnology, Sirius University of Science and Technology, 1 Olympic Ave., 354340 Sochi, Russia; (V.S.P.); (D.V.S.)
- Laboratory of Combined Treatment, N.N. Blokhin Cancer Research Center, 115478 Moscow, Russia
- Department of Biochemistry, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Darina V. Sokolova
- Department of Biotechnology, Sirius University of Science and Technology, 1 Olympic Ave., 354340 Sochi, Russia; (V.S.P.); (D.V.S.)
- Laboratory of Combined Treatment, N.N. Blokhin Cancer Research Center, 115478 Moscow, Russia
- Department of Biochemistry, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Dmitry O. Korolev
- Institute for Urology and Reproductive Health, Sechenov University, 119992 Moscow, Russia; (D.O.K.); (N.V.P.); (A.Z.V.)
| | - Natalia V. Potoldykova
- Institute for Urology and Reproductive Health, Sechenov University, 119992 Moscow, Russia; (D.O.K.); (N.V.P.); (A.Z.V.)
| | - Andrey Z. Vinarov
- Institute for Urology and Reproductive Health, Sechenov University, 119992 Moscow, Russia; (D.O.K.); (N.V.P.); (A.Z.V.)
| | - Alessandro Parodi
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (M.R.-R.); (A.S.F.); (A.V.B.); (N.V.G.); (A.P.)
- Department of Biotechnology, Sirius University of Science and Technology, 1 Olympic Ave., 354340 Sochi, Russia; (V.S.P.); (D.V.S.)
| | - Andrey A. Zamyatnin
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (M.R.-R.); (A.S.F.); (A.V.B.); (N.V.G.); (A.P.)
- Department of Biotechnology, Sirius University of Science and Technology, 1 Olympic Ave., 354340 Sochi, Russia; (V.S.P.); (D.V.S.)
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
- Department of Immunology, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, UK
| |
Collapse
|
12
|
Ma K, Chen X, Liu W, Chen S, Yang C, Yang J. CTSB is a negative prognostic biomarker and therapeutic target associated with immune cells infiltration and immunosuppression in gliomas. Sci Rep 2022; 12:4295. [PMID: 35277559 PMCID: PMC8917123 DOI: 10.1038/s41598-022-08346-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 03/07/2022] [Indexed: 12/29/2022] Open
Abstract
Previous researches have demonstrated the meaning of CTSB for the progress of several tumors, whereas few clues about its immunological characteristic in gliomas. Here we systematically explored its biologic features and clinical significance for gliomas. 699 glioma cases of TCGA and 325 glioma cases of CGGA were respectively included as training and validating cohorts. R software was used for data analysis and mapping. We found that CTSB was remarkably highly-expressed for HGG, IDH wild type, 1p19q non-codeletion type, MGMT promoter unmethylation type and mesenchymal gliomas. CTSB could specifically and sensitively indicate mesenchymal glioma. Upregulated CTSB was an independent hazard correlated with poor survival. CTSB-related biological processes in gliomas chiefly concentrated on immunoreaction and inflammation response. Then we proved that CTSB positively related to most inflammatory metagenes except IgG, including HCK, LCK, MHC II, STAT1 and IFN. More importantly, the levels of glioma-infiltrating immune cells were positively associated with the expression of CTSB, especially for TAMs, MDSCs and Tregs. In conclusion, CTSB is closely related to the malignant pathological subtypes, worse prognosis, immune cells infiltration and immunosuppression of gliomas, which make it a promising biomarker and potential target in the diagnosis, treatment and prognostic assessment of gliomas.
Collapse
Affiliation(s)
- Kaiming Ma
- Department of Neurosurgery, Peking University Third Hospital, 49 North Garden Rd, Haidian District, Beijing, 100191, China.,Center for Precision Neurosurgery and Oncology of Peking University Health Science Center, Beijing, China
| | - Xin Chen
- Department of Neurosurgery, Peking University Third Hospital, 49 North Garden Rd, Haidian District, Beijing, 100191, China.,Center for Precision Neurosurgery and Oncology of Peking University Health Science Center, Beijing, China
| | - Weihai Liu
- Department of Neurosurgery, Peking University Third Hospital, 49 North Garden Rd, Haidian District, Beijing, 100191, China.,Center for Precision Neurosurgery and Oncology of Peking University Health Science Center, Beijing, China
| | - Suhua Chen
- Department of Neurosurgery, Peking University Third Hospital, 49 North Garden Rd, Haidian District, Beijing, 100191, China.,Center for Precision Neurosurgery and Oncology of Peking University Health Science Center, Beijing, China
| | - Chenlong Yang
- Department of Neurosurgery, Peking University Third Hospital, 49 North Garden Rd, Haidian District, Beijing, 100191, China
| | - Jun Yang
- Department of Neurosurgery, Peking University Third Hospital, 49 North Garden Rd, Haidian District, Beijing, 100191, China. .,Center for Precision Neurosurgery and Oncology of Peking University Health Science Center, Beijing, China.
| |
Collapse
|
13
|
Jing X, Hu H, Sun Y, Yu B, Cong H, Shen Y. The Intracellular and Extracellular Microenvironment of Tumor Site: The Trigger of Stimuli-Responsive Drug Delivery Systems. SMALL METHODS 2022; 6:e2101437. [PMID: 35048560 DOI: 10.1002/smtd.202101437] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/29/2021] [Indexed: 06/14/2023]
Abstract
The tumor microenvironment (TME), including intracellular and extracellular microenvironment, contains many biochemical indicators (such as acidity/alkalinity, oxygen content, and enzymatic activity) that are different from the normal physiological environment. These abnormal biochemical indicators can accelerate the heterogeneity of tumors, but on the other hand, they also provide opportunities for the design of intelligent drug delivery systems (DDSs). The TME-responsive DDSs have shown great potential in reducing the side effects of chemotherapy and improving the curative effect of tumors. In this review, the abnormal biochemical indicators of TME are introduced in detail from both the extracellular and intracellular aspects. In view of the various physiological barriers encountered during drug delivery, the strategy of constructing TME-responsive DDSs is discussed. By summarizing the typical research progress, the authors prospect the development of TME-responsive DDS in the future.
Collapse
Affiliation(s)
- Xiaodong Jing
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, China
| | - Hao Hu
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, China
| | - Yanzhen Sun
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, China
| | - Bing Yu
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, China
- State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao, 266071, China
| | - Hailin Cong
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, China
- State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao, 266071, China
| | - Youqing Shen
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, Center for Bionanoengineering, and Department of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang, 310027, China
| |
Collapse
|
14
|
Bonafé GA, Boschiero MN, Sodré AR, Ziegler JV, Rocha T, Ortega MM. Natural Plant Compounds: Does Caffeine, Dipotassium Glycyrrhizinate, Curcumin, and Euphol Play Roles as Antitumoral Compounds in Glioblastoma Cell Lines? Front Neurol 2022; 12:784330. [PMID: 35300350 PMCID: PMC8923017 DOI: 10.3389/fneur.2021.784330] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 11/29/2021] [Indexed: 12/21/2022] Open
Abstract
Many plant-derived compounds are shown to be promising antitumor therapeutic agents by enhancing apoptosis-related pathways and cell cycle impairment in tumor cells, including glioblastoma (GBM) cell lines. We aimed to review four natural plant compounds effective in GBM cell lines as caffeine, dipotassium glycyrrhizinate (DPG), curcumin, and euphol. Furthermore, antitumoral effect of these plant compounds on GBM cell lines through microRNAs (miRs) modulation was investigated. However, only DPG and curcumin were found as effective on miR modulation. Caffeine arrests GBM cell cycle in G0/G1 phase by cyclin-dependent kinases (CDK) complex inhibition and by decreasing BCL-2 and increasing FOXO1 expression levels causing greater apoptotic activity. Caffeine can also directly inhibit IP3R3, p38 phosphorylation, and rho-associated protein kinase (ROCK), decreasing cell invasion and migration capacity or indirectly by inhibiting the tissue inhibitor metalloproteinase-1 (TIMP-1) and integrins β1 and β3, leading to lower matrix metalloproteinases, MMP-2 and MMP-9. DPG presents antitumoral effect in GBM cells related to nuclear factor kappa B (NF-κB) pathway suppression by IRAK2 and TRAF6-mediating miR-16 and miR-146a, respectively. More recently, it was observed that DPG upregulated miR-4443 and miR-3620, responsible for post-transcriptional inhibition of the NF-κB pathway by CD209 and TNC modulation, respectively leading to lower MMP-9 and migration capacity. Curcumin is able to increase miR-223-3p, miR-133a-3p, miR-181a-5p, miR-34a-5p, miR-30c-5p, and miR-1290 expression leading to serine or threonine kinase (AKT) pathway impairment and also it decreases miR-27a-5p, miR-221-3p, miR-21-5p, miR-125b-5p, and miR-151-3p expression causing p53-BCL2 pathway inhibition and consequently, cellular apoptosis. Interestingly, lower expression of miR-27a by curcumin action enhanced the C/EBP homologous protein(CHOP) expression, leading to paraptosis. Curcumin can inhibit miR-21 expression and consequently activate apoptosis through caspase 3 and death receptor (DR) 4 and 5 activation. Autophagy is controlled by the LC-3 protein that interacts with Atg family for the LC3-II formation and autophagy activation. Euphol can enhance LC3-II levels directly in GBM cells or inhibits tumor invasion and migration through PDK1 modulation.
Collapse
Affiliation(s)
- Gabriel Alves Bonafé
- Laboratory of Cell and Molecular Tumor Biology and Bioactive Compounds, São Francisco University Medical School, São Paulo, Brazil
| | - Matheus Negri Boschiero
- Laboratory of Cell and Molecular Tumor Biology and Bioactive Compounds, São Francisco University Medical School, São Paulo, Brazil
| | - André Rodrigues Sodré
- Laboratory of Cell and Molecular Tumor Biology and Bioactive Compounds, São Francisco University Medical School, São Paulo, Brazil
| | | | - Thalita Rocha
- Postgraduate Program in Biomaterials and Regenerative Medicine, Faculty of Medical Sciences and Health, Pontifical Catholic University of São Paulo, São Paulo, Brazil
| | - Manoela Marques Ortega
- Laboratory of Cell and Molecular Tumor Biology and Bioactive Compounds, São Francisco University Medical School, São Paulo, Brazil
- *Correspondence: Manoela Marques Ortega
| |
Collapse
|
15
|
Antibacterial, Antiradical and Antiproliferative Potential of Green, Roasted, and Spent Coffee Extracts. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12041938] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The phytochemical compositions of green coffee beans (GB), roasted coffee (RC), and the solid residue known as spent coffee grounds (SCG) have been associated with beneficial physiological effects. The objective of this study was to analyze the total phenolic compounds, antiradical scavenging ability, antibacterial activity, and antiproliferative activity on cancer cells of aqueous and ethanolic extracts of GB, RC, and SCG samples. The total phenolic content was quantified by Folin–Ciocalteu assay, while the antiradical activity was evaluated by ABTS●+ and DPPH radical assays, antibacterial activity was determined using the microtiter broth dilution method, and antiproliferative activity was evaluated by MTT assay in lung carcinoma cells (A549) and cervical cancer cells (C33A); furthermore, apoptosis and cell cycle arrest were evaluated by flow cytometry. Ethanolic extracts of RC and SCG showed the highest content of total phenols. The SCG ethanolic extract exhibited the lowest inhibitory capacity 50 (IC50) values for free radicals. The SCG extracts also had the lowest MIC values in bacteria. In antiproliferative assays, SCG extracts exhibited a significant decrease in viability in both cell lines, as well as increased apoptotic cells and promoted cell cycle arrest. The higher content of total phenols and antiradical activity of SCG ethanolic extracts was related to their antiproliferative activity in cancer cells, as well as their antibacterial activity against clinical isolates; therefore, the utilization of SCG adds value to an abundant and inexpensive residue.
Collapse
|
16
|
Pranata R, Feraldho A, Lim MA, Henrina J, Vania R, Golden N, July J. Coffee and tea consumption and the risk of glioma: a systematic review and dose-response meta-analysis. Br J Nutr 2022; 127:78-86. [PMID: 33750490 DOI: 10.1017/s0007114521000830] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In this systematic review and dose-response meta-analysis, we aimed to assess whether coffee and tea consumption is related to the risk of glioma. We performed a systematic literature search using PubMed, Embase, Scopus and the EuropePMC from the inception of database up until 1 October 2020. Exposures in the present study were coffee and tea consumption, the main outcome was the incidence of glioma. The present study compares the association between the exposure of coffee and tea with the incidence of glioma, and the results are reported in relative risks (RR). There are 12 unique studies comprising of 1 960 731 participants with 2987 glioma cases. Higher coffee consumption was associated with a statistically non-significant trend towards lower risk of glioma (RR 0·77 (95 % CI 0·55, 1·03), P= 0·11; I2:75·27 %). Meta-regression showed that the association between coffee and glioma was reduced by smoking (P= 0·029). Higher tea consumption was associated with a lower risk of glioma (RR 0·84 (95 % CI 0·71, 0·98), P= 0·030; I2:16·42 %). Sensitivity analysis by removal of case-control studies showed that higher coffee consumption (RR 0·85 (95 % CI 0·72, 1·00), P= 0·046; I2:0 %) and higher tea consumption (RR 0·81 (95 % CI 0·70, 0·93), P= 0·004; I2:0 %, Pnon-linearity = 0·140) were associated with lower risk of glioma. Dose-response meta-analysis showed that every one cup of coffee per day decreases the risk of glioma by 3 % (RR 0·97 (95 % CI 0·94, 0·99), P= 0·016, Pnon-linearity = 0·054) and every one cup of tea per day decreases the risk of glioma by 3 % (RR 0·97 (95 % CI 0·94, 1·00), P= 0·048). This meta-analysis showed apparent association between coffee and tea intake and risk of glioma.
Collapse
Affiliation(s)
- Raymond Pranata
- Faculty of Medicine, Universitas Pelita Harapan, Tangerang, Indonesia
| | - Andrea Feraldho
- Clinical and Public Health Nutrition, Division of Medicine, University College London, London, UK
| | | | | | - Rachel Vania
- Faculty of Medicine, Universitas Pelita Harapan, Tangerang, Indonesia
- Division of Plastic, Reconstructive and Aesthetic, Department of Surgery, Faculty of Medicine, Udayana University, Sanglah General Hospital, Denpasar, Bali, Indonesia
| | - Nyoman Golden
- Department of Neurosurgery, Sanglah General Hospital, School of Medicine, Udayana University, Denpasar, Bali, Indonesia
| | - Julius July
- Department of Neurosurgery, Medical Faculty of Pelita Harapan University, Neuroscience Centre Siloam Hospital, Lippo Village Tangerang, Indonesia
| |
Collapse
|
17
|
Treatment of glioblastoma with re-purposed renin-angiotensin system modulators: Results of a phase I clinical trial. J Clin Neurosci 2021; 95:48-54. [PMID: 34929651 DOI: 10.1016/j.jocn.2021.11.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 10/16/2021] [Accepted: 11/21/2021] [Indexed: 12/15/2022]
Abstract
Glioblastoma is the most common and most aggressive primary brain cancer in adults. Standard treatment of glioblastoma consisting of maximal safe resection, adjuvant radiotherapy and chemotherapy with temozolomide, results in an overall median survival of 14.6 months. The aggressive nature of glioblastoma has been attributed to the presence of glioblastoma stem cells which express components of the renin-angiotensin system (RAS). This phase I clinical trial investigated the tolerability and efficacy of a treatment targeting the RAS and its converging pathways in patients with glioblastoma. Patients who had relapsed following standard treatment of glioblastoma who met the trial criteria were commenced on dose-escalated oral RAS modulators (propranolol, aliskiren, cilazapril, celecoxib, curcumin with piperine, aspirin, and metformin). Of the 17 patients who were enrolled, ten completed full dose-escalation of the treatment. The overall median survival was 19.9 (95% CI:14.1-25.7) months. Serial FET-PET/CTs showed a reduction in both tumor volume and uptake in one patient, an increase in tumor uptake in nine patients with decreased (n = 1), unchanged (n = 1) and increased (n = 7) tumor volume, in the ten patients who had completed full dose-escalation of the treatment. Two patients experienced mild side effects and all patients had preservation of quality of life and performance status during the treatment. There is a trend towards increased survival by 5.3 months although it was not statistically significant. These encouraging results warrant further clinical trials on this potential novel, well-tolerated and cost-effective therapeutic option for patients with glioblastoma.
Collapse
|
18
|
Meisaprow P, Aksorn N, Vinayanuwattikun C, Chanvorachote P, Sukprasansap M. Caffeine Induces G0/G1 Cell Cycle Arrest and Inhibits Migration through Integrin αv, β3, and FAK/Akt/c-Myc Signaling Pathway. Molecules 2021; 26:7659. [PMID: 34946741 PMCID: PMC8706725 DOI: 10.3390/molecules26247659] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/14/2021] [Accepted: 12/15/2021] [Indexed: 12/12/2022] Open
Abstract
Lung cancer is recognized as a major cause of mortality worldwide owing to its metastatic activity. Given the lack of solid information regarding the possible effects of caffeine, one of the most consumed natural psychoactive substances, on molecular signaling pathways implicated in the aggressive behavior of lung cancer, our study aimed to evaluate the effect and mechanism of caffeine on metastasis-related mechanisms. The results revealed that caffeine treatment at concentrations of 0-500 µM caused no direct cytotoxic effects on NCI-H23 cells. Treatment of cells with caffeine showed good potential to inhibit cell proliferation at 48 h and induced significant cell cycle arrest at the G0/G1 phase. Concerning metastasis, caffeine was shown to reduce filopodia formation, inhibit migration and invasion capability, and reduce the ability of cancer cells to survive and grow in an anchorage-independent manner. Moreover, caffeine could attenuate the formation of 3D tumor spheroids in cancer stem cell (CSC)-enriched populations. With regard to mechanisms, we found that caffeine significantly altered the integrin pattern of the treated cells and caused the downregulation of metastasis-associated integrins, namely, integrins αv and β3. Subsequently, the downstream signals, including protein signaling and transcription factors, namely, phosphorylated focal adhesion kinase (p-FAK), phosphorylated protein kinase B (p-Akt), cell division cycle 42 (Cdc42), and c-Myc, were significantly decreased in caffeine-exposed cells. Taken together, our novel data on caffeine-inhibiting mechanism in relation to metastasis in lung cancer could provide insights into the impact of caffeine intake on human diseases and conditions.
Collapse
Affiliation(s)
- Pichitchai Meisaprow
- Graduate Student in Master of Science Program in Nutrition, Faculty of Medicine Ramathibodi Hospital and Institute of Nutrition, Mahidol University, Bangkok 10400, Thailand;
| | - Nithikoon Aksorn
- Department of Clinical Pathology, Faculty of Medicine Vajira Hospital, Navamindradhiraj University, Bangkok 10300, Thailand;
| | - Chanida Vinayanuwattikun
- Division of Medical Oncology, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand;
| | - Pithi Chanvorachote
- Cell-Based Drug and Health Product Development Research Unit, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Monruedee Sukprasansap
- Food Toxicology Unit, Institute of Nutrition, Mahidol University, Salaya Campus, Nakhon Pathom 73170, Thailand
| |
Collapse
|
19
|
Tilaoui M, Ait Mouse H, Zyad A. Update and New Insights on Future Cancer Drug Candidates From Plant-Based Alkaloids. Front Pharmacol 2021; 12:719694. [PMID: 34975465 PMCID: PMC8716855 DOI: 10.3389/fphar.2021.719694] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 11/23/2021] [Indexed: 02/06/2023] Open
Abstract
Cancer is a complex multifactorial disease that results from alterations in many physiological and biochemical functions. Over the last few decades, it has become clear that cancer cells can acquire multidrug resistance to conventional anticancer drugs, resulting in tumor relapse. Thus, there is a continuous need to discover new and effective anticancer drugs. Natural products from plants have served as a primary source of cancer drugs and continue to provide new plant-derived anticancer drugs. The present review describes plant-based alkaloids, which have been reported as active or potentially active in cancer treatment within the past 4 years (2017-2020), both in preclinical research and/or in clinical trials. In addition, recent insights into the possible molecular mechanism of action of alkaloid prodrugs naturally present in plants are also highlighted.
Collapse
Affiliation(s)
- Mounir Tilaoui
- Experimental Oncology and Natural Substances Team, Cellular and Molecular Immuno-pharmacology, Faculty of Sciences and Technology, Sultan Moulay Slimane University, Beni-Mellal, Morocco
| | | | | |
Collapse
|
20
|
The Renin-Angiotensin System in the Tumor Microenvironment of Glioblastoma. Cancers (Basel) 2021; 13:cancers13164004. [PMID: 34439159 PMCID: PMC8392691 DOI: 10.3390/cancers13164004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 08/03/2021] [Accepted: 08/06/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Glioblastoma (GB) is the most aggressive brain cancer in humans. Patient survival outcomes have remained dismal despite intensive research over the past 50 years, with a median overall survival of only 14.6 months. We highlight the critical role of the renin–angiotensin system (RAS) on GB cancer stem cells and the tumor microenvironment which, in turn, influences cancer stem cells in driving tumorigenesis and treatment resistance. We present recent developments and underscore the need for further research into the GB tumor microenvironment. We discuss the novel therapeutic targeting of the RAS using existing commonly available medications and utilizing model systems to further this critical investigation. Abstract Glioblastoma (GB) is an aggressive primary brain tumor. Despite intensive research over the past 50 years, little advance has been made to improve the poor outcome, with an overall median survival of 14.6 months following standard treatment. Local recurrence is inevitable due to the quiescent cancer stem cells (CSCs) in GB that co-express stemness-associated markers and components of the renin–angiotensin system (RAS). The dynamic and heterogeneous tumor microenvironment (TME) plays a fundamental role in tumor development, progression, invasiveness, and therapy resistance. There is increasing evidence showing the critical role of the RAS in the TME influencing CSCs via its upstream and downstream pathways. Drugs that alter the hallmarks of cancer by modulating the RAS present a potential new therapeutic alternative or adjunct to conventional treatment of GB. Cerebral and GB organoids may offer a cost-effective method for evaluating the efficacy of RAS-modulating drugs on GB. We review the nexus between the GB TME, CSC niche, and the RAS, and propose re-purposed RAS-modulating drugs as a potential therapeutic alternative or adjunct to current standard therapy for GB.
Collapse
|
21
|
Travers S, Litofsky NS. Daily Lifestyle Modifications to Improve Quality of Life and Survival in Glioblastoma: A Review. Brain Sci 2021; 11:brainsci11050533. [PMID: 33922443 PMCID: PMC8146925 DOI: 10.3390/brainsci11050533] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 04/19/2021] [Accepted: 04/22/2021] [Indexed: 12/21/2022] Open
Abstract
Survival in glioblastoma remains poor despite advancements in standard-of-care treatment. Some patients wish to take a more active role in their cancer treatment by adopting daily lifestyle changes to improve their quality of life or overall survival. We review the available literature through PubMed and Google Scholar to identify laboratory animal studies, human studies, and ongoing clinical trials. We discuss which health habits patients adopt and which have the most promise in glioblastoma. While results of clinical trials available on these topics are limited, dietary restrictions, exercise, use of supplements and cannabis, and smoking cessation all show some benefit in the comprehensive treatment of glioblastoma. Marital status also has an impact on survival. Further clinical trials combining standard treatments with lifestyle modifications are necessary to quantify their survival advantages.
Collapse
|
22
|
Quesnel A, Karagiannis GS, Filippou PS. Extracellular proteolysis in glioblastoma progression and therapeutics. Biochim Biophys Acta Rev Cancer 2020; 1874:188428. [PMID: 32956761 DOI: 10.1016/j.bbcan.2020.188428] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 09/03/2020] [Accepted: 09/04/2020] [Indexed: 12/20/2022]
Abstract
Gliomas encompass highly invasive primary central nervous system (CNS) tumours of glial cell origin with an often-poor clinical prognosis. Of all gliomas, glioblastoma is the most aggressive form of primary brain cancer. Current treatments in glioblastoma are insufficient due to the invasive nature of brain tumour cells, which typically results in local tumour recurrence following treatment. The latter represents the most important cause of mortality in glioblastoma and underscores the necessity for an in-depth understanding of the underlying mechanisms. Interestingly, increased synthesis and secretion of several proteolytic enzymes within the tumour microenvironment, such as matrix metalloproteinases, lysosomal proteases, cathepsins and kallikreins for extracellular-matrix component degradation may play a major role in the aforementioned glioblastoma invasion mechanisms. These proteolytic networks are key players in establishing and maintaining a tumour microenvironment that promotes tumour cell survival, proliferation, and migration. Indeed, the targeted inhibition of these proteolytic enzymes has been a promisingly useful therapeutic strategy for glioblastoma management in both preclinical and clinical development. We hereby summarize current advances on the biology of the glioblastoma tumour microenvironment, with a particular emphasis on the role of proteolytic enzyme families in glioblastoma invasion and progression, as well as on their subsequent prognostic value as biomarkers and their therapeutic targeting in the era of precision medicine.
Collapse
Affiliation(s)
- Agathe Quesnel
- School of Health & Life Sciences, Teesside University, Middlesbrough TS1 3BX, United Kingdom; National Horizons Centre, Teesside University, 38 John Dixon Ln, Darlington, DL1 1HG, United Kingdom
| | - George S Karagiannis
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, New York, USA; Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, New York, USA; Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Panagiota S Filippou
- School of Health & Life Sciences, Teesside University, Middlesbrough TS1 3BX, United Kingdom; National Horizons Centre, Teesside University, 38 John Dixon Ln, Darlington, DL1 1HG, United Kingdom.
| |
Collapse
|
23
|
Tumor microenvironment-induced structure changing drug/gene delivery system for overcoming delivery-associated challenges. J Control Release 2020; 323:203-224. [PMID: 32320817 DOI: 10.1016/j.jconrel.2020.04.026] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 04/15/2020] [Accepted: 04/16/2020] [Indexed: 02/07/2023]
Abstract
Nano-drug/gene delivery systems (DDS) are powerful weapons for the targeted delivery of various therapeutic molecules in treatment of tumors. Nano systems are being extensively investigated for drug and gene delivery applications because of their exceptional ability to protect the payload from degradation in vivo, prolong circulation of the nanoparticles (NPs), realize controlled release of the contents, reduce side effects, and enhance targeted delivery among others. However, the specific properties required for a DDS vary at different phase of the complex delivery process, and these requirements are often conflicting, including the surface charge, particle size, and stability of DDS, which severely reduces the efficiency of the drug/gene delivery. Therefore, researchers have attempted to fabricate structure, size, or charge changeable DDS by introducing various tumor microenvironment (TME) stimuli-responsive elements into the DDS to meet the varying requirements at different phases of the delivery process, thus improving drug/gene delivery efficiency. This paper summarizes the most recent developments in TME stimuli-responsive DDS and addresses the aforementioned challenges.
Collapse
|
24
|
Efferth T, Oesch F. Repurposing of plant alkaloids for cancer therapy: Pharmacology and toxicology. Semin Cancer Biol 2019; 68:143-163. [PMID: 31883912 DOI: 10.1016/j.semcancer.2019.12.010] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 12/15/2019] [Indexed: 02/08/2023]
Abstract
Drug repurposing (or repositioning) is an emerging concept to use old drugs for new treatment indications. Phytochemicals isolated from medicinal plants have been largely neglected in this context, although their pharmacological activities have been well investigated in the past, and they may have considerable potentials for repositioning. A grand number of plant alkaloids inhibit syngeneic or xenograft tumor growth in vivo. Molecular modes of action in cancer cells include induction of cell cycle arrest, intrinsic and extrinsic apoptosis, autophagy, inhibition of angiogenesis and glycolysis, stress and anti-inflammatory responses, regulation of immune functions, cellular differentiation, and inhibition of invasion and metastasis. Numerous underlying signaling processes are affected by plant alkaloids. Furthermore, plant alkaloids suppress carcinogenesis, indicating chemopreventive properties. Some plant alkaloids reveal toxicities such as hepato-, nephro- or genotoxicity, which disqualifies them for repositioning purposes. Others even protect from hepatotoxicity or cardiotoxicity of xenobiotics and established anticancer drugs. The present survey of the published literature clearly demonstrates that plant alkaloids have the potential for repositioning in cancer therapy. Exploitation of the chemical diversity of natural alkaloids may enrich the candidate pool of compounds for cancer chemotherapy and -prevention. Their further preclinical and clinical development should follow the same stringent rules as for any other synthetic drug as well. Prospective randomized, placebo-controlled clinical phase I and II trials should be initiated to unravel the full potential of plant alkaloids for drug repositioning.
Collapse
Affiliation(s)
- Thomas Efferth
- Department of Pharmaceutical Biology, Johannes Gutenberg University, Mainz, Germany.
| | - Franz Oesch
- Institute of Toxicology, Medical Center, Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
25
|
Tan DC, Roth IM, Wickremesekera AC, Davis PF, Kaye AH, Mantamadiotis T, Stylli SS, Tan ST. Therapeutic Targeting of Cancer Stem Cells in Human Glioblastoma by Manipulating the Renin-Angiotensin System. Cells 2019; 8:cells8111364. [PMID: 31683669 PMCID: PMC6912312 DOI: 10.3390/cells8111364] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 10/23/2019] [Accepted: 10/29/2019] [Indexed: 12/11/2022] Open
Abstract
Patients with glioblastoma (GB), a highly aggressive brain tumor, have a median survival of 14.6 months following neurosurgical resection and adjuvant chemoradiotherapy. Quiescent GB cancer stem cells (CSCs) invariably cause local recurrence. These GB CSCs can be identified by embryonic stem cell markers, express components of the renin-angiotensin system (RAS) and are associated with circulating CSCs. Despite the presence of circulating CSCs, GB patients rarely develop distant metastasis outside the central nervous system. This paper reviews the current literature on GB growth inhibition in relation to CSCs, circulating CSCs, the RAS and the novel therapeutic approach by repurposing drugs that target the RAS to improve overall symptom-free survival and maintain quality of life.
Collapse
Affiliation(s)
- David Ch Tan
- Department of Neurosurgery, Wellington Regional Hospital, Wellington 6021, New Zealand.
| | - Imogen M Roth
- Gillies McIndoe Research Institute, Wellington 6021, New Zealand.
| | - Agadha C Wickremesekera
- Department of Neurosurgery, Wellington Regional Hospital, Wellington 6021, New Zealand.
- Gillies McIndoe Research Institute, Wellington 6021, New Zealand.
- Department of Surgery, The University of Melbourne, Parkville, Victoria 3050, Australia.
| | - Paul F Davis
- Gillies McIndoe Research Institute, Wellington 6021, New Zealand.
| | - Andrew H Kaye
- Department of Surgery, The University of Melbourne, Parkville, Victoria 3050, Australia.
- Department of Neurosurgery, Hadassah Hebrew University Medical Centre, Jerusalem 91120, Israel.
| | - Theo Mantamadiotis
- Department of Surgery, The University of Melbourne, Parkville, Victoria 3050, Australia.
| | - Stanley S Stylli
- Department of Surgery, The University of Melbourne, Parkville, Victoria 3050, Australia.
- Department of Neurosurgery, The Royal Melbourne Hospital, Parkville, Victoria 3050, Australia.
| | - Swee T Tan
- Gillies McIndoe Research Institute, Wellington 6021, New Zealand.
- Department of Surgery, The University of Melbourne, Parkville, Victoria 3050, Australia.
- Wellington Regional Plastic, Maxillofacial & Burns Unit, Hutt Hospital, Lower Hutt 5040, New Zealand.
| |
Collapse
|
26
|
Caffeine enhances the anti-tumor effect of 5-fluorouracil via increasing the production of reactive oxygen species in hepatocellular carcinoma. Med Oncol 2019; 36:97. [DOI: 10.1007/s12032-019-1323-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 10/15/2019] [Indexed: 01/19/2023]
|
27
|
Pérez-Pérez D, Reyes-Vidal I, Chávez-Cortez EG, Sotelo J, Magaña-Maldonado R. Methylxanthines: Potential Therapeutic Agents for Glioblastoma. Pharmaceuticals (Basel) 2019; 12:ph12030130. [PMID: 31500285 PMCID: PMC6789489 DOI: 10.3390/ph12030130] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 08/24/2019] [Accepted: 09/01/2019] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma (GBM) is the most common and aggressive primary brain tumor. Currently, treatment is ineffective and the median overall survival is 20.9 months. The poor prognosis of GBM is a consequence of several altered signaling pathways that favor the proliferation and survival of neoplastic cells. One of these pathways is the deregulation of phosphodiesterases (PDEs). These enzymes participate in the development of GBM and may have value as therapeutic targets to treat GBM. Methylxanthines (MXTs) such as caffeine, theophylline, and theobromine are PDE inhibitors and constitute a promising therapeutic anti-cancer agent against GBM. MTXs also regulate various cell processes such as proliferation, migration, cell death, and differentiation; these processes are related to cancer progression, making MXTs potential therapeutic agents in GBM.
Collapse
Affiliation(s)
- Daniel Pérez-Pérez
- PECEM, Faculty of Medicine, National Autonomous University of México, México City 04510, Mexico
- Neuroimmunology and Neuro-oncology Unit, National Institute of Neurology and Neurosurgery, México City 14269, Mexico
| | - Iannel Reyes-Vidal
- Neuroimmunology and Neuro-oncology Unit, National Institute of Neurology and Neurosurgery, México City 14269, Mexico
| | - Elda Georgina Chávez-Cortez
- Neuroimmunology and Neuro-oncology Unit, National Institute of Neurology and Neurosurgery, México City 14269, Mexico
| | - Julio Sotelo
- Neuroimmunology and Neuro-oncology Unit, National Institute of Neurology and Neurosurgery, México City 14269, Mexico
| | - Roxana Magaña-Maldonado
- Neuroimmunology and Neuro-oncology Unit, National Institute of Neurology and Neurosurgery, México City 14269, Mexico.
| |
Collapse
|
28
|
Sánchez-Martín V, Jiménez-García L, Herranz S, Luque A, Acebo P, Amesty Á, Estévez-Braun A, de Las Heras B, Hortelano S. α-Hispanolol Induces Apoptosis and Suppresses Migration and Invasion of Glioblastoma Cells Likely via Downregulation of MMP-2/9 Expression and p38MAPK Attenuation. Front Pharmacol 2019; 10:935. [PMID: 31551765 PMCID: PMC6733979 DOI: 10.3389/fphar.2019.00935] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 07/22/2019] [Indexed: 12/14/2022] Open
Abstract
α-Hispanolol (α-H) is a labdane diterpenoid that has been shown to induce apoptosis in several human cancer cells. However, the effect of α-H in human glioblastoma cells has not been described. In the present work, we have investigated the effects of α-H on apoptosis, migration, and invasion of human glioblastoma cells with the aim of identifying the molecular targets underlying its mechanism of action. The results revealed that α-H showed significant cytotoxicity against human glioma cancer cell lines U87 and U373 in a concentration- and time-dependent manner. This effect was higher in U87 cells and linked to apoptosis, as revealed the increased percentage of sub-G1 population by cell cycle analysis and acquisition of typical features of apoptotic cell morphology. Apoptosis was also confirmed by significant presence of annexin V-positive cells and caspase activation. Pretreatment with caspase inhibitors diminishes the activities of caspase 8, 9, and 3 and maintains the percentage of viable glioblastoma cells, indicating that α-H induced cell apoptosis through both the extrinsic and the intrinsic pathways. Moreover, we also found that α-H downregulated the anti-apoptotic Bcl-2 and Bcl-xL proteins and activated the pro-apoptotic Bid and Bax proteins. On the other hand, α-H exhibited inhibitory effects on the migration and invasion of U87 cells in a concentration-dependent manner. Furthermore, additional experiments showed that α-H treatment reduced the enzymatic activities and protein levels of matrix metalloproteinase MMP-2 and MMP-9 and increased the expression of TIMP-1 inhibitor, probably via p38MAPK regulation. Finally, xenograft assays confirmed the anti-glioma efficacy of α-H. Taken together, these findings suggest that α-H may exert anti-tumoral effects in vitro and in vivo through the inhibition of cell proliferation and invasion as well as by the induction of apoptosis in human glioblastoma cells. This research describes α-H as a new drug that may improve the therapeutic efficacy against glioblastoma tumors.
Collapse
Affiliation(s)
- Vanesa Sánchez-Martín
- Unidad de Terapias Farmacológicas, Área de Genética Humana, Instituto de Investigación de Enfermedades Raras (IIER), Instituto de Salud Carlos III, Madrid, Spain.,Departamento de Farmacología, Farmacognosia y Botánica, Facultad de Farmacia, Universidad Complutense de Madrid (UCM), Madrid, Spain
| | - Lidia Jiménez-García
- Unidad de Terapias Farmacológicas, Área de Genética Humana, Instituto de Investigación de Enfermedades Raras (IIER), Instituto de Salud Carlos III, Madrid, Spain
| | - Sandra Herranz
- Unidad de Terapias Farmacológicas, Área de Genética Humana, Instituto de Investigación de Enfermedades Raras (IIER), Instituto de Salud Carlos III, Madrid, Spain
| | - Alfonso Luque
- Unidad de Terapias Farmacológicas, Área de Genética Humana, Instituto de Investigación de Enfermedades Raras (IIER), Instituto de Salud Carlos III, Madrid, Spain
| | - Paloma Acebo
- Unidad de Terapias Farmacológicas, Área de Genética Humana, Instituto de Investigación de Enfermedades Raras (IIER), Instituto de Salud Carlos III, Madrid, Spain
| | - Ángel Amesty
- Departamento de Química Orgánica, Instituto Universitario de Bio-Orgánica Antonio González, Universidad de La Laguna, La Laguna, Tenerife, Spain
| | - Ana Estévez-Braun
- Departamento de Química Orgánica, Instituto Universitario de Bio-Orgánica Antonio González, Universidad de La Laguna, La Laguna, Tenerife, Spain
| | - Beatriz de Las Heras
- Departamento de Farmacología, Farmacognosia y Botánica, Facultad de Farmacia, Universidad Complutense de Madrid (UCM), Madrid, Spain
| | - Sonsoles Hortelano
- Unidad de Terapias Farmacológicas, Área de Genética Humana, Instituto de Investigación de Enfermedades Raras (IIER), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
29
|
Mijanović O, Branković A, Panin AN, Savchuk S, Timashev P, Ulasov I, Lesniak MS. Cathepsin B: A sellsword of cancer progression. Cancer Lett 2019; 449:207-214. [PMID: 30796968 PMCID: PMC6488514 DOI: 10.1016/j.canlet.2019.02.035] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 02/12/2019] [Accepted: 02/13/2019] [Indexed: 12/13/2022]
Abstract
Clinical, biochemical and molecular biology studies have identified lysosome-encapsulated cellular proteases as critical risk factors for cancer progression. Cathepsins represent a group of such proteases aimed at maintenance of cellular homeostasis. Nevertheless, recent reports suggest that Cathepsin B executes other cellular programs such as controlling tumor growth, migration, invasion, angiogenesis, and metastases development. In fact, elevated levels of Cathepsins are found under different pathological conditions including inflammation, infection, neurodegenerative disease, and cancer. Furthermore, the discovery of Cathepsin B secretion and function as an extracellular matrix protein has broadened our appreciation for the impact of Cathepsin B on cancer progression. Underneath a façade of an intracellular protease with limited therapeutic potential hides a central role of cathepsins in extracellular functions. Moreover, this role is incredibly diverse from one condition to the next - from driving caspase-dependent apoptosis to facilitating tumor neovascularization and metastasis. Here we discuss the role of Cathepsin B in the oncogenic process and perspective the use of Cathepsin B for diagnostic and therapeutic applications.
Collapse
Affiliation(s)
- Olja Mijanović
- Institute of Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow, 119991, Russia
| | - Ana Branković
- Department of Forensics, Academy of Criminalistic and Police Studies, Belgrade, Serbia
| | - Alexander N Panin
- Moscow State University of Food Production, 11, Volokolamskoe Shosse, Moscow, 125080, Russia
| | - Solomiia Savchuk
- The University of Illinois at Chicago (UIC), Chicago, IL, USA; Northwestren University, Chicago, IL, 60611, USA
| | - Peter Timashev
- Institute of Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow, 119991, Russia
| | - Ilya Ulasov
- Institute of Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow, 119991, Russia
| | - Maciej S Lesniak
- The University of Illinois at Chicago (UIC), Chicago, IL, USA; Northwestren University, Chicago, IL, 60611, USA.
| |
Collapse
|
30
|
Yen GC, Tsai CM, Lu CC, Weng CJ. Recent progress in natural dietary non-phenolic bioactives on cancers metastasis. J Food Drug Anal 2018; 26:940-964. [PMID: 29976413 PMCID: PMC9303016 DOI: 10.1016/j.jfda.2018.05.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 05/04/2018] [Accepted: 05/15/2018] [Indexed: 12/20/2022] Open
Abstract
From several decades ago to now, cancer continues to be the leading cause of death worldwide, and metastasis is the major cause of cancer-related deaths. For health benefits, there is a great desire to use non-chemical therapy such as nutraceutical supplementation to prevent pathology development. Over 10,000 different natural bioactives or phytochemicals have been known that possessing potential preventive or supplementary effects for various diseases including cancer. Previously, the in vitro and in vivo anti-invasive and anti-metastatic activities of phenolic acids, monophenol, polyphenol and their derivatives and flavonoids and their derivatives have been reviewed. However, a vast number of natural dietary compounds other than phenolics have been demonstrated to potentially possess the ability to inhibit the invasion and metastasis of various cancers. In this review, we summarize the studies in recent decade on in vitro and in vivo effects and molecular mechanisms of natural bioactives, excluding the phenolics in food, in cancer invasion and metastasis. By combining this review of non-phenolics with the previous phenolics reviews, the puzzle for the contribution of natural dietary bioactives on cancer invasive or/and metastatic progress will be almost complete and more clear.
Collapse
Affiliation(s)
- Gow-Chin Yen
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung, Taiwan; Graduate Institute of Food Safety, National Chung Hsing University, Taichung, Taiwan
| | - Chiung-Man Tsai
- Tainan Hospital, Ministry of Health and Welfare, Tainan City, Taiwan
| | - Chi-Cheng Lu
- Department of Pharmacy, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Chia-Jui Weng
- Department of Living Services Industry, Tainan University of Technology, Tainan City, Taiwan.
| |
Collapse
|
31
|
Liang FR, He HS, Zhang CW, Xu XM, Zeng ZP, Yuan JP, Hong YH, Wang JH. Molecular cloning and functional characterization of cathepsin B from Nile tilapia (Oreochromis niloticus). Int J Biol Macromol 2018; 116:71-83. [PMID: 29730007 DOI: 10.1016/j.ijbiomac.2018.04.160] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 04/26/2018] [Accepted: 04/29/2018] [Indexed: 02/08/2023]
Abstract
Cathepsin B (CatB) has been widely known for its hydrolytic ability and involvement in the innate immunity. However, the mechanism of CatB from teleosts participating in immunoregulation remains poorly understood; and the sequence of CatB from Nile tilapia (NtCatB) has not been cloned and characterized. In this study, the coding sequence of NtCatB was cloned, and then characterized by bioinformatic analysis and heterologous expression. The deduced amino acid sequence (330-aa) of NtCatB contains the representative features of CatB. Quantitative real-time PCR revealed the extensive mRNA expression of NtCatB in six tissues of healthy Nile tilapia, and its transcription level was significantly up-regulated after Streptococcus agalactiae challenge. NtCatB may interact with some immunological function proteins and take part in the regulatory pathway. These results suggest that NtCatB is likely to be involved in the immune reaction. The mature region (residues 79-328, mNtCatB) of NtCatB was cloned and transferred to pET-28a for expressing the recombinant protein. The purified recombinant mNtCatB was verified with the activity of 992.34 U mg-1 min-1 under the optimal condition using a substrate hydrolyzing assay. The recombinant cystatin-A1-like can effectively inhibit the activity of the recombinant mNtCatB, and their binding form was predicted by molecular docking. Our results contribute to elucidating the immunological functions of NtCatB.
Collapse
Affiliation(s)
- Fu-Rui Liang
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, School of Marine Sciences, Sun Yat-Sen University, Guangzhou 510006, People's Republic of China.
| | - Hui-Shi He
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, School of Marine Sciences, Sun Yat-Sen University, Guangzhou 510006, People's Republic of China.
| | - Chu-Wen Zhang
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, School of Marine Sciences, Sun Yat-Sen University, Guangzhou 510006, People's Republic of China.
| | - Xiao-Ming Xu
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, School of Marine Sciences, Sun Yat-Sen University, Guangzhou 510006, People's Republic of China.
| | - Zhao-Ping Zeng
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, School of Marine Sciences, Sun Yat-Sen University, Guangzhou 510006, People's Republic of China.
| | - Jian-Ping Yuan
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, School of Marine Sciences, Sun Yat-Sen University, Guangzhou 510006, People's Republic of China; South China Sea Bioresource Exploitation and Utilization Collaborative Innovation Center, School of Marine Sciences, Sun Yat-Sen University, Guangzhou 510006, People's Republic of China.
| | - Yue-Hui Hong
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, School of Marine Sciences, Sun Yat-Sen University, Guangzhou 510006, People's Republic of China.
| | - Jiang-Hai Wang
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, School of Marine Sciences, Sun Yat-Sen University, Guangzhou 510006, People's Republic of China; South China Sea Bioresource Exploitation and Utilization Collaborative Innovation Center, School of Marine Sciences, Sun Yat-Sen University, Guangzhou 510006, People's Republic of China.
| |
Collapse
|
32
|
Yang SL, Kuo FH, Chen PN, Hsieh YH, Yu NY, Yang WE, Hsieh MJ, Yang SF. Andrographolide suppresses the migratory ability of human glioblastoma multiforme cells by targeting ERK1/2-mediated matrix metalloproteinase-2 expression. Oncotarget 2017; 8:105860-105872. [PMID: 29285298 PMCID: PMC5739685 DOI: 10.18632/oncotarget.22407] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 09/23/2017] [Indexed: 11/25/2022] Open
Abstract
Glioblastoma multiforme (GBM) can be a fatal tumor because of difficulties in treating the related metastasis. Andrographolide is the bioactive component of the Andrographis paniculata. Andrographolide possesses the anti-inflammatory activity and inhibits the growth of various cancers; however, its effect on GBM cancer motility remains largely unknown. In this study, we examined the antimetastatic properties of andrographolide in human GBM cells. Our results revealed that andrographolide inhibited the invasion and migration abilities of GBM8401 and U251 cells. Furthermore, andrographolide inhibited matrix metalloproteinase (MMP)-2 activity and expression. Real-time PCR and promoter activity assays indicated that andrographolide inhibited MMP-2 expression at the transcriptional level. Such inhibitory effects were associated with the suppression of CREB DNA-binding activity and CREB expression. Mechanistically, andrographolide inhibited the cell motility of GBM8401 cells through the extracellular-regulated kinase (ERK) 1/2 pathway, and the blocking of the ERK 1/2 pathway could reverse MMP-2-mediated cell motility. In conclusion, CREB is a crucial target of andrographolide for suppressing MMP-2-mediated cell motility in GBM cells. Therefore, a combination of andrographolide and an ERK inhibitor might be a good strategy for preventing GBM metastasis.
Collapse
Affiliation(s)
- Shih-Liang Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Traditional Chinese Medicine, Taichung Hospital, Ministry of Health and Welfare, Taichung, Taiwan
| | - Fu-Hsuan Kuo
- Neurological Institute, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Pei-Ni Chen
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan.,Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Yi-Hsien Hsieh
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan.,Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Nuo-Yi Yu
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Wei-En Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Ming-Ju Hsieh
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Cancer Research Center, Changhua Christian Hospital, Changhua, Taiwan.,Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
33
|
Liu H, Zhou Y, Tang L. Caffeine induces sustained apoptosis of human gastric cancer cells by activating the caspase‑9/caspase‑3 signalling pathway. Mol Med Rep 2017; 16:2445-2454. [PMID: 28677810 PMCID: PMC5547974 DOI: 10.3892/mmr.2017.6894] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 04/25/2017] [Indexed: 12/21/2022] Open
Abstract
Caffeine is one of the most widely consumed substances found in beverages, and has demonstrated anticancer effects in several types of cancer. The present study aimed to examine the anticancer effects of caffeine on gastric cancer (GC) cells (MGC‑803 and SGC‑7901) in vitro, and to determine whether the apoptosis‑related caspase‑9/-3 pathway is associated with these effects. The sustained antiproliferative effects of caffeine on gastric cancer were also investigated. GC cell viability and proliferation were evaluated using cell counting and colony forming assays, following treatment with various concentrations of caffeine. Flow cytometry was performed to assess cell cycle dynamics and apoptosis. Western blot analysis was conducted to detect the activity of the caspase‑9/-3 pathway. The results indicated that caffeine treatment significantly suppressed GC cell growth and viability and induced apoptosis by activating the caspase‑9/-3 pathway. Furthermore, the anticancer effects of caffeine appeared to be sustained, as the caspase‑9/-3 pathway remained active following caffeine withdrawal. In conclusion, caffeine may function as a sustained anticancer agent by activating the caspase‑9/-3 pathway, which indicates that it may be useful as a therapeutic candidate in gastric cancer.
Collapse
Affiliation(s)
- Hanyang Liu
- Department of Gastrointestinal Surgery, Nanjing Medical University, Affiliated Changzhou No. 2 People's Hospital, Changzhou, Jiangsu 213000, P.R. China
| | - Yan Zhou
- Department of Gastrointestinal Surgery, Nanjing Medical University, Affiliated Changzhou No. 2 People's Hospital, Changzhou, Jiangsu 213000, P.R. China
| | - Liming Tang
- Department of Gastrointestinal Surgery, Nanjing Medical University, Affiliated Changzhou No. 2 People's Hospital, Changzhou, Jiangsu 213000, P.R. China
| |
Collapse
|
34
|
Cadoná FC, Rosa JL, Schneider T, Cubillos-Rojas M, Sánchez-Tena S, Azzolin VF, Assmann CE, Machado AK, Ribeiro EE, da Cruz IBM. Guaraná, a Highly Caffeinated Food, Presentsin vitroAntitumor Activity in Colorectal and Breast Cancer Cell Lines by Inhibiting AKT/mTOR/S6K and MAPKs Pathways. Nutr Cancer 2017; 69:800-810. [DOI: 10.1080/01635581.2017.1324994] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Francine C. Cadoná
- Graduate Program in Biochemistry and Toxicology, Federal University of Santa Maria, Santa Maria, Brazil
| | - Jose L. Rosa
- Departament de Ciències Fisiològiques II, Campus de Bellvitge, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Taiane Schneider
- Departament de Ciències Fisiològiques II, Campus de Bellvitge, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Monica Cubillos-Rojas
- Departament de Ciències Fisiològiques II, Campus de Bellvitge, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Susana Sánchez-Tena
- Departament de Ciències Fisiològiques II, Campus de Bellvitge, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Verônica F. Azzolin
- Graduate Program in Pharmacology, Federal University of Santa Maria, Santa Maria, Brazil
| | - Charles E. Assmann
- Graduate Program in Biochemistry and Toxicology, Federal University of Santa Maria, Santa Maria, Brazil
| | | | - Euler E. Ribeiro
- Third Age Open University, University of Amazonas State, Manaus, Brazil
| | - Ivana Beatrice M. da Cruz
- Graduate Program in Biochemistry and Toxicology, Federal University of Santa Maria, Santa Maria, Brazil
- Graduate Program in Pharmacology, Federal University of Santa Maria, Santa Maria, Brazil
- Graduate Program in Gerontology, Federal University of Santa Maria, Santa Maria, Brazil
| |
Collapse
|
35
|
Pounis G, Tabolacci C, Costanzo S, Cordella M, Bonaccio M, Rago L, D'Arcangelo D, Filippo Di Castelnuovo A, de Gaetano G, Donati MB, Iacoviello L, Facchiano F. Reduction by coffee consumption of prostate cancer risk: Evidence from the Moli-sani cohort and cellular models. Int J Cancer 2017; 141:72-82. [PMID: 28436066 DOI: 10.1002/ijc.30720] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 03/01/2017] [Accepted: 03/20/2017] [Indexed: 01/14/2023]
Abstract
Meta-analytic data on the effect of coffee in prostate cancer risk are controversial. Caffeine as a bioactive compound of coffee has not yet been studied in deep in vitro. Our study aimed at evaluating in a population cohort the effect of Italian-style coffee consumption on prostate cancer risk and at investigating in vitro the potential antiproliferative and antimetastatic activity of caffeine on prostate cancer cell lines. 6,989 men of the Moli-sani cohort aged ≥50 years were followed for a mean of 4.24 ± 1.35 years and 100 new prostate cancer cases were identified. The European Prospective Investigation into Cancer and Nutrition-Food Frequency Questionnaire was used for the dietary assessment and the evaluation of Italian-style coffee consumption. Two human prostate cancer cell lines, PC-3 and DU145, were tested with increasing concentrations of caffeine, and their proliferative/metastatic features were evaluated. The newly diagnosed prostate cancer participants presented lower coffee consumption (60.1 ± 51.3 g/day) compared to the disease-free population (74.0 ± 51.7 g/day) (p < 0.05). Multiadjusted analysis showed that the subjects at highest consumption (>3 cups/day) had 53% lower prostate cancer risk as compared to participants at the lowest consumption (0-2 cups/day) (p = 0.02). Both human prostate cancer cell lines treated with caffeine showed a significant reduction in their proliferative and metastatic behaviors (p < 0.05). In conclusion, reduction by Italian-style coffee consumption of prostate cancer risk (>3 cups/day) was observed in epidemiological level. Caffeine appeared to exert both antiproliferative and antimetastatic activity on two prostate cancer cell lines, thus providing a cellular confirmation for the cohort study results.
Collapse
Affiliation(s)
- George Pounis
- Department of Epidemiology and Prevention, IRCCS Istituto Neurologico Mediterraneo - NEUROMED, Pozzilli, (IS), Italy
| | - Claudio Tabolacci
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, ISS, Rome, Italy
| | - Simona Costanzo
- Department of Epidemiology and Prevention, IRCCS Istituto Neurologico Mediterraneo - NEUROMED, Pozzilli, (IS), Italy
| | - Martina Cordella
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, ISS, Rome, Italy
| | - Marialaura Bonaccio
- Department of Epidemiology and Prevention, IRCCS Istituto Neurologico Mediterraneo - NEUROMED, Pozzilli, (IS), Italy
| | - Livia Rago
- Epicomed Research Srl. Campobasso, Italy
| | - Daniela D'Arcangelo
- Laboratory of Vascular Pathology. Istituto Dermopatico dell'Immacolata-IRCCS, FLMM, Rome, Italy
| | | | - Giovanni de Gaetano
- Department of Epidemiology and Prevention, IRCCS Istituto Neurologico Mediterraneo - NEUROMED, Pozzilli, (IS), Italy
| | - Maria Benedetta Donati
- Department of Epidemiology and Prevention, IRCCS Istituto Neurologico Mediterraneo - NEUROMED, Pozzilli, (IS), Italy
| | - Licia Iacoviello
- Department of Epidemiology and Prevention, IRCCS Istituto Neurologico Mediterraneo - NEUROMED, Pozzilli, (IS), Italy.,Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Francesco Facchiano
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, ISS, Rome, Italy
| | -
- The Moli-sani study investigators are listed in the appendix
| |
Collapse
|
36
|
Sun J, Luo Q, Liu L, Yang X, Zhu S, Song G. Salinomycin attenuates liver cancer stem cell motility by enhancing cell stiffness and increasing F-actin formation via the FAK-ERK1/2 signalling pathway. Toxicology 2017; 384:1-10. [PMID: 28395993 DOI: 10.1016/j.tox.2017.04.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Revised: 04/05/2017] [Accepted: 04/06/2017] [Indexed: 02/07/2023]
Abstract
Salinomycin has recently been identified as an antitumour drug for several types of cancer stem cell (CSC) treatments. However, the effects of salinomycin on the migratory and invasive properties of liver cancer stem cells (LCSCs) are unclear. In present study, we investigated the effect of salinomycin on the migration and invasion of LCSCs, and examined the molecular mechanisms underlying the anticancer effects of salinomycin. Here we showed that the migration and invasion of LCSCs were significantly suppressed in a salinomycin dose-dependent manner. Moreover, western blot analysis showed that salinomycin repressed the phosphorylation of focal adhesion kinase (FAK) and extracellular signal-regulated kinase (ERK1/2). Taken together, these findings provide new evidence that salinomycin suppresses the migration and invasion of LCSCs by inhibiting the expression of the FAK-ERK1/2 signalling pathway. In addition, the analysis of the mechanical properties showed that salinomycin increased cell stiffness in LCSCs via the FAK, and ERK1/2 pathways, suggesting that the inhibition of LCSC migration might partially contribute to the increase in cell stiffness stimulated by salinomycin. To further examine the role of salinomycin on cell motility and stiffness, the actin cytoskeleton of LCSCs was detected. The increased F-actin filaments in LCSCs induced by salinomycin reflected the increase in cell stiffness and the decrease in cell migration. Overall, these results showed that salinomycin inhibits the migration and invasion of LCSCs through the dephosphorylated FAK and ERK1/2 pathways, reflecting the changes in cell stiffness resulting from the increased actin cytoskeleton.
Collapse
Affiliation(s)
- Jinghui Sun
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, People's Republic of China; School of Medical Laboratory Science, Chengdu Medical College, Chengdu 610500, People's Republic of China
| | - Qing Luo
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, People's Republic of China
| | - Lingling Liu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, People's Republic of China
| | - Xianjiong Yang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, People's Republic of China; Department of Chemistry, School of Basic Medical Sciences, Guizhou Medical University, Guiyang 500025, People's Republic of China
| | - Shunqin Zhu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, People's Republic of China; School of Life Sciences, Southwest University, Chongqing 400715, People's Republic of China
| | - Guanbin Song
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, People's Republic of China.
| |
Collapse
|
37
|
Wu H, Li J, Xu D, Jv D, Meng X, Qiao P, Cui T, Shi B. The 37-kDa laminin receptor precursor regulates the malignancy of human glioma cells. Cell Biochem Funct 2017; 34:516-521. [PMID: 27748570 DOI: 10.1002/cbf.3225] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 09/08/2016] [Accepted: 09/12/2016] [Indexed: 11/12/2022]
Abstract
Glioma is one of the most common brain tumors and one of the most aggressive cancers. Although extensive progress has been made regarding to the diagnosis and treatment, the mortality in glioma patients is still high. Therefore, finding new therapeutic targets to the glioma is critical to the advancement in cancer treatment. Recently, the 37-kDa laminin receptor precursor (37LRP) was reported to play important roles in occurrence of some types of cancer, indicating that this molecule may function as a key regulator in the tumor migration and metastasis. However, there is still no report to elucidate the correlation between 37LRP expression and glioma genesis and development. In this study, we found the higher expression of 37LRP in the glioma cells compared with the normal brain cells. We also indicated that the downregulation of 37LRP could affect the glioma biomarker expression and also weaken the proliferative, migratory, and metastatic capacity of glioma cells in vitro. Furthermore, 37LRP silencing inhibited the glioma tumor growth in vivo. Collectively, these data demonstrated that 37LRP regulates the metastasis of glioma cells in vitro and tumor growth in vivo, suggesting that 37LRP may function as a potential molecular target in the glioma treatment.
Collapse
Affiliation(s)
- Hongjie Wu
- Department of Neurosurgery, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China, 471003.
| | - Jing Li
- Department of Neurosurgery, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China, 471003
| | - Dongxiao Xu
- Department of Neurosurgery, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China, 471003
| | - Donghui Jv
- Department of Neurosurgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China, 150086
| | - Xiaofeng Meng
- Department of Neurosurgery, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China, 471003
| | - Peng Qiao
- Department of Neurosurgery, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China, 471003
| | - Tao Cui
- Department of Neurosurgery, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China, 471003
| | - Baozhong Shi
- Department of Neurosurgery, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China, 471003
| |
Collapse
|
38
|
Selected Literature Watch. JOURNAL OF CAFFEINE RESEARCH 2016. [DOI: 10.1089/jcr.2016.29002.slw] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|