1
|
Park JS, Ma YQ, Wang F, Ma H, Sui G, Rustamov N, Han M, Son Y, Park CW, Han SB, Hong JT, Jeong LS, Lee J, Roh YS. A3AR antagonism mitigates metabolic dysfunction-associated steatotic liver disease by exploiting monocyte-derived Kupffer cell necroptosis and inflammation resolution. Metabolism 2025; 164:156114. [PMID: 39732364 DOI: 10.1016/j.metabol.2024.156114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/17/2024] [Accepted: 12/20/2024] [Indexed: 12/30/2024]
Abstract
BACKGROUND & AIMS Metabolic dysfunction-associated steatotic liver (MASLD) progression is driven by chronic inflammation and fibrosis, largely influenced by Kupffer cell (KC) dynamics, particularly replenishment of pro-inflammatory monocyte-derived KCs (MoKCs) due to increased death of embryo-derived KCs. Adenosine A3 receptor (A3AR) plays a key role in regulating metabolism and immune responses, making it a promising therapeutic target. This study aimed to investigate the impact of selective A3AR antagonism for regulation of replenished MoKCs, thereby improving MASLD. APPROACH & RESULTS A3AR expression was significantly elevated in KCs from both patients with MASLD and fast-food diet (FFD)-fed mice. A3AR knockout (KO) mice displayed marked improvements in hepatic inflammation and fibrosis along with a reduction in CLEC4F-positive KCs. The spatial transcriptomics of these KCs revealed disrupted mitochondrial integrity, increased oxidative stress, and enhanced cell death due to A3AR deletion. Similarly, in vivo FM101 treatment, a highly potent and selective antagonist of A3AR with a truncated 4'-thioadenosine structure, mitigated FFD-induced MASLD in mice. Mechanistically, FM101 induces β-arrestin2-mediated A3AR degradation, leading to mitochondrial dysfunction-mediated necroptosis in KCs. Consistently, A3AR was highly expressed in monocyte-derived macrophages in MASLD patients, with strong correlations with macrophage activation and monocyte chemoattractant gene sets. Thus, FM101 induced necroptosis in pro-inflammatory MoKCs, facilitating anti-inflammatory effects. CONCLUSIONS This study demonstrated that inhibiting A3AR via FM101 or genetic deletion alleviates MASLD by inducing mitochondrial dysfunction and subsequent necroptosis in MoKCs, establishing FM101 as a promising therapeutic strategy for MASLD.
Collapse
Affiliation(s)
- Jeong-Su Park
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, South Korea
| | - Yuan-Qiang Ma
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, South Korea
| | - Feng Wang
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, South Korea
| | - Hwan Ma
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, South Korea
| | - Guoyan Sui
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, South Korea
| | - Nodir Rustamov
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, South Korea
| | - Minyeong Han
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, South Korea
| | - Yejin Son
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, South Korea
| | - Chun-Woong Park
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, South Korea
| | - Sang-Bae Han
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, South Korea
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, South Korea
| | - Lak Shin Jeong
- Research and Development Center, Future Medicine Co., Ltd, Seongnam, South Korea; College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Jin Lee
- Department of Pathology, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA.
| | - Yoon Seok Roh
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, South Korea.
| |
Collapse
|
2
|
Tosh DK, Pavan M, Clark AA, Lammers J, Villano S, Marri S, Sgambellone S, Choi S, Lee J, Ivancich MS, Bock HA, Campbell RG, Lewicki SA, Levitan IM, Chen E, Liu N, Demby T, Gavrilova O, Gao ZG, Lucarini L, McCorvy JD, Jacobson KA. Potent and Selective Human 5-HT 2B Serotonin Receptor Antagonists: 4'-Cyano-(N)-methanocarba-adenosines by Synthetic Serendipity. J Med Chem 2024; 67:21264-21291. [PMID: 39589936 PMCID: PMC11715225 DOI: 10.1021/acs.jmedchem.4c02174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2024]
Abstract
Rigidified nucleoside derivatives with (N)-methanocarba replacement of ribose have been repurposed as peripheral subtype-selective 5-HT2B serotonin receptor antagonists for heart and lung fibrosis and intestinal/vascular conditions. 4'-Cyano derivative 40 (MRS8209; Ki, 4.27 nM) was 47-fold (human binding, but not rat and mouse) and 724-fold (functionally) selective at 5-HT2BR, compared to antitarget 5-HT2CR, and predicted to form a stable receptor complex using docking and molecular dynamics. 4'-Cyano substituents enhanced 5-HT2BR affinity (typically 4-5-fold compared to 4'-CH2OH), depending on an N6 group larger than methyl. Asymmetric N6 groups (4'-cyano-2-halo derivatives 33-35 and 37) provided potent 5-HT2BR Ki values (7-22 nM). A 4'-CH2CN substituent was less effective than 4'-CN at increasing 5-HT2BR affinity, while a 4'-CHF2 group produced high 5-HT2B affinity/selectivity. A 2-benzylthio-adenine group with unsubstituted 6-NH2 shifted the typical selectivity pattern toward potent 5-HT2C binding. Thus, the SAR suggests that N6-cyclopentyl-4'-cyano modifications are promising, with an interdependence among the substituent positions.
Collapse
Affiliation(s)
- Dilip K Tosh
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Matteo Pavan
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Allison A Clark
- Department of Cell Biology, Neurobiology, and Anatomy, Neuroscience Research Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, Wisconsin 53226, United States
| | - Josie Lammers
- Department of Cell Biology, Neurobiology, and Anatomy, Neuroscience Research Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, Wisconsin 53226, United States
| | - Serafina Villano
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology, University of Florence, Viale Gaetano Pieraccini, 6, Florence 50139, Italy
| | - Silvia Marri
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology, University of Florence, Viale Gaetano Pieraccini, 6, Florence 50139, Italy
| | - Silvia Sgambellone
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology, University of Florence, Viale Gaetano Pieraccini, 6, Florence 50139, Italy
| | - Suebin Choi
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Jihyun Lee
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Marko S Ivancich
- Department of Cell Biology, Neurobiology, and Anatomy, Neuroscience Research Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, Wisconsin 53226, United States
| | - Hailey A Bock
- Department of Cell Biology, Neurobiology, and Anatomy, Neuroscience Research Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, Wisconsin 53226, United States
| | - Ryan G Campbell
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Sarah A Lewicki
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Ian M Levitan
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Eric Chen
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Naili Liu
- Mouse Metabolism Core, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Tamar Demby
- Mouse Metabolism Core, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Oksana Gavrilova
- Mouse Metabolism Core, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Zhan-Guo Gao
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Laura Lucarini
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology, University of Florence, Viale Gaetano Pieraccini, 6, Florence 50139, Italy
| | - John D McCorvy
- Department of Cell Biology, Neurobiology, and Anatomy, Neuroscience Research Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, Wisconsin 53226, United States
| | - Kenneth A Jacobson
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institutes of Health, Bethesda, Maryland 20892, United States
| |
Collapse
|
3
|
Kim M, Naik SD, Jarhad DB, Aswar VR, Tripathi SK, Aslam MA, Huh JY, Jeong LS. Stereochemical influence of 4'-methyl substitutions on truncated 4'-thioadenosine derivatives: Impact on A 3 adenosine receptor binding and antagonism. Bioorg Chem 2024; 153:107901. [PMID: 39447347 DOI: 10.1016/j.bioorg.2024.107901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 10/10/2024] [Accepted: 10/17/2024] [Indexed: 10/26/2024]
Abstract
Herein, we investigated the stereochemical effects of 4'-methyl substitution on A3 adenosine receptor (A3AR) ligands by synthesizing and evaluating a series of truncated 4'-thioadenosine derivatives featuring 4'-α-methyl, 4'-β-methyl, and 4',4'-dimethyl substitutions. We successfully synthesized these derivatives, using the stereoselective addition of an organometallic reagent, KSAc-mediated sulfur cyclization, and Vorbrüggen condensation. Binding assays demonstrated that the 4'-β-methyl substitution conferred the highest affinity for A3AR, with compound 1 h exhibiting a Ki = 3.5 nM, followed by the 4',4'-dimethyl and 4'-α-methyl substitutions. Notably, despite the absence of the 5'-OH group, compound 1 h unexpectedly displayed partial agonism. Computational docking studies indicated that compound 1 h, the β-methyl derivative, adopted a South conformation and maintained strong interactions within the receptor, including a critical interaction with Thr94, a residue known to be notable for agonistic effects. Conversely, compound 2 h, the α-methyl derivative, also adopted a South conformation but resulted in a flattened structure that hindered interactions with Thr94 and Asn250. The dimethyl derivative 3 h exhibited steric clashes with Thr94, contributing to a reduction in binding affinity. However, the docking results for 3 h indicated a North conformation, suggesting that the change in sugar conformation due to the additional 4'-methyl group altered the angle between the α-methyl group and the sugar plane, enabling binding despite the increased steric bulk. These findings suggest that not only do the substituents and their stereochemistry influence receptor-ligand interactions, but the conformation and the resulting spatial orientation of the substituents also play a crucial role in modulating receptor-ligand interaction. This stereochemical insight offers a valuable framework for the design of new, selective, and potent A3AR ligands, potentially facilitating the development of novel therapeutics for A3AR-related diseases such as glaucoma, inflammation, and cancer.
Collapse
Affiliation(s)
- Minjae Kim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, South Korea
| | - Siddhi D Naik
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, South Korea; Government College of Arts Science and Commerce, Khandola Marcela, Goa, India
| | - Dnyandev B Jarhad
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, South Korea
| | - Vikas R Aswar
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, South Korea
| | - Sushil Kumar Tripathi
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, South Korea
| | - Muhammad Arif Aslam
- College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Joo Young Huh
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea; Department of Global Innovative Drugs, The Graduate School of Chung-Ang University, Seoul 06974, Republic of Korea
| | - Lak Shin Jeong
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, South Korea; Future Medicine Co., Ltd, 54 Changup-ro, Sujeong-gu, Seongnam, Gyeonggi-do 13449, Republic of Korea.
| |
Collapse
|
4
|
Shan L, Gao L, Chai Y, Li K, Yu J, Liang F, Qin J, Ni Y, Sun P. Cordycepin improves hyperactivation and acrosome reaction through adenosine receptors during human sperm capacitation in vitro. Reprod Biol Endocrinol 2024; 22:143. [PMID: 39533327 PMCID: PMC11555834 DOI: 10.1186/s12958-024-01318-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Sperm capacitation is a prerequisite for natural or in vitro fertilization. After capacitation, sperm become hyperactivated and undergo an acrosome reaction, which helps them penetrate the oocyte. Cordycepin, a bioactive compound first isolated from Cordyceps militaris, is an adenosine analog with numerous physiological activities. However, its effects on sperm capacitation remain unclear. This study aims to elucidate the effects and mechanisms of cordycepin on human sperm capacitation. METHODS During in vitro capacitation culture, healthy human sperm were treated with cordycepin (20, 100, 500 µM). Sperm motility and hyperactivation were detected using a computer-assisted sperm analyzer. Sperm acrosome reaction was measured using fluorescein isothiocyanate-conjugated Pisum sativum agglutinin. Sperm protein kinase A (PKA) activity was analyzed using an ELISA kit. The levels of sperm protein tyrosine phosphorylation were detected by western blotting. Sperm DNA damage was detected by a sperm chromatin dispersion assay. Reactive oxygen species (ROS) were measured using the fluorescence probe 2',7'-dichlorodihydrofluorescein diacetate. The expression and localization of adenosine receptors were analyzed by western blotting and immunofluorescence. The specific inhibitors of adenosine receptors were used to confirm their effects on cordycepin-induced sperm capacitation. Finally, molecular docking was performed to analyze the interaction between cordycepin and adenosine receptors. RESULTS Cordycepin improved hyperactivated sperm motility, acrosome reaction, PKA activity, and protein tyrosine phosphorylation during capacitation while having no obvious effects on sperm ROS or DNA damage. Four adenosine receptor subtypes were expressed in human sperm, but their localizations differed. Inhibition of adenosine receptors significantly decreased cordycepin-induced sperm hyperactivation and the acrosome reaction. Molecular docking showed that cordycepin can bind to the four subtypes of adenosine receptors. CONCLUSION Cordycepin may promote human sperm capacitation through adenosine receptor-mediated signaling pathways. These findings may be useful for assisted reproductive technology and animal breeding.
Collapse
Affiliation(s)
- Lijun Shan
- School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang, China
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Linmei Gao
- School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang, China
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yuhao Chai
- School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang, China
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Kun Li
- School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang, China
- Zhejiang Provincial Laboratory of Experimental Animal's & Nonclinical Laboratory Studies, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Jianmin Yu
- School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Fei Liang
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Jiangfeng Qin
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Ya Ni
- School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang, China
- Zhejiang Provincial Laboratory of Experimental Animal's & Nonclinical Laboratory Studies, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Peibei Sun
- School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang, China.
- Zhejiang Provincial Laboratory of Experimental Animal's & Nonclinical Laboratory Studies, Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
5
|
Yan A, Rosa AL, Chhablani PP, Chhablani J. Caffeine and Vision: Effects on the Eye. Turk J Ophthalmol 2024; 54:291-300. [PMID: 39463170 PMCID: PMC11589232 DOI: 10.4274/tjo.galenos.2024.43895] [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: 04/03/2024] [Accepted: 05/29/2024] [Indexed: 10/29/2024] Open
Abstract
Caffeine, commonly found in coffee and tea, affects various aspects of eye health as it blocks adenosine receptors, impacting tear production, intraocular pressure, macular perfusion, and choroidal thickness. However, its connection with eye conditions like glaucoma and cataracts remains uncertain due to conflicting research findings. Some studies suggest potential benefits for cataracts, while others warn against frequent caffeine intake in glaucoma and surgical scenarios due to possible increases in intraocular pressure. Conflicting evidence also exists regarding its effects on dry eye, macular degeneration, myopia/hyperopia, diabetic retinopathy, retinopathy of prematurity, and central serous retinopathy. Caffeine does not seem to be a risk factor for dry eye, although studies have shown that caffeine may offer protection against wet age-related macular degeneration, and the metabolite 7-methylxanthine could be a more promising treatment for myopia. Moreover, caffeine can potentially cause tremors and might hinder surgical performance, especially in less experienced surgeons. Recommendations from experts vary, highlighting the need for further research to fully understand how caffeine affects the eye. Individuals genetically predisposed to glaucoma should be cautious due to the possibility of clinically significant elevations in intraocular pressure with caffeine consumption. For delicate procedures like microsurgery, where tremors can be detrimental, caution should be exercised with caffeine. This review underscores the importance of additional studies to provide clearer insights and prudent recommendations regarding caffeine's impact on eye health.
Collapse
Affiliation(s)
- Audrey Yan
- West Virginia School of Osteopathic Medicine, Department of Medicine, Lewisburg, USA
| | - Antonio La Rosa
- IRCCS Humanitas Research Hospital, Department of Ophthalmology, Milan, Italy
| | - Preeti Patil Chhablani
- University of Pittsburgh School of Medicine, Department of Ophthalmology, Pittsburg, USA
| | - Jay Chhablani
- University of Pittsburgh School of Medicine, Department of Ophthalmology, Pittsburg, USA
| |
Collapse
|
6
|
Ming Y, Gong Y, Fu X, Ouyang X, Peng Y, Pu W. Small-molecule-based targeted therapy in liver cancer. Mol Ther 2024; 32:3260-3287. [PMID: 39113358 PMCID: PMC11489561 DOI: 10.1016/j.ymthe.2024.08.001] [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: 11/22/2023] [Revised: 03/13/2024] [Accepted: 08/02/2024] [Indexed: 08/23/2024] Open
Abstract
Liver cancer is one of the most prevalent malignant tumors worldwide. According to the Barcelona Clinic Liver Cancer staging criteria, clinical guidelines provide tutorials to clinical management of liver cancer at their individual stages. However, most patients diagnosed with liver cancer are at advanced stage; therefore, many researchers conduct investigations on targeted therapy, aiming to improve the overall survival of these patients. To date, small-molecule-based targeted therapies are highly recommended (first line: sorafenib and lenvatinib; second line: regorafenib and cabozantinib) by current the clinical guidelines of the American Society of Clinical Oncology, European Society for Medical Oncology, and National Comprehensive Cancer Network. Herein, we summarize the small-molecule-based targeted therapies in liver cancer, including the approved and preclinical therapies as well as the therapies under clinical trials, and introduce their history of discovery, clinical trials, indications, and molecular mechanisms. For drug resistance, the revealed mechanisms of action and the combination therapies are also discussed. In fact, the known small-molecule-based therapies still have limited clinical benefits to liver cancer patients. Therefore, we analyze the current status and give our ideas for the urgent issues and future directions in this field, suggesting clues for novel techniques in liver cancer treatment.
Collapse
Affiliation(s)
- Yue Ming
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Yanqiu Gong
- National Clinical Research Center for Geriatrics and Department of General Practice, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xuewen Fu
- Jinhua Huanke Environmental Technology Co., Ltd., Jinhua 321000, China
| | - Xinyu Ouyang
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610064, China; West China School of Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yong Peng
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610064, China; Frontier Medical Center, Tianfu Jincheng Laboratory, Chengdu 610212, China.
| | - Wenchen Pu
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610064, China; West China School of Medicine, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
7
|
Kim M, Choi H, Nayak A, Tripathi SK, Aswar VR, Gaikwad VB, Jacobson KA, Jeong LS. Structure-Activity Relationship of Truncated 4'-Selenonucleosides: A 3 Adenosine Receptor Activity and Binding Selectivity. ACS Med Chem Lett 2024; 15:1620-1626. [PMID: 39420956 PMCID: PMC11482266 DOI: 10.1021/acsmedchemlett.4c00344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 08/18/2024] [Accepted: 08/19/2024] [Indexed: 10/19/2024] Open
Abstract
This study explored the impact of structural modifications on truncated 4'-selenonucleosides as ligands for the A3 adenosine receptor (AR). We synthesized and evaluated a series of these compounds for their binding affinities, functional activities, and structural interactions by using computational modeling. The SAR study revealed that all compounds exhibited selective and notable hA3AR binding, among which 6l (K i = 5.2 nM) and 6m (K i = 5.7 nM) were found as the best binding compounds. The representative N 6-cyclopropyl compound 6m was found to be a partial agonist, contrasting with the antagonist profiles of truncated 4'-oxo and 4'-thionucleosides. Computational docking highlighted 6m's unique interaction with Thr94 at the A3AR binding site. This research not only advances our understanding of A3AR ligand interactions but also highlights the potential of truncated 4'-selenonucleosides as effective A3AR modulators.
Collapse
Affiliation(s)
- Minjae Kim
- Research
Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Korea
| | - Hongseok Choi
- Research
Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Korea
| | - Akshata Nayak
- Bangalore
University, Gnanabharati Campus, Bangalore 560056, India
| | - Sushil K. Tripathi
- Research
Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Korea
| | - Vikas R. Aswar
- Research
Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Korea
| | - Vidyasagar B. Gaikwad
- Research
Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Korea
| | - Kenneth A. Jacobson
- Molecular
Recognition Section, Laboratory of Bioorganic Chemistry, National
Institute of Diabetes and Digestive and Kidney Disease, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Lak Shin Jeong
- Research
Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Korea
- Future
Medicine Co., Ltd, 54
Changup-ro, Sujeong-gu, Seongnam, Gyeonggi-do 13449, Republic of Korea
| |
Collapse
|
8
|
Valladão SC, França AP, Pandolfo P, Dos Santos-Rodrigues A. Adenosinergic system and nucleoside transporters in attention deficit hyperactivity disorder: Current findings. Neurosci Biobehav Rev 2024; 164:105771. [PMID: 38880409 DOI: 10.1016/j.neubiorev.2024.105771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 06/07/2024] [Accepted: 06/11/2024] [Indexed: 06/18/2024]
Abstract
Attention deficit hyperactivity disorder (ADHD) is a neurodevelopmental disorder with high heterogeneity that can affect individuals of any age. It is characterized by three main symptoms: inattention, hyperactivity, and impulsivity. These neurobehavioral alterations and neurochemical and pharmacological findings are mainly attributed to unbalanced catecholaminergic signaling, especially involving dopaminergic pathways within prefrontal and striatal areas. Dopamine receptors and transporters are not solely implicated in this imbalance, as evidence indicates that the dopaminergic signaling is modulated by adenosine activity. To this extent, alterations in adenosinergic signaling are probably involved in ADHD. Here, we review the current knowledge about adenosine's role in the modulation of chemical, behavioral and cognitive parameters of ADHD, especially regarding dopaminergic signaling. Current literature usually links adenosine receptors signaling to the dopaminergic imbalance found in ADHD, but there is evidence that equilibrative nucleoside transporters (ENTs) could also be implicated as players in dopaminergic signaling alterations seen in ADHD, since their involvement in other neurobehavioral impairments.
Collapse
Affiliation(s)
- Sofia Corrêa Valladão
- Graduate Program of Neurosciences and Department of Neurobiology, Institute of Biology, Universidade Federal Fluminense, Niterói, Brazil; Graduate Program of Physiology and Pharmacology, Biomedical Institute, Universidade Federal Fluminense, Niterói, Brazil.
| | - Angela Patricia França
- Graduate Program in Neuroscience, Centre of Biological Sciences, Federal University of Santa Catarina (UFSC), Brazil; Graduate Program in Medical Sciences, Centre of Health Sciences, Federal University of Santa Catarina, Brazil.
| | - Pablo Pandolfo
- Graduate Program of Neurosciences and Department of Neurobiology, Institute of Biology, Universidade Federal Fluminense, Niterói, Brazil; Graduate Program of Physiology and Pharmacology, Biomedical Institute, Universidade Federal Fluminense, Niterói, Brazil.
| | - Alexandre Dos Santos-Rodrigues
- Graduate Program of Neurosciences and Department of Neurobiology, Institute of Biology, Universidade Federal Fluminense, Niterói, Brazil.
| |
Collapse
|
9
|
Li J, Wang D, Hao X, Li Y, Gao H, Fan Y, Fang B, Guo Y. Exploring the high-quality ingredients and mechanisms of Da Chuanxiong Formula in the treatment of neuropathic pain based on network pharmacology, molecular docking, and molecular dynamics simulation. Biomed Pharmacother 2024; 178:117195. [PMID: 39068852 DOI: 10.1016/j.biopha.2024.117195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 07/10/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024] Open
Abstract
Da Chuanxiong Formula (DCXF) is a traditional herbal prescription used for pain management. It consists of Chuanxiong Rhizoma (CR) and Gastrodiae Rhizoma (GR). Despite its long history of use, the underlying therapeutic mechanism of DCXF remains insufficiently understood. Therefore, in this study, key target genes were obtained through network pharmacology research methods and molecular docking techniques, including transient receptor potential vanilloid 1 (TRPV1), adenosine A2a receptor (ADORA2A), nuclear receptor subfamily 3 group C member 1 (NR3C1), and protein kinase C beta (PRKCB). Molecular dynamics simulations demonstrated the favorable binding between all four key genes and their corresponding compounds. Notably, chronic constriction injury (CCI) treatment resulted in a significant decrease in mechanical threshold and thermal latency period for rat foot contraction, which was ameliorated upon administration of DCXF. Furthermore, real-time quantitative reverse transcription PCR (RT-qPCR) and western blot (WB) analyses indicated an upregulation of TRPV1, ADORA2A, NR3C1, and PRKCB expression in the rat dorsal root ganglion following CCI, which was attenuated by treatment with DCXF. The expressions of inflammatory factors, including tumor necrosis factor-α (TNF-α), interleukin 1 beta (IL-1β), and interleukin 6 (IL-6), in the rat dorsal root ganglion were assessed using ELISA, confirming consistent trends with the aforementioned findings. The results of this study offer a promising theoretical foundation for the utilization of DCXF in the treatment of neuropathic pain (NP).
Collapse
Affiliation(s)
- Jinshi Li
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, China
| | - Dongxu Wang
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, China
| | - Xiaotong Hao
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, China
| | - Yuan Li
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, China
| | - Hairong Gao
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, China
| | - Yiting Fan
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, China
| | - Bo Fang
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, China.
| | - Yang Guo
- Department of Surgical Oncology, Breast Surgery, General Surgery, The First Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
10
|
Fathi M, Zarei A, Moghimi A, Jalali P, Salehi Z, Gholamin S, Jadidi-Niaragh F. Combined cancer immunotherapy based on targeting adenosine pathway and PD-1/PDL-1 axis. Expert Opin Ther Targets 2024; 28:757-777. [PMID: 39305018 DOI: 10.1080/14728222.2024.2405090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 09/12/2024] [Indexed: 10/02/2024]
Abstract
INTRODUCTION Cancer immunotherapy has revolutionized the field of oncology, offering new hope to patients with advanced malignancies. Tumor-induced immunosuppression limits the effectiveness of current immunotherapeutic strategies, such as PD-1/PDL-1 checkpoint inhibitors. Adenosine, a purine nucleoside molecule, is crucial to this immunosuppression because it stops T cells from activating and helps regulatory T cells grow. Targeting the adenosine pathway and blocking PD-1/PDL-1 is a potential way to boost the immune system's response to tumors. AREAS COVERED This review discusses the current understanding of the adenosine pathway in tumor immunology and the preclinical and clinical data supporting the combination of adenosine pathway inhibitors with PD-1/PDL-1 blockade. We also discuss the challenges and future directions for developing combination immunotherapy targeting the adenosine pathway and the PD-1/PDL-1 axis for cancer treatment. EXPERT OPINION The fact that the adenosine signaling pathway controls many immune system processes suggests that it has a wide range of therapeutic uses. Within the next five years, there will be tremendous progress in this area, and the standard of care for treating malignant tumors will have switched from point-to-point therapy to the integration of immunological networks comprised of multiple signaling pathways, like the adenosine axis.
Collapse
Affiliation(s)
- Mehrdad Fathi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Asieh Zarei
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ata Moghimi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pooya Jalali
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Centre, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Salehi
- Hematology, Oncology and Stem Cell Transplantation Research Center, Research Institute for Oncology, Hematology and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran
| | - Sharareh Gholamin
- City of Hope Beckman Research Institute and Medical Center, Duarte, CA, USA
- City of Hope Department of Radiation Oncology, Duarte, CA, USA
| | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
11
|
Huang X, Chorianopoulou A, Kalkounou P, Georgiou M, Pousias A, Davies A, Pearce A, Harris M, Lambrinidis G, Marakos P, Pouli N, Kolocouris A, Lougiakis N, Ladds G. Hit-to-Lead Optimization of Heterocyclic Carbonyloxycarboximidamides as Selective Antagonists at Human Adenosine A3 Receptor. J Med Chem 2024; 67:13117-13146. [PMID: 39073853 PMCID: PMC11320584 DOI: 10.1021/acs.jmedchem.4c01092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/18/2024] [Accepted: 07/01/2024] [Indexed: 07/30/2024]
Abstract
Antagonism of the human adenosine A3 receptor (hA3R) has potential therapeutic application. Alchemical relative binding free energy calculations of K18 and K32 suggested that the combination of a 3-(2,6-dichlorophenyl)-isoxazolyl group with 2-pyridinyl at the ends of a carbonyloxycarboximidamide group should improve hA3R affinity. Of the 25 new analogues synthesized, 37 and 74 showed improved hA3R affinity compared to K18 (and K32). This was further improved through the addition of a bromine group to the 2-pyridinyl at the 5-position, generating compound 39. Alchemical relative binding free energy calculations, mutagenesis studies and MD simulations supported the compounds' binding pattern while suggesting that the bromine of 39 inserts deep into the hA3R orthosteric pocket, so highlighting the importance of rigidification of the carbonyloxycarboximidamide moiety. MD simulations highlighted the importance of rigidification of the carbonyloxycarboximidamide, while suggesting that the bromine of 39 inserts deep into the hA3R orthosteric pocket, which was supported through mutagenesis studies 39 also selectively antagonized endogenously expressed hA3R in nonsmall cell lung carcinoma cells, while pharmacokinetic studies indicated low toxicity enabling in vivo evaluation. We therefore suggest that 39 has potential for further development as a high-affinity hA3R antagonist.
Collapse
Affiliation(s)
- Xianglin Huang
- Department
of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, U.K.
| | - Anna Chorianopoulou
- Laboratory
of Medicinal Chemistry, Section of Pharmaceutical Chemistry, Department
of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou 15771, Athens, Greece
| | - Panagoula Kalkounou
- Laboratory
of Medicinal Chemistry, Section of Pharmaceutical Chemistry, Department
of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou 15771, Athens, Greece
| | - Maria Georgiou
- Laboratory
of Medicinal Chemistry, Section of Pharmaceutical Chemistry, Department
of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou 15771, Athens, Greece
| | - Athanasios Pousias
- Laboratory
of Medicinal Chemistry, Section of Pharmaceutical Chemistry, Department
of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou 15771, Athens, Greece
| | - Amy Davies
- Department
of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, U.K.
| | - Abigail Pearce
- Department
of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, U.K.
| | - Matthew Harris
- Department
of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, U.K.
| | - George Lambrinidis
- Laboratory
of Medicinal Chemistry, Section of Pharmaceutical Chemistry, Department
of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou 15771, Athens, Greece
| | - Panagiotis Marakos
- Laboratory
of Medicinal Chemistry, Section of Pharmaceutical Chemistry, Department
of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou 15771, Athens, Greece
| | - Nicole Pouli
- Laboratory
of Medicinal Chemistry, Section of Pharmaceutical Chemistry, Department
of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou 15771, Athens, Greece
| | - Antonios Kolocouris
- Laboratory
of Medicinal Chemistry, Section of Pharmaceutical Chemistry, Department
of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou 15771, Athens, Greece
| | - Nikolaos Lougiakis
- Laboratory
of Medicinal Chemistry, Section of Pharmaceutical Chemistry, Department
of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou 15771, Athens, Greece
| | - Graham Ladds
- Department
of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, U.K.
| |
Collapse
|
12
|
Venugopala KN, Buccioni M. Current Understanding of the Role of Adenosine Receptors in Cancer. Molecules 2024; 29:3501. [PMID: 39124905 PMCID: PMC11313767 DOI: 10.3390/molecules29153501] [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: 06/08/2024] [Revised: 07/21/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
Abstract
Cancer, a complex array of diseases, involves the unbridled proliferation and dissemination of aberrant cells in the body, forming tumors that can infiltrate neighboring tissues and metastasize to distant sites. With over 200 types, each cancer has unique attributes, risks, and treatment avenues. Therapeutic options encompass surgery, chemotherapy, radiation therapy, hormone therapy, immunotherapy, targeted therapy, or a blend of these methods. Yet, these treatments face challenges like late-stage diagnoses, tumor diversity, severe side effects, drug resistance, targeted drug delivery hurdles, and cost barriers. Despite these hurdles, advancements in cancer research, encompassing biology, genetics, and treatment, have enhanced early detection methods, treatment options, and survival rates. Adenosine receptors (ARs), including A1, A2A, A2B, and A3 subtypes, exhibit diverse roles in cancer progression, sometimes promoting or inhibiting tumor growth depending on the receptor subtype, cancer type, and tumor microenvironment. Research on AR ligands has revealed promising anticancer effects in lab studies and animal models, hinting at their potential as cancer therapeutics. Understanding the intricate signaling pathways and interactions of adenosine receptors in cancer is pivotal for crafting targeted therapies that optimize benefits while mitigating drawbacks. This review delves into each adenosine receptor subtype's distinct roles and signaling pathways in cancer, shedding light on their potential as targets for improving cancer treatment outcomes.
Collapse
Affiliation(s)
- Katharigatta Narayanaswamy Venugopala
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia
- Department of Biotechnology and Food Science, Faculty of Applied Sciences, Durban University of Technology, Durban 4001, South Africa
| | - Michela Buccioni
- School of Pharmacy, Medicinal Chemistry Unit, ChIP, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy;
| |
Collapse
|
13
|
Akbar H, Jarosinski KW. Temporal Dynamics of Purinergic Receptor Expression in the Lungs of Marek's Disease (MD) Virus-Infected Chickens Resistant or Susceptible to MD. Viruses 2024; 16:1130. [PMID: 39066292 PMCID: PMC11281646 DOI: 10.3390/v16071130] [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: 06/08/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
Marek's disease virus (MDV) is an economic concern for the poultry industry due to its poorly understood pathophysiology. Purinergic receptors (PRs) are potential therapeutic targets for viral infections, including herpesviruses, prompting our investigation into their role in MDV pathogenesis. The current study is part of an experimental series analyzing the expression of PRs during MDV infection. To address the early or short-acting P2 PR responses during natural MDV infection, we performed an "exposure" experiment where age-matched chickens were exposed to experimentally infected shedders to initiate natural infection. In addition, select non-PR regulatory gene responses were measured. Two groups of naïve contact chickens (n = 5/breed/time point) from MD-resistant (White Leghorns: WL) and -susceptible (Pure Columbian) chicken lines were housed separately with experimentally infected PC (×PC) and WL (×WL) chickens for 6 or 24 h. Whole lung lavage cells (WLLC) were collected, RNA was extracted, and RT-qPCR assays were used to measure specific PR responses. In addition, other potentially important markers in pathophysiology were measured. Our study revealed that WL chickens exhibited higher P1 PR expression during natural infection. WL chickens also showed higher expression of P1A3 and P2X3 at 6 and 24 h when exposed to PC-infected chickens. P2X5 and P2Y1 showed higher expression at 6 h, while P2Y5 showed higher expression at 6 and 24 h; regardless of the chicken line, PC chickens exhibited higher expression of P2X2, P2Y8, P2Y10, P2Y13, and P2Y14 when exposed to either group of infected chickens. In addition, MDV infection altered the expression of DDX5 in both WL and PC groups exposed to PC-infected birds only. However, irrespective of the source of exposure, BCL2 and ANGPTL4 showed higher expression in both WL and PC. The expression of STAT1A and STAT5A was influenced by time and breed, with major changes observed in STAT5A. CAT and SOD1 expression significantly increased in both WL and PC birds, regardless of the source of infection. GPX1 and GPX2 expression also increased in both WL and PC, although overall lower expression was observed in PC chickens at 24 h compared to 6 h. Our data suggest systemic changes in the host during early infection, indicated by the altered expression of PRs, DDX5, BCL2, ANGPTL4, and other regulatory genes during early MDV infection. The relative expression of these responses in PC and WL chickens suggests they may play a key role in their response to natural MDV infection in the lungs and long-term pathogenesis and survival.
Collapse
Affiliation(s)
| | - Keith W. Jarosinski
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61802, USA;
| |
Collapse
|
14
|
Sequeira L, Benfeito S, Fernandes C, Lima I, Peixoto J, Alves C, Machado CS, Gaspar A, Borges F, Chavarria D. Drug Development for Alzheimer's and Parkinson's Disease: Where Do We Go Now? Pharmaceutics 2024; 16:708. [PMID: 38931832 PMCID: PMC11206728 DOI: 10.3390/pharmaceutics16060708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 05/15/2024] [Accepted: 05/22/2024] [Indexed: 06/28/2024] Open
Abstract
Neurodegenerative diseases (NDs) are a set of progressive, chronic, and incurable diseases characterized by the gradual loss of neurons, culminating in the decline of cognitive and/or motor functions. Alzheimer's disease (AD) and Parkinson's disease (PD) are the most common NDs and represent an enormous burden both in terms of human suffering and economic cost. The available therapies for AD and PD only provide symptomatic and palliative relief for a limited period and are unable to modify the diseases' progression. Over the last decades, research efforts have been focused on developing new pharmacological treatments for these NDs. However, to date, no breakthrough treatment has been discovered. Hence, the development of disease-modifying drugs able to halt or reverse the progression of NDs remains an unmet clinical need. This review summarizes the major hallmarks of AD and PD and the drugs available for pharmacological treatment. It also sheds light on potential directions that can be pursued to develop new, disease-modifying drugs to treat AD and PD, describing as representative examples some advances in the development of drug candidates targeting oxidative stress and adenosine A2A receptors.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Fernanda Borges
- CIQUP-IMS—Centro de Investigação em Química da Universidade do Porto, Institute of Molecular Sciences, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, R. Campo Alegre s/n, 4169-007 Porto, Portugal
| | - Daniel Chavarria
- CIQUP-IMS—Centro de Investigação em Química da Universidade do Porto, Institute of Molecular Sciences, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, R. Campo Alegre s/n, 4169-007 Porto, Portugal
| |
Collapse
|
15
|
Arora C, Matic M, Bisceglia L, Di Chiaro P, De Oliveira Rosa N, Carli F, Clubb L, Nemati Fard LA, Kargas G, Diaferia GR, Vukotic R, Licata L, Wu G, Natoli G, Gutkind JS, Raimondi F. The landscape of cancer-rewired GPCR signaling axes. CELL GENOMICS 2024; 4:100557. [PMID: 38723607 PMCID: PMC11099383 DOI: 10.1016/j.xgen.2024.100557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 02/17/2024] [Accepted: 04/10/2024] [Indexed: 05/15/2024]
Abstract
We explored the dysregulation of G-protein-coupled receptor (GPCR) ligand systems in cancer transcriptomics datasets to uncover new therapeutics opportunities in oncology. We derived an interaction network of receptors with ligands and their biosynthetic enzymes. Multiple GPCRs are differentially regulated together with their upstream partners across cancer subtypes and are associated to specific transcriptional programs and to patient survival patterns. The expression of both receptor-ligand (or enzymes) partners improved patient stratification, suggesting a synergistic role for the activation of GPCR networks in modulating cancer phenotypes. Remarkably, we identified many such axes across several cancer molecular subtypes, including many involving receptor-biosynthetic enzymes for neurotransmitters. We found that GPCRs from these actionable axes, including, e.g., muscarinic, adenosine, 5-hydroxytryptamine, and chemokine receptors, are the targets of multiple drugs displaying anti-growth effects in large-scale, cancer cell drug screens, which we further validated. We have made the results generated in this study freely available through a webapp (gpcrcanceraxes.bioinfolab.sns.it).
Collapse
Affiliation(s)
- Chakit Arora
- Laboratorio di Biologia Bio@SNS, Scuola Normale Superiore, Piazza dei Cavalieri 7, 56126 Pisa, Italy
| | - Marin Matic
- Laboratorio di Biologia Bio@SNS, Scuola Normale Superiore, Piazza dei Cavalieri 7, 56126 Pisa, Italy
| | - Luisa Bisceglia
- Laboratorio di Biologia Bio@SNS, Scuola Normale Superiore, Piazza dei Cavalieri 7, 56126 Pisa, Italy
| | - Pierluigi Di Chiaro
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milano, Italy
| | - Natalia De Oliveira Rosa
- Laboratorio di Biologia Bio@SNS, Scuola Normale Superiore, Piazza dei Cavalieri 7, 56126 Pisa, Italy
| | - Francesco Carli
- Laboratorio di Biologia Bio@SNS, Scuola Normale Superiore, Piazza dei Cavalieri 7, 56126 Pisa, Italy
| | - Lauren Clubb
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Lorenzo Amir Nemati Fard
- Laboratorio di Biologia Bio@SNS, Scuola Normale Superiore, Piazza dei Cavalieri 7, 56126 Pisa, Italy
| | - Giorgos Kargas
- Laboratorio di Biologia Bio@SNS, Scuola Normale Superiore, Piazza dei Cavalieri 7, 56126 Pisa, Italy
| | - Giuseppe R Diaferia
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milano, Italy
| | - Ranka Vukotic
- Azienda Ospedaliero-Universitaria Pisana, Via Roma, 67, 56126 Pisa, Italy
| | - Luana Licata
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Guanming Wu
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR, USA
| | - Gioacchino Natoli
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milano, Italy
| | - J Silvio Gutkind
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA.
| | - Francesco Raimondi
- Laboratorio di Biologia Bio@SNS, Scuola Normale Superiore, Piazza dei Cavalieri 7, 56126 Pisa, Italy; Laboratorio di Biologia Bio@SNS, Scuola Normale Superiore, Piazza dei Cavalieri 7, 56126 Pisa, Italy.
| |
Collapse
|
16
|
Sakao K, Sho C, Miyata T, Takara K, Oda R, Hou DX. Verification of In Vitro Anticancer Activity and Bioactive Compounds in Cordyceps Militaris-Infused Sweet Potato Shochu Spirits. Molecules 2024; 29:2119. [PMID: 38731610 PMCID: PMC11085083 DOI: 10.3390/molecules29092119] [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: 04/06/2024] [Revised: 04/24/2024] [Accepted: 05/01/2024] [Indexed: 05/13/2024] Open
Abstract
Many liqueurs, including spirits infused with botanicals, are crafted not only for their taste and flavor but also for potential medicinal benefits. However, the scientific evidence supporting their medicinal effects remains limited. This study aims to verify in vitro anticancer activity and bioactive compounds in shochu spirits infused with Cordyceps militaris, a Chinese medicine. The results revealed that a bioactive fraction was eluted from the spirit extract with 40% ethanol. The infusion time impacted the inhibitory effect of the spirit extract on the proliferation of colon cancer-derived cell line HCT-116 cells, and a 21-day infusion showed the strongest inhibitory effect. Furthermore, the spirit extract was separated into four fractions, A-D, by high-performance liquid chromatography (HPLC), and Fractions B, C, and D, but not A, exerted the effects of proliferation inhibition and apoptotic induction of HCT-116 cells and HL-60 cells. Furthermore, Fractions B, C, and D were, respectively, identified as adenosine, cordycepin, and N6-(2-hydroxyethyl)-adenosine (HEA) by comprehensive chemical analyses, including proton nuclear magnetic resonance (1H-NMR), Fourier transform infrared spectroscopy (FT-IR), and electrospray ionization mass spectrometry (ESI-MS). To better understand the bioactivity mechanisms of cordycepin and HEA, the agonist and antagonist tests of the A3 adenosine receptor (A3AR) were performed. Cell viability was suppressed by cordycepin, and HEA was restored by the A3AR antagonist MR1523, suggesting that cordycepin and HEA possibly acted as agonists to activate A3ARs to inhibit cell proliferation. Molecular docking simulations revealed that both adenosine and cordycepin bound to the same pocket site of A3ARs, while HEA exhibited a different binding pattern, supporting a possible explanation for the difference in their bioactivity. Taken together, the present study demonstrated that cordycepin and HEA were major bioactive ingredients in Cordyceps militaries-infused sweet potato shochu spirits, which contributed to the in vitro anticancer activity.
Collapse
Affiliation(s)
- Kozue Sakao
- The United Graduate School of Agricultural Sciences, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-0065, Japan; (K.S.); (T.M.); (K.T.)
- Graduate School of Agriculture, Forestry and Fisheries, Kagoshima University, Kagoshima 890-0065, Japan
| | - Cho Sho
- Kirishima Shuzo Co., Ltd., 4-28-1 Shimokawahigashi, Miyakonojo, Miyazaki 885-8588, Japan;
| | - Takeshi Miyata
- The United Graduate School of Agricultural Sciences, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-0065, Japan; (K.S.); (T.M.); (K.T.)
- Graduate School of Agriculture, Forestry and Fisheries, Kagoshima University, Kagoshima 890-0065, Japan
| | - Kensaku Takara
- The United Graduate School of Agricultural Sciences, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-0065, Japan; (K.S.); (T.M.); (K.T.)
- Faculty of Agriculture, University of the Ryukyus, 1 Senbaru, Nishihara, Okinawa 903-0213, Japan
| | - Rio Oda
- Graduate School of Agriculture, Forestry and Fisheries, Kagoshima University, Kagoshima 890-0065, Japan
| | - De-Xing Hou
- The United Graduate School of Agricultural Sciences, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-0065, Japan; (K.S.); (T.M.); (K.T.)
- Graduate School of Agriculture, Forestry and Fisheries, Kagoshima University, Kagoshima 890-0065, Japan
| |
Collapse
|
17
|
Cai H, Guo S, Xu Y, Sun J, Li J, Xia Z, Jiang Y, Xie X, Xu HE. Cryo-EM structures of adenosine receptor A 3AR bound to selective agonists. Nat Commun 2024; 15:3252. [PMID: 38627384 PMCID: PMC11021478 DOI: 10.1038/s41467-024-47207-6] [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: 10/17/2023] [Accepted: 03/22/2024] [Indexed: 04/19/2024] Open
Abstract
The adenosine A3 receptor (A3AR), a key member of the G protein-coupled receptor family, is a promising therapeutic target for inflammatory and cancerous conditions. The selective A3AR agonists, CF101 and CF102, are clinically significant, yet their recognition mechanisms remained elusive. Here we report the cryogenic electron microscopy structures of the full-length human A3AR bound to CF101 and CF102 with heterotrimeric Gi protein in complex at 3.3-3.2 Å resolution. These agonists reside in the orthosteric pocket, forming conserved interactions via their adenine moieties, while their 3-iodobenzyl groups exhibit distinct orientations. Functional assays reveal the critical role of extracellular loop 3 in A3AR's ligand selectivity and receptor activation. Key mutations, including His3.37, Ser5.42, and Ser6.52, in a unique sub-pocket of A3AR, significantly impact receptor activation. Comparative analysis with the inactive A2AAR structure highlights a conserved receptor activation mechanism. Our findings provide comprehensive insights into the molecular recognition and signaling of A3AR, paving the way for designing subtype-selective adenosine receptor ligands.
Collapse
Affiliation(s)
- Hongmin Cai
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
| | - Shimeng Guo
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Youwei Xu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Jun Sun
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Junrui Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Zhikan Xia
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yi Jiang
- Lingang Laboratory, Shanghai, China
| | - Xin Xie
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research, Institute for Drug Discovery, Yantai, China.
| | - H Eric Xu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
| |
Collapse
|
18
|
Choi Y, Jeong JY, Hong S. Highly Sensitive Real-Time Monitoring of Adenosine Receptor Activities in Nonsmall Cell Lung Cancer Cells Using Carbon Nanotube Field-Effect Transistors. ACS APPLIED MATERIALS & INTERFACES 2024; 16:2101-2109. [PMID: 38166368 DOI: 10.1021/acsami.3c14492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2024]
Abstract
Adenosine metabolism through adenosine receptors plays a critical role in lung cancer biology. Although recent studies showed the potential of targeting adenosine receptors as drug targets for lung cancer treatment, conventional methods for investigating receptor activities often suffer from various drawbacks, including low sensitivity and slow analysis speed. In this study, adenosine receptor activities in nonsmall cell lung cancer (NSCLC) cells were monitored in real time with high sensitivity through a carbon nanotube field-effect transistor (CNT-FET). In this method, we hybridized a CNT-FET with NSCLC cells expressing A2A and A2B adenosine receptors to construct a hybrid platform. This platform could detect adenosine, an endogenous ligand of adenosine receptors, down to 1 fM in real time and sensitively discriminate adenosine among other nucleosides. Furthermore, we could also utilize the platform to detect adenosine in complicated environments, such as human serum. Notably, our hybrid platform allowed us to monitor pharmacological effects between adenosine and other drugs, including dipyridamole and theophylline, even in human serum samples. These results indicate that the NSCLC cell-hybridized CNT-FET can be a practical tool for biomedical applications, such as the evaluation and screening of drug-candidate substances.
Collapse
Affiliation(s)
- Yoonji Choi
- Department of Physics and Astronomy, and Institute of Applied Physics, Seoul National University, Seoul 08826, Republic of Korea
| | - Jin-Young Jeong
- Post-Silicon Semiconductor Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Seunghun Hong
- Department of Physics and Astronomy, and Institute of Applied Physics, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
19
|
Mazziotta C, Badiale G, Cervellera CF, Tognon M, Martini F, Rotondo JC. Regulatory mechanisms of circular RNAs during human mesenchymal stem cell osteogenic differentiation. Theranostics 2024; 14:143-158. [PMID: 38164139 PMCID: PMC10750202 DOI: 10.7150/thno.89066] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 10/01/2023] [Indexed: 01/03/2024] Open
Abstract
Human osteogenic differentiation is a complex and well-orchestrated process which involves a plethora of molecular players and cellular processes. A growing number of studies have underlined that circular RNAs (circRNAs) play an important regulatory role during human osteogenic differentiation. CircRNAs are single-stranded, covalently closed non-coding RNA molecules that are acquiring increased attention as epigenetic regulators of gene expression. Given their intrinsic high conformational stability, abundance, and specificity, circRNAs can undertake various biological activities in order to regulate multiple cellular processes, including osteogenic differentiation. The most recent evidence indicates that circRNAs control human osteogenesis by preventing the inhibitory activity of miRNAs on their downstream target genes, using a competitive endogenous RNA mechanism. The aim of this review is to draw attention to the currently known regulatory mechanisms of circRNAs during human osteogenic differentiation. Specifically, we provide an understanding of recent advances in research conducted on various human mesenchymal stem cell types that underlined the importance of circRNAs in regulating osteogenesis. A comprehensive understanding of the underlying regulatory mechanisms of circRNA in osteogenesis will improve knowledge on the molecular processes of bone growth, resulting in the potential development of novel preclinical and clinical studies and the discovery of novel diagnostic and therapeutic tools for bone disorders.
Collapse
Affiliation(s)
- Chiara Mazziotta
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
- Center for Studies on Gender Medicine - Department of Medical Sciences, University of Ferrara. 64/b, Fossato di Mortara Street. Ferrara, Italy
| | - Giada Badiale
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | | | - Mauro Tognon
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Fernanda Martini
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
- Center for Studies on Gender Medicine - Department of Medical Sciences, University of Ferrara. 64/b, Fossato di Mortara Street. Ferrara, Italy
- Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy
| | - John Charles Rotondo
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
- Center for Studies on Gender Medicine - Department of Medical Sciences, University of Ferrara. 64/b, Fossato di Mortara Street. Ferrara, Italy
| |
Collapse
|
20
|
Mazziotta C, Iaquinta MR, Tramarin ML, Badiale G, Cervellera CF, Tonnini G, Patergnani S, Pinton P, Lanza G, Gafà R, Tognon M, Martini F, De Mattei M, Rotondo JC. Hsa-microRNA-1249-3p/Homeobox A13 axis modulates the expression of β-catenin gene in human epithelial cells. Sci Rep 2023; 13:22872. [PMID: 38129477 PMCID: PMC10739948 DOI: 10.1038/s41598-023-49837-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 12/12/2023] [Indexed: 12/23/2023] Open
Abstract
Intercellular adhesion is a key function for epithelial cells. The fundamental mechanisms relying on epithelial cell adhesion have been partially uncovered. Hsa-microRNA-1249-3p (hsa-miR-1249-3p) plays a role in the epithelial mesenchymal transition in carcinoma cells, but its physiological function in epithelial cells is unknown. We aimed to investigate the role and molecular mechanisms of hsa-miR-1249-3p on epithelial cell functions. Hsa-miR-1249-3p was overexpressed in human epithelial cells and uterine cervical tissues, compared to cervical carcinoma cells and precancerous tissues, respectively. Hsa-miR-1249-3p was analyzed to verify its regulatory function on Homeobox A13 (HOXA13) target gene and its downstream cell adhesion gene β-catenin. Functional experiments indicated that hsa-miR-1249-3p inhibition prompted the mRNA and protein overexpression of HOXA13 which, in turn, led to the β-catenin protein expression. Moreover, hsa-miR-1249-3p inhibition induced a strong colony forming ability in epithelial cells, suggesting the miR involvement in cell adhesion machinery. These data indicate that hsa-miR-1249-3p regulates the expression of HOXA13 and its downstream cell adhesion gene β-catenin, possible resulting in cell adhesion modification in epithelial cells. This study will allow the set-up of further investigations aimed at exploring the relationship between the hsa-miR-1249-3p/HOXA13 axis and downstream cell adhesion genes.
Collapse
Affiliation(s)
- Chiara Mazziotta
- Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121, Ferrara, Italy
- Center for Studies on Gender Medicine, Department of Medical Sciences, University of Ferrara, 44121, Ferrara, Italy
| | - Maria Rosa Iaquinta
- Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121, Ferrara, Italy
- Center for Studies on Gender Medicine, Department of Medical Sciences, University of Ferrara, 44121, Ferrara, Italy
| | - Maria Letizia Tramarin
- Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121, Ferrara, Italy
| | - Giada Badiale
- Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121, Ferrara, Italy
| | - Christian Felice Cervellera
- Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121, Ferrara, Italy
| | - Giulia Tonnini
- Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121, Ferrara, Italy
| | - Simone Patergnani
- Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121, Ferrara, Italy
- Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121, Ferrara, Italy
| | - Paolo Pinton
- Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121, Ferrara, Italy
- Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121, Ferrara, Italy
| | - Giovanni Lanza
- Department of Translational Medicine, University of Ferrara, 44121, Ferrara, Italy
| | - Roberta Gafà
- Department of Translational Medicine, University of Ferrara, 44121, Ferrara, Italy
| | - Mauro Tognon
- Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121, Ferrara, Italy
| | - Fernanda Martini
- Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121, Ferrara, Italy
- Center for Studies on Gender Medicine, Department of Medical Sciences, University of Ferrara, 44121, Ferrara, Italy
- Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121, Ferrara, Italy
| | - Monica De Mattei
- Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121, Ferrara, Italy.
| | - John Charles Rotondo
- Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121, Ferrara, Italy.
- Center for Studies on Gender Medicine, Department of Medical Sciences, University of Ferrara, 44121, Ferrara, Italy.
| |
Collapse
|
21
|
Youssef R, Maniar R, Khan J, Mesa H. Metabolic Interplay in the Tumor Microenvironment: Implications for Immune Function and Anticancer Response. Curr Issues Mol Biol 2023; 45:9753-9767. [PMID: 38132455 PMCID: PMC10742411 DOI: 10.3390/cimb45120609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/26/2023] [Accepted: 11/29/2023] [Indexed: 12/23/2023] Open
Abstract
Malignant tumors exhibit rapid growth and high metabolic rates, similar to embryonic stem cells, and depend on aerobic glycolysis, known as the "Warburg effect". This understanding has enabled the use of radiolabeled glucose analogs in tumor staging and therapeutic response assessment via PET scans. Traditional treatments like chemotherapy and radiotherapy target rapidly dividing cells, causing significant toxicity. Despite immunotherapy's impact on solid tumor treatment, gaps remain, leading to research on cancer cell evasion of immune response and immune tolerance induction via interactions with the tumor microenvironment (TME). The TME, consisting of immune cells, fibroblasts, vessels, and the extracellular matrix, regulates tumor progression and therapy responses. TME-targeted therapies aim to transform this environment from supporting tumor growth to impeding it and fostering an effective immune response. This review examines the metabolic disparities between immune cells and cancer cells, their impact on immune function and therapeutic targeting, the TME components, and the complex interplay between cancer cells and nontumoral cells. The success of TME-targeted therapies highlights their potential to achieve better cancer control or even a cure.
Collapse
Affiliation(s)
- Reem Youssef
- Department of Laboratory Medicine and Pathology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Rohan Maniar
- Division of Hematology/Oncology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Jaffar Khan
- Department of Laboratory Medicine and Pathology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Hector Mesa
- Department of Laboratory Medicine and Pathology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
22
|
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
|
23
|
Federico S, Persico M, Trevisan L, Biasinutto C, Bolcato G, Salmaso V, Da Ros T, Gianferrara T, Prencipe F, Kachler S, Klotz KN, Pacor S, Moro S, Spalluto G. [1,2,4]Triazolo[1,5-c]pyrimidines as Tools to Investigate A 3 Adenosine Receptors in Cancer Cell Lines. ChemMedChem 2023; 18:e202300299. [PMID: 37675643 DOI: 10.1002/cmdc.202300299] [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: 06/09/2023] [Revised: 08/31/2023] [Accepted: 09/06/2023] [Indexed: 09/07/2023]
Abstract
The A3 adenosine receptor is an interesting target whose role in cancer is controversial. In this work, a structural investigation at the 2-position of the [1,2,4]triazolo[1,5-c]pyrimidine nucleus was performed, finding new potent and selective A3 adenosine receptor antagonists such as the ethyl 2-(4-methoxyphenyl)-5-(methylamino)-[1,2,4]triazolo[1,5-c]pyrimidine-8-carboxylate (20, DZ123) that showed a Ki value of 0.47 nM and an exceptional selectivity profile over the other adenosine receptor subtypes. Computational studies were performed to rationalize the affinity and the selectivity profile of the tested compounds at the A3 adenosine receptor and the A1 and A2A adenosine receptors. Compound 20 was tested on both A3 adenosine receptor positive cell lines (CHO-A3 AR transfected, THP1 and HCT16) and on A3 negative cancer cell lines, showing no effect in the latter and a pro-proliferative effect at a low concentration in the former. These interesting results pave the way to further investigation on both the mechanism involved and potential therapeutic applications.
Collapse
Affiliation(s)
- Stephanie Federico
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Via Licio Giorgieri 1, 34127, Trieste, Italy
| | - Margherita Persico
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Via Licio Giorgieri 1, 34127, Trieste, Italy
| | - Letizia Trevisan
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Via Licio Giorgieri 1, 34127, Trieste, Italy
| | - Chiara Biasinutto
- Department of Life Sciences, University of Trieste, Via Licio Giorgieri 5, 34127, Trieste, Italy
| | - Giovanni Bolcato
- Molecular Modeling Section (MMS), Department of Pharmaceutical and Pharmacological Sciences, University of Padova, via Marzolo 5, 35131, Padova, Italy
| | - Veronica Salmaso
- Molecular Modeling Section (MMS), Department of Pharmaceutical and Pharmacological Sciences, University of Padova, via Marzolo 5, 35131, Padova, Italy
| | - Tatiana Da Ros
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Via Licio Giorgieri 1, 34127, Trieste, Italy
| | - Teresa Gianferrara
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Via Licio Giorgieri 1, 34127, Trieste, Italy
| | - Filippo Prencipe
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Via Licio Giorgieri 1, 34127, Trieste, Italy
| | - Sonja Kachler
- Rudolf-Virchow-Zentrum -, Center for Integrative and Translational Bioimaging, University of Würzburg, Josef-Schneider-Str. 2, 97080, Würzburg, Germany
| | - Karl-Norbert Klotz
- Institut für Pharmakologie und Toxikologie, University of Würzburg, Versbacher Str. 9, 97078, Würzburg, Germany
| | - Sabrina Pacor
- Department of Life Sciences, University of Trieste, Via Licio Giorgieri 5, 34127, Trieste, Italy
| | - Stefano Moro
- Molecular Modeling Section (MMS), Department of Pharmaceutical and Pharmacological Sciences, University of Padova, via Marzolo 5, 35131, Padova, Italy
| | - Giampiero Spalluto
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Via Licio Giorgieri 1, 34127, Trieste, Italy
| |
Collapse
|
24
|
Itzhak I, Bareket-Samish A, Fishman P. Namodenoson Inhibits the Growth of Pancreatic Carcinoma via Deregulation of the Wnt/β-catenin, NF-κB, and RAS Signaling Pathways. Biomolecules 2023; 13:1584. [PMID: 38002266 PMCID: PMC10669398 DOI: 10.3390/biom13111584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 10/17/2023] [Accepted: 10/23/2023] [Indexed: 11/26/2023] Open
Abstract
Namodenoson, an A3 adenosine receptor (A3AR) agonist, is currently being used in a phase III trial in advanced liver cancer. We examined the anti-growth effect of namodenoson on pancreatic carcinoma cells and investigated the molecular mechanism involved. BxPC-3 pancreatic carcinoma cells were cultured with namodenoson (5-20 nM for 24 h at 37 °C), and the Presto Blue assay was used to monitor cell growth. Western blot analyses were performed on BxPC-3 cells (20 nM namodenoson for 24 h at 37 °C) to evaluate the expression levels of cell growth regulatory proteins. In vivo studies involved the subcutaneous inoculation of BxPC-3 cells into nude mice, randomizing the mice into namodenoson (10 μg/kg twice daily for 35 days) vs. control, and monitoring tumor size twice weekly. Treatment with namodenoson was associated with the significant dose-dependent inhibition of BxPC-3 cell growth, which was mitigated by the A3AR antagonist MRS1523. Western blot analyses showed that namodenoson treatment modulated the expression of NF-κB, as well as proteins in the Wnt/β-catenin and the RAS signaling pathways, leading to the upregulation of apoptotic proteins (Bad, Bax). In vivo studies also showed the significant inhibition of pancreatic carcinoma tumor growth with namodenoson. In conclusion, our findings support the continued development of namodenoson as a treatment for pancreatic cancer.
Collapse
|
25
|
Wang Y, Zhang X, Tang G, Deng P, Qin Y, Han J, Wang S, Sun X, Li D, Chen Z. The causal relationship between gut microbiota and bone mineral density: a Mendelian randomization study. Front Microbiol 2023; 14:1268935. [PMID: 37937216 PMCID: PMC10625988 DOI: 10.3389/fmicb.2023.1268935] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 10/09/2023] [Indexed: 11/09/2023] Open
Abstract
Background The gut microbiota has emerged as an intriguing and potentially influential factor in regulating bone health. However, the causal effect of the gut microbiota on bone mineral density (BMD) appears to differ throughout various life stages. Methods We conducted a Mendelian randomization (MR) analysis to investigate the potential causal relationship between gut microbiota and BMD in five distinct age groups: 0-15, 15-30, 30-45, 45-60, and 60 years and older. The analysis employed three different methods, namely MR-Egger, weighted median, and Inverse-variance weighting, to ensure the robustness of our findings, a series of sensitivity analyses were also conducted, such as horizontal pleiotropy tests, heterogeneity tests, and leave-one-out sensitivity tests. Results In the age group of 0-15 years, Eubacterium_fissicatena_group and Eubacterium_hallii_group were identified as risk factors for BMD. During the 15-30 age group, Phascolarctobacterium, Roseburia, and Ruminococcaceae_UCG_003 were found to be protective factors for BMD. In the 30-45 age group, Lachnospira genus demonstrated a protective effect on BMD, while Barnesiella and Lactococcus were identified as risk factors for BMD. Moving on to the 45-60 age group, Eubacterium_ventriosum_group, Lachnospiraceae_UCG_004, and Subdoligranulum were observed to be protective factors for BMD, while Eubacterium_coprostanoligenes_group, Fusicatenibacter, and Lactococcus were associated with an increased risk of BMD. In individuals aged 60 years and older, Fusicatenibacter and Ruminococcaceae_UCG_002 were also noted as risk factors for BMD. Conversely, Eubacterium_ruminantium_group, Ruminococcus_gauvreauii_group, Alistipes, and Coprococcus_3 were found to be protective factors for BMD, whereas Barnesiella and Sellimonas were identified as risk factors for BMD. Conclusion A robust causal relationship between gut microbiota and bone mineral density (BMD) exists throughout all stages of life, with Firmicutes phylum being the primary group associated with BMD across age groups. Gut microbiota linked with BMD primarily belong to the Firmicutes phylum across age groups. The diversity of gut microbiota phyla associated with BMD depicts relatively stable patterns during the ages of 0-45 years. However, for individuals aged 45 years and above, there is an observed increase in the number of gut microbiota species linked with BMD, and by the age of 60 years, a trend toward an increase in the Bacteroidetes phylum categories is proposed.
Collapse
Affiliation(s)
- Ying Wang
- Beijing University of Chinese Medicine, Beijing, China
- Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Xuejian Zhang
- Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Guangjun Tang
- Beijing University of Chinese Medicine, Beijing, China
- Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Pin Deng
- Institute of Basic Theory of Traditional Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuyan Qin
- Beijing University of Chinese Medicine, Beijing, China
- Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Jinglu Han
- Beijing University of Chinese Medicine, Beijing, China
- Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Shulong Wang
- Beijing University of Chinese Medicine, Beijing, China
- Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Xiaojie Sun
- Beijing University of Chinese Medicine, Beijing, China
- Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Dongxiao Li
- Beijing University of Chinese Medicine, Beijing, China
- Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Zhaojun Chen
- Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| |
Collapse
|
26
|
Marco-Bonilla M, Herencia R, Fresnadillo M, Huete-Toral F, Carracedo G, Largo R, Herrero-Beaumont G, Mediero A. Dipyridamole activates adenosine A2B receptor and AMPK/cAMP signaling and promotes myogenic differentiation of myoblastic C2C12 cells. Front Pharmacol 2023; 14:1247664. [PMID: 37771723 PMCID: PMC10522837 DOI: 10.3389/fphar.2023.1247664] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 08/28/2023] [Indexed: 09/30/2023] Open
Abstract
Introduction: Sarcopenia is defined as a loss of muscle mass and strength. ATP homeostasis is crucial during myogenesis. We determined how the purinergic system modulates myogenesis using dipyridamole (blocks adenosine taken up by the cells) and tenofovir (inhibits ATP release) in a myoblast cell line. Methods: C2C12 cells were differentiated in the presence/absence of tenofovir/dipyridamole, with/without the A2B selective inhibitor PSB-603. Extra-/intracellular nucleotides were examined via HPLC. The expression of muscle differentiation proteins (Pax7, Mif5, MyoD, MyoG, and MHC), PKA/CREB, adenosine receptors (A1, A2A, A2B, and A3), ATP-channel pannexin-1 and the P2X7 receptor was analyzed via WB and RT-PCR. cAMP and AMPK activation was measured. Results: Tenofovir increased intracellular ATP and reduced extracellular adenosine, decreasing Pax7 expression and increasing MHC expression prematurely. Dipyridamole increased intracellular AMP and extracellular adenosine, counteracting the premature myogenesis promoted by tenofovir. All adenosine receptors were expressed during differentiation with dipyridamole, increasing A2B expression. Tenofovir maintained inactive AMPK and decreased cAMP levels, as well as PKAα and pCREB expression, which were recovered with dipyridamole. Discussion: Adenosine and ATP act as mediators in muscle myogenesis. The blockade of ATP release by tenofovir promotes premature myogenesis, with dipyridamole counteracting the premature differentiation promoted by tenofovir via the adenosine A2B receptor and cAMP/AMPK pathways. Therefore, dipyridamole might be of interest as a therapeutic approach in sarcopenia.
Collapse
Affiliation(s)
| | - Raquel Herencia
- Bone and Joint Research Unit, FIIS-Fundación Jiménez Díaz UAM, Madrid, Spain
| | - María Fresnadillo
- Bone and Joint Research Unit, FIIS-Fundación Jiménez Díaz UAM, Madrid, Spain
| | - Fernando Huete-Toral
- Ocupharm Group Research, Faculty of Optic and Optometry, University Complutense of Madrid, Madrid, Spain
| | - Gonzalo Carracedo
- Ocupharm Group Research, Faculty of Optic and Optometry, University Complutense of Madrid, Madrid, Spain
- Department of Optometry and Vision, Faculty of Optic and Optometry, University Complutense of Madrid, Madrid, Spain
| | - Raquel Largo
- Bone and Joint Research Unit, FIIS-Fundación Jiménez Díaz UAM, Madrid, Spain
| | | | - Aránzazu Mediero
- Bone and Joint Research Unit, FIIS-Fundación Jiménez Díaz UAM, Madrid, Spain
| |
Collapse
|
27
|
Tosh D, Fisher CL, Salmaso V, Wan TC, Campbell RG, Chen E, Gao ZG, Auchampach JA, Jacobson KA. First Potent Macrocyclic A 3 Adenosine Receptor Agonists Reveal G-Protein and β-Arrestin2 Signaling Preferences. ACS Pharmacol Transl Sci 2023; 6:1288-1305. [PMID: 37705595 PMCID: PMC10496144 DOI: 10.1021/acsptsci.3c00126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Indexed: 09/15/2023]
Abstract
(N)-Methanocarba adenosine derivatives (A3 adenosine receptor (AR) agonists containing bicyclo[3.1.0]hexane replacing furanose) were chain-extended at N6 and C2 positions with terminal alkenes for ring closure. The resulting macrocycles of 17-20 atoms retained affinity, indicating a spatially proximal orientation of these receptor-bound chains, consistent with molecular modeling of 12. C2-Arylethynyl-linked macrocycle 19 was more A3AR-selective than 2-ether-linked macrocycle 12 (both 5'-methylamides, human (h) A3AR affinities (Ki): 22.1 and 25.8 nM, respectively), with lower mouse A3AR affinities. Functional hA3AR comparison of two sets of open/closed analogues in β-arrestin2 and Gi/o protein assays showed certain signaling preferences divergent from reference agonist Cl-IB-MECA 1. The potencies of 1 at all three Gαi isoforms were slightly less than its hA3AR binding affinity (Ki: 1.4 nM), while the Gαi1 and Gαi2 potencies of macrocycle 12 were roughly an order of magnitude higher than its radioligand binding affinity. Gαi2-coupling was enhanced in macrocycle 12 (EC50 2.56 nM, ∼40% greater maximal efficacy than 1). Di-O-allyl precursor 18 cyclized to form 19, increasing the Gαi1 potency by 7.5-fold. The macrocycles 12 and 19 and their open precursors 11 and 18 potently stimulated β-arrestin2 recruitment, with EC50 values (nM) of 5.17, 4.36, 1.30, and 4.35, respectively, and with nearly 50% greater efficacy compared to 1. This example of macrocyclization altering the coupling pathways of small-molecule (nonpeptide) GPCR agonists is the first for potent and selective macrocyclic AR agonists. These initial macrocyclic derivatives can serve as a guide for the future design of macrocyclic AR agonists displaying unanticipated pharmacology.
Collapse
Affiliation(s)
- Dilip
K. Tosh
- Laboratory
of Bioorganic Chemistry, National Institute of Diabetes and Digestive
and Kidney Disease, National Institutes
of Health, 9000 Rockville
Pike, Bethesda, Maryland 20892, United States
| | - Courtney L. Fisher
- Department
of Pharmacology & Toxicology and the Cardiovascular Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, Wisconsin 53226, United States
| | - Veronica Salmaso
- Laboratory
of Bioorganic Chemistry, National Institute of Diabetes and Digestive
and Kidney Disease, National Institutes
of Health, 9000 Rockville
Pike, Bethesda, Maryland 20892, United States
- Molecular
Modeling Section, Department of Pharmaceutical and Pharmacological
Sciences, University of Padua, Padua 35131, Italy
| | - Tina C. Wan
- Department
of Pharmacology & Toxicology and the Cardiovascular Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, Wisconsin 53226, United States
| | - Ryan G. Campbell
- Laboratory
of Bioorganic Chemistry, National Institute of Diabetes and Digestive
and Kidney Disease, National Institutes
of Health, 9000 Rockville
Pike, Bethesda, Maryland 20892, United States
| | - Eric Chen
- Laboratory
of Bioorganic Chemistry, National Institute of Diabetes and Digestive
and Kidney Disease, National Institutes
of Health, 9000 Rockville
Pike, Bethesda, Maryland 20892, United States
| | - Zhan-Guo Gao
- Laboratory
of Bioorganic Chemistry, National Institute of Diabetes and Digestive
and Kidney Disease, National Institutes
of Health, 9000 Rockville
Pike, Bethesda, Maryland 20892, United States
| | - John A. Auchampach
- Department
of Pharmacology & Toxicology and the Cardiovascular Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, Wisconsin 53226, United States
| | - Kenneth A. Jacobson
- Laboratory
of Bioorganic Chemistry, National Institute of Diabetes and Digestive
and Kidney Disease, National Institutes
of Health, 9000 Rockville
Pike, Bethesda, Maryland 20892, United States
| |
Collapse
|
28
|
Pottie E, Suresh RR, Jacobson KA, Stove CP. Assay-Dependent Inverse Agonism at the A 3 Adenosine Receptor: When Neutral Is Not Neutral. ACS Pharmacol Transl Sci 2023; 6:1266-1274. [PMID: 37705594 PMCID: PMC10496142 DOI: 10.1021/acsptsci.3c00071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Indexed: 09/15/2023]
Abstract
The A3 adenosine receptor (A3AR) is implicated in a variety of (patho)physiological conditions. While most research has focused on agonists and antagonists, inverse agonism at A3AR has been scarcely studied. Therefore, this study aimed at exploring inverse agonism, using two previously engineered cell lines (hA3ARLgBiT-SmBiTβarr2 and hA3ARLgBiT-SmBiTminiGαi), both employing the NanoBiT technology. The previously established inverse agonist PSB-10 showed a decrease in basal signal in the β-arrestin 2 (βarr2) but not the miniGαi recruitment assay, indicative of inverse agonism in the former assay. Control experiments confirmed the specificity and reversibility of this observation. Evaluation of a set of presumed neutral antagonists (MRS7907, MRS7799, XAC, and MRS1220) revealed that all displayed concentration-dependent signal decreases when tested in the A3AR-βarr2 recruitment assay, yielding EC50 and Emax values for inverse agonism. Conversely, in the miniGαi recruitment assay, no signal decreases were observed. To assess whether this observation was caused by the inability of the ligands to induce inverse agonism in the G protein pathway, or rather by a limitation inherent to the employed A3AR-miniGαi recruitment assay, a GloSensor cAMP assay was performed. The outcome of the latter also suggests inverse agonism by the presumed neutral antagonists in this latter assay. These findings emphasize the importance of prior characterization of ligands in the relevant test system. Moreover, it showed the suitability of the NanoBiT βarr2 recruitment and the GloSensor cAMP assays to capture inverse agonism at the A3AR, as opposed to the NanoBiT miniGαi recruitment assay.
Collapse
Affiliation(s)
- Eline Pottie
- Laboratory
of Toxicology, Department of Bioanalysis, Faculty of Pharmaceutical
Sciences, Ghent University, Campus Heymans, Ottergemsesteenweg
460, B-9000 Ghent, Belgium
| | - R. Rama Suresh
- Laboratory
of Bioorganic Chemistry, National Institute
of Diabetes & Digestive & Kidney Diseases, National Institutes
of Health, Bethesda, Maryland 20802, United States
| | - Kenneth A. Jacobson
- Laboratory
of Bioorganic Chemistry, National Institute
of Diabetes & Digestive & Kidney Diseases, National Institutes
of Health, Bethesda, Maryland 20802, United States
| | - Christophe P. Stove
- Laboratory
of Toxicology, Department of Bioanalysis, Faculty of Pharmaceutical
Sciences, Ghent University, Campus Heymans, Ottergemsesteenweg
460, B-9000 Ghent, Belgium
| |
Collapse
|
29
|
Jia W, Huang Z, Zhou L, Liou YC, Di Virgilio F, Ulrich H, Illes P, Zhang W, Huang C, Tang Y. Purinergic signalling in cancer therapeutic resistance: From mechanisms to targeting strategies. Drug Resist Updat 2023; 70:100988. [PMID: 37413937 DOI: 10.1016/j.drup.2023.100988] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 06/05/2023] [Accepted: 06/23/2023] [Indexed: 07/08/2023]
Abstract
Purinergic signalling, consisting of extracellular purines and purinergic receptors, modulates cell proliferation, invasion and immunological reaction during cancer progression. Here, we focus on current evidence that suggests the crucial role of purinergic signalling in mediating cancer therapeutic resistance, the major obstacle in cancer treatment. Mechanistically, purinergic signalling can modulate the tumor microenvironment (TME), epithelial-mesenchymal transition (EMT) and anti-tumor immunity, thus affecting drug sensitivity of tumor cells. Currently, some agents attempting to target purinergic signalling either in tumor cells or in tumor-associated immune cells are under preclinical or clinical investigation. Moreover, nano-based delivery technologies significantly improve the efficacy of agents targeting purinergic signalling. In this review article, we summarize the mechanisms of purinergic signalling in promoting cancer therapeutic resistance and discuss the potentials and challenges of targeting purinergic signalling in future cancer treatment.
Collapse
Affiliation(s)
- Wenhui Jia
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Zhao Huang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Li Zhou
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Yih-Cherng Liou
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore 117573, Singapore; Department of Biological Sciences, Faculty of Science, National University of Singapore, 14 Science Drive 4, Singapore 117573, Singapore
| | | | - Henning Ulrich
- International Joint Research Centre on Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China; Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Peter Illes
- International Joint Research Centre on Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China; Rudolf-Boehm-Institut für Pharmakologie und Toxikologie, Universitaet Leipzig, Leipzig, Germany
| | - Wei Zhang
- Mental Health Center and Psychiatric Laboratory, the State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Canhua Huang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China; Institute of TCM-Based Stress Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Yong Tang
- International Joint Research Centre on Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China; School of Health and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China; Acupuncture and Chronobiology Key Laboratory of Sichuan Province, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China.
| |
Collapse
|
30
|
Beerkens BL, Snijders IM, Snoeck J, Liu R, Tool ATJ, Le Dévédec SE, Jespers W, Kuijpers TW, van Westen GJ, Heitman LH, IJzerman AP, van der Es D. Development of an Affinity-Based Probe to Profile Endogenous Human Adenosine A3 Receptor Expression. J Med Chem 2023; 66:11399-11413. [PMID: 37531576 PMCID: PMC10461224 DOI: 10.1021/acs.jmedchem.3c00854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Indexed: 08/04/2023]
Abstract
The adenosine A3 receptor (A3AR) is a G protein-coupled receptor (GPCR) that exerts immunomodulatory effects in pathophysiological conditions such as inflammation and cancer. Thus far, studies toward the downstream effects of A3AR activation have yielded contradictory results, thereby motivating the need for further investigations. Various chemical and biological tools have been developed for this purpose, ranging from fluorescent ligands to antibodies. Nevertheless, these probes are limited by their reversible mode of binding, relatively large size, and often low specificity. Therefore, in this work, we have developed a clickable and covalent affinity-based probe (AfBP) to target the human A3AR. Herein, we show validation of the synthesized AfBP in radioligand displacement, SDS-PAGE, and confocal microscopy experiments as well as utilization of the AfBP for the detection of endogenous A3AR expression in flow cytometry experiments. Ultimately, this AfBP will aid future studies toward the expression and function of the A3AR in pathologies.
Collapse
Affiliation(s)
- Bert L.
H. Beerkens
- Division
of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333
CC Leiden, The Netherlands
| | - Inge M. Snijders
- Division
of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333
CC Leiden, The Netherlands
| | - Joep Snoeck
- Division
of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333
CC Leiden, The Netherlands
| | - Rongfang Liu
- Division
of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333
CC Leiden, The Netherlands
| | - Anton T. J. Tool
- Department
of Molecular Hematology, Sanquin Research, Plesmalaan 125, 1066 CX Amsterdam, The Netherlands
| | - Sylvia E. Le Dévédec
- Division
of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333
CC Leiden, The Netherlands
| | - Willem Jespers
- Division
of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333
CC Leiden, The Netherlands
| | - Taco W. Kuijpers
- Department
of Molecular Hematology, Sanquin Research, Plesmalaan 125, 1066 CX Amsterdam, The Netherlands
- Department
of Pediatric Immunology, Rheumatology and Infectious Diseases, Emma
Children’s Hospital, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Gerard J.P. van Westen
- Division
of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333
CC Leiden, The Netherlands
| | - Laura H. Heitman
- Division
of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333
CC Leiden, The Netherlands
- Oncode
Institute, Einsteinweg
55, 2333 CC Leiden, The Netherlands
| | - Adriaan P. IJzerman
- Division
of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333
CC Leiden, The Netherlands
| | - Daan van der Es
- Division
of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333
CC Leiden, The Netherlands
| |
Collapse
|
31
|
Fu K, Hui C, Wang X, Ji T, Li X, Sun R, Xing C, Fan X, Gao Y, Su L. Torpor-like Hypothermia Induced by A1 Adenosine Receptor Agonist: A Novel Approach to Protect against Neuroinflammation. Int J Mol Sci 2023; 24:11036. [PMID: 37446216 DOI: 10.3390/ijms241311036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 06/27/2023] [Accepted: 06/29/2023] [Indexed: 07/15/2023] Open
Abstract
Hypothermia is a promising clinical therapy for acute injuries, including neural damage, but it also faces practical limitations due to the complexities of the equipment and procedures required. This study investigates the use of the A1 adenosine receptor (A1AR) agonist N6-cyclohexyladenosine (CHA) as a more accessible method to induce steady, torpor-like hypothermic states. Additionally, this study investigates the protective potential of CHA against LPS-induced sepsis and neuroinflammation. Our results reveal that CHA can successfully induce a hypothermic state by activating a neuronal circuit similar to the one that induces physiological torpor. This state is characterized by maintaining a steady core body temperature below 28 °C. We further found that this torpor-like state effectively mitigates neuroinflammation and preserves the integrity of the blood-brain barrier during sepsis, thereby limiting the infiltration of inflammatory factors into the central nervous system. Instead of being a direct effect of CHA, this protective effect is attributed to inhibiting pro-inflammatory responses in macrophages and reducing oxidative stress damage in endothelial cells under systemic hypothermia. These results suggest that A1AR agonists such as CHA could potentially be potent neuroprotective agents against neuroinflammation. They also shed light on possible future directions for the application of hypothermia-based therapies in the treatment of sepsis and other neuroinflammatory conditions.
Collapse
Affiliation(s)
- Kang Fu
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Chunlei Hui
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
| | - Xinyuan Wang
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Tingting Ji
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Xiuqing Li
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Rui Sun
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Chunlei Xing
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
| | - Xi Fan
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Yuanqing Gao
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Li Su
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
| |
Collapse
|
32
|
Cervellera CF, Mazziotta C, Di Mauro G, Iaquinta MR, Mazzoni E, Torreggiani E, Tognon M, Martini F, Rotondo JC. Immortalized erythroid cells as a novel frontier for in vitro blood production: current approaches and potential clinical application. Stem Cell Res Ther 2023; 14:139. [PMID: 37226267 PMCID: PMC10210309 DOI: 10.1186/s13287-023-03367-8] [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: 01/17/2023] [Accepted: 05/05/2023] [Indexed: 05/26/2023] Open
Abstract
BACKGROUND Blood transfusions represent common medical procedures, which provide essential supportive therapy. However, these procedures are notoriously expensive for healthcare services and not without risk. The potential threat of transfusion-related complications, such as the development of pathogenic infections and the occurring of alloimmunization events, alongside the donor's dependence, strongly limits the availability of transfusion units and represents significant concerns in transfusion medicine. Moreover, a further increase in the demand for donated blood and blood transfusion, combined with a reduction in blood donors, is expected as a consequence of the decrease in birth rates and increase in life expectancy in industrialized countries. MAIN BODY An emerging and alternative strategy preferred over blood transfusion is the in vitro production of blood cells from immortalized erythroid cells. The high survival capacity alongside the stable and longest proliferation time of immortalized erythroid cells could allow the generation of a large number of cells over time, which are able to differentiate into blood cells. However, a large-scale, cost-effective production of blood cells is not yet a routine clinical procedure, as being dependent on the optimization of culture conditions of immortalized erythroid cells. CONCLUSION In our review, we provide an overview of the most recent erythroid cell immortalization approaches, while also describing and discussing related advancements of establishing immortalized erythroid cell lines.
Collapse
Affiliation(s)
- Christian Felice Cervellera
- Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121, Ferrara, Italy
| | - Chiara Mazziotta
- Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121, Ferrara, Italy
- Department of Medical Sciences, Center for Studies on Gender Medicine, University of Ferrara, 64/b, Fossato di Mortara Street, 44121, Ferrara, Italy
| | - Giulia Di Mauro
- Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121, Ferrara, Italy
| | - Maria Rosa Iaquinta
- Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121, Ferrara, Italy
- Department of Medical Sciences, Center for Studies on Gender Medicine, University of Ferrara, 64/b, Fossato di Mortara Street, 44121, Ferrara, Italy
| | - Elisa Mazzoni
- Department of Chemical, Pharmaceutical and Agricultural Sciences-DOCPAS, University of Ferrara, 44121, Ferrara, Italy
| | - Elena Torreggiani
- Department of Chemical, Pharmaceutical and Agricultural Sciences-DOCPAS, University of Ferrara, 44121, Ferrara, Italy
| | - Mauro Tognon
- Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121, Ferrara, Italy
| | - Fernanda Martini
- Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121, Ferrara, Italy.
- Department of Medical Sciences, Center for Studies on Gender Medicine, University of Ferrara, 64/b, Fossato di Mortara Street, 44121, Ferrara, Italy.
- Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121, Ferrara, Italy.
| | - John Charles Rotondo
- Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121, Ferrara, Italy.
- Department of Medical Sciences, Center for Studies on Gender Medicine, University of Ferrara, 64/b, Fossato di Mortara Street, 44121, Ferrara, Italy.
| |
Collapse
|
33
|
Rafii S, Ghouzlani A, Naji O, Ait Ssi S, Kandoussi S, Lakhdar A, Badou A. A 2AR as a Prognostic Marker and a Potential Immunotherapy Target in Human Glioma. Int J Mol Sci 2023; 24:6688. [PMID: 37047660 PMCID: PMC10095519 DOI: 10.3390/ijms24076688] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/05/2023] [Accepted: 03/14/2023] [Indexed: 04/07/2023] Open
Abstract
Gliomas are considered one of the most malignant tumors in the body. The immune system has the ability to control the initiation and development of tumors, including gliomas. Thus, immune cells find themselves controlled by various molecular pathways, inhibiting their activation, such as the immunosuppressive adenosine 2A receptor (A2AR). Our objective was to establish the expression profile and role of A2AR at the transcriptomic level, using real-time RT-PCR in Moroccan glioma patients, in addition to TCGA and CGGA cohorts. The real-time RT-PCR results in Moroccan patients showed that high expression of this gene was associated with poor survival in males. Our study on the CGGA cohort corroborated these results. In addition, there was a positive association of A2AR with T-cell exhaustion genes. A2AR also correlated strongly with genes that are primarily enriched in focal adhesion and extracellular matrix interactions, inducing epithelial mesenchymal transition, angiogenesis, and glioma growth. However, in the TCGA cohort, the A2AR showed results that were different from the two previously examined cohorts. In fact, this gene was instead linked to a good prognosis in patients with the astrocytoma histological type. The correlation and enrichment results reinforced the prognostic role of A2AR in this TCGA cohort, in which its high expression was shown to be related to lymphocyte differentiation and a successful cytolytic response, suggesting a more efficient anti-tumor immune response. Correlations and differential analyses based on A2AR gene expression, to understand the cause of the association of this gene with two different prognoses (CGGA males and TCGA Astrocytoma), showed that the overexpression of A2AR in Chinese male patients could be associated with the overexpression of extracellular adenosine, which binds to A2AR to induce immunosuppression and consequently a poor prognosis. However, in the second group (TCGA astrocytomas), the overexpression of the gene could be associated with an adenosine deficiency, and therefore this receptor does not undergo activation. The absence of A2AR activation in these patients may have protected them from immunosuppression, which could reflect the good prognosis. A2AR can be considered a promising therapeutic target in male CGGA and Moroccan patients with gliomas.
Collapse
Affiliation(s)
- Soumaya Rafii
- Immuno-Genetics and Human Pathologies Laboratory, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca 20000, Morocco
| | - Amina Ghouzlani
- Immuno-Genetics and Human Pathologies Laboratory, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca 20000, Morocco
| | - Oumayma Naji
- Immuno-Genetics and Human Pathologies Laboratory, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca 20000, Morocco
| | - Saadia Ait Ssi
- Immuno-Genetics and Human Pathologies Laboratory, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca 20000, Morocco
| | - Sarah Kandoussi
- Immuno-Genetics and Human Pathologies Laboratory, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca 20000, Morocco
| | | | - Abdallah Badou
- Immuno-Genetics and Human Pathologies Laboratory, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca 20000, Morocco
- Mohammed VI Center for Research and Innovation, Rabat, Morocco and Mohammed VI University of Sciences and Health, Casablanca 82403, Morocco
| |
Collapse
|
34
|
Rahimi MR, Semenova EA, Larin AK, Kulemin NA, Generozov EV, Łubkowska B, Ahmetov II, Golpasandi H. The ADORA2A TT Genotype Is Associated with Anti-Inflammatory Effects of Caffeine in Response to Resistance Exercise and Habitual Coffee Intake. Nutrients 2023; 15:nu15071634. [PMID: 37049474 PMCID: PMC10097079 DOI: 10.3390/nu15071634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/21/2023] [Accepted: 03/27/2023] [Indexed: 03/30/2023] Open
Abstract
Caffeine is an adenosine A2A receptor (ADORA2A) antagonist with ergogenic and anti-inflammatory effects. Previous studies have reported that the ADORA2A gene regulates glutamate metabolism and immune responses, with the ADORA2A rs5751876 TT genotype (with high sensitivity to caffeine) showing larger ergogenic effect following caffeine ingestion. We therefore hypothesized that the TT genotype would be associated with greater anti-inflammatory effects of caffeine in response to exercise, and with higher coffee intake in physically active individuals. The aim of the present study was twofold: (1) to investigate the association of the ADORA2A variant with the anti-inflammatory effects of caffeine in response to intense resistance exercise (RE), and (2) to analyze the association of the rs5751876 with coffee intake in physically active individuals (n = 134). Fifteen resistance-trained athletes participated in a randomized, double-blind, placebo-controlled cross-over study, where they consumed 6 mg/kg of caffeine or placebo one hour prior to performing an RE protocol. Blood samples were taken immediately from the arterial vein before, immediately after, and 15 min after RE for the analysis of inflammatory markers myeloperoxidase (MPO) and acetylcholinesterase (AChE). We found that the ADORA2A TT genotype carriers experienced lower exercise-induced inflammatory responses (p < 0.05 for AchE) when compared to the C allele carriers (i.e., CC/CT) one hour following the ingestion of caffeine. Furthermore, the ADORA2A TT genotype was positively associated with coffee intake (p = 0.0143; irrespective of CYP1A2 rs762551 polymorphism). In conclusion, we found that the ADORA2A gene polymorphism is associated with anti-inflammatory effects of caffeine in response to resistance exercise, as well as with habitual coffee intake in physically active individuals.
Collapse
|
35
|
Natural and synthetic compounds for glioma treatment based on ROS-mediated strategy. Eur J Pharmacol 2023:175537. [PMID: 36871663 DOI: 10.1016/j.ejphar.2023.175537] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 01/08/2023] [Accepted: 01/23/2023] [Indexed: 03/06/2023]
Abstract
Glioma is the most frequent and most malignant tumor of the central nervous system (CNS),accounting for about 50% of all CNS tumor and approximately 80% of the malignant primary tumors in the CNS. Patients with glioma benefit from surgical resection, chemo- and radio-therapy. However these therapeutical strategies do not significantly improve the prognosis, nor increase survival rates owing to restricted drug contribution in the CNS and to the malignant characteristics of glioma. Reactive oxygen species (ROS) are important oxygen-containing molecules that regulate tumorigenesis and tumor progression. When ROS accumulates to cytotoxic levels, this can lead to anti-tumor effects. Multiple chemicals used as therapeutic strategies are based on this mechanism. They regulate intracellular ROS levels directly or indirectly, resulting in the inability of glioma cells to adapt to the damage induced by these substances. In the current review, we summarize the natural products, synthetic compounds and interdisciplinary techniques used for the treatment of glioma. Their possible molecular mechanisms are also presented. Some of them are also used as sensitizers: they modulate ROS levels to improve the outcomes of chemo- and radio-therapy. In addition, we summarize some new targets upstream or downstream of ROS to provide ideas for developing new anti-glioma therapies.
Collapse
|
36
|
Akbar H, Fasick JJ, Ponnuraj N, Jarosinski KW. Purinergic signaling during Marek's disease in chickens. Sci Rep 2023; 13:2044. [PMID: 36739336 PMCID: PMC9899245 DOI: 10.1038/s41598-023-29210-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Purinergic receptors (PRs) have been reported as potential therapeutic targets for many viral infections including herpesviruses, which urges the investigation into their role in Marek's disease (MD), a herpesvirus induced cancer in chickens that is an important pathogen for the poultry industry. MD is caused by MD virus (MDV) that has a similar viral life cycle as human varicella zoster virus in that it is shed from infected epithelial skin cells and enters the host through the respiratory route. In this report, PR responses during natural MDV infection and disease progression was examined in MD-resistant white Leghorns (WL) and MD-susceptible Pure Columbian (PC) chickens during natural infection. Whole lung lavage cells (WLLC) and liver tissue samples were collected from chickens infected but showing no clinical signs of MD (Infected) or presenting with clinical disease (Diseased). RNA was extracted followed by RT-qPCR analysis with gene specific primers against members of the P1, P2X, and P2Y PR families. Differential expression (p < 0.05) was observed in breed and disease conditions. Some PRs showed tissue specific expression (P1A1, P2X1, and P2X6 in WLLC) whereas others responded to MDV infection only in MD-susceptible (PC) chickens (P1A2A, P2X1, P2X5, P2X7). P2Y PRs had differential expression in both chicken lines in response to MDV infection and MD progression. This study is the first to our knowledge to examine PR responses during MDV infection and disease progression. These results suggest PR signaling may an important area of research for MDV replication and MD.
Collapse
Affiliation(s)
- Haji Akbar
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Julia J Fasick
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Nagendraprabhu Ponnuraj
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Keith W Jarosinski
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
37
|
Mazziotta C, Tognon M, Martini F, Torreggiani E, Rotondo JC. Probiotics Mechanism of Action on Immune Cells and Beneficial Effects on Human Health. Cells 2023; 12:cells12010184. [PMID: 36611977 PMCID: PMC9818925 DOI: 10.3390/cells12010184] [Citation(s) in RCA: 192] [Impact Index Per Article: 96.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/12/2022] [Accepted: 12/29/2022] [Indexed: 01/03/2023] Open
Abstract
Immune cells and commensal microbes in the human intestine constantly communicate with and react to each other in a stable environment in order to maintain healthy immune activities. Immune system-microbiota cross-talk relies on a complex network of pathways that sustain the balance between immune tolerance and immunogenicity. Probiotic bacteria can interact and stimulate intestinal immune cells and commensal microflora to modulate specific immune functions and immune homeostasis. Growing evidence shows that probiotic bacteria present important health-promoting and immunomodulatory properties. Thus, the use of probiotics might represent a promising approach for improving immune system activities. So far, few studies have been reported on the beneficial immune modulatory effect of probiotics. However, many others, which are mainly focused on their metabolic/nutritional properties, have been published. Therefore, the mechanisms behind the interaction between host immune cells and probiotics have only been partially described. The present review aims to collect and summarize the most recent scientific results and the resulting implications of how probiotic bacteria and immune cells interact to improve immune functions. Hence, a description of the currently known immunomodulatory mechanisms of probiotic bacteria in improving the host immune system is provided.
Collapse
Affiliation(s)
- Chiara Mazziotta
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
- Center for Studies on Gender Medicine, Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121 Ferrara, Italy
| | - Mauro Tognon
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Fernanda Martini
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
- Center for Studies on Gender Medicine, Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121 Ferrara, Italy
- Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy
| | - Elena Torreggiani
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
- Correspondence: (E.T.); (J.C.R.); Tel.: +39-053-2455-557 (E.T.); +39-053-245-5536 (J.C.R.)
| | - John Charles Rotondo
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
- Center for Studies on Gender Medicine, Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121 Ferrara, Italy
- Correspondence: (E.T.); (J.C.R.); Tel.: +39-053-2455-557 (E.T.); +39-053-245-5536 (J.C.R.)
| |
Collapse
|
38
|
Mazziotta C, Cervellera CF, Lanzillotti C, Touzé A, Gaboriaud P, Tognon M, Martini F, Rotondo JC. MicroRNA dysregulations in Merkel cell carcinoma: Molecular mechanisms and clinical applications. J Med Virol 2023; 95:e28375. [PMID: 36477874 DOI: 10.1002/jmv.28375] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 11/22/2022] [Accepted: 11/25/2022] [Indexed: 12/13/2022]
Abstract
Merkel cell carcinoma (MCC) is an aggressive skin malignancy with two distinct etiologies. The first, which accounts for the highest proportion, is caused by Merkel cell polyomavirus (MCPyV), a DNA tumor virus. A second, UV-induced, MCC form has also been identified. Few MCC diagnostic, prognostic, and therapeutic options are available. MicroRNAs (miRNAs) are small noncoding RNA molecules, which play a key role in regulating various physiologic cellular functions including cell cycling, proliferation, differentiation, and apoptosis. Numerous miRNAs are dysregulated in cancer, by acting as either tumor suppressors or oncomiRs. The aim of this review is to collect, summarize, and discuss recent findings on miRNAs whose dysregulation has been assumed to play a role in MCC. The potential clinical application of miRNAs as diagnostic and prognostic biomarkers in MCC is also described. In the future, miRNAs will potentially gain clinical significance for the improvement of MCC diagnostic, prognostic, and therapeutic options.
Collapse
Affiliation(s)
- Chiara Mazziotta
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy.,Department of Medical Sciences, Center for Studies on Gender Medicine, University of Ferrara, Ferrara, Italy
| | | | - Carmen Lanzillotti
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy.,Department of Medical Sciences, Center for Studies on Gender Medicine, University of Ferrara, Ferrara, Italy
| | - Antoine Touzé
- "Biologie des infections à polyomavirus" Team, UMR INRAE 1282, University of Tours, Tours, France
| | - Pauline Gaboriaud
- "Biologie des infections à polyomavirus" Team, UMR INRAE 1282, University of Tours, Tours, France
| | - Mauro Tognon
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Fernanda Martini
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy.,Department of Medical Sciences, Center for Studies on Gender Medicine, University of Ferrara, Ferrara, Italy.,Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - John Charles Rotondo
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy.,Department of Medical Sciences, Center for Studies on Gender Medicine, University of Ferrara, Ferrara, Italy
| |
Collapse
|
39
|
Fakhoury HMA, Elahi MA, Al Sarheed S, Al Dubayee M, Alshahrani A, Zhra M, Almassri A, Aljada A. Gene Expression Profiling of Peripheral Blood Mononuclear Cells in Type 2 Diabetes: An Exploratory Study. Medicina (B Aires) 2022; 58:medicina58121829. [PMID: 36557031 PMCID: PMC9787392 DOI: 10.3390/medicina58121829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 11/28/2022] [Accepted: 12/06/2022] [Indexed: 12/15/2022] Open
Abstract
Background and Objectives: Visceral obesity is associated with chronic low-grade inflammation that predisposes to metabolic syndrome. Indeed, infiltration of adipose tissue with immune-inflammatory cells, including 'classical' inflammatory M1 and anti-inflammatory 'alternative' M2 macrophages, causes the release of a variety of bioactive molecules, resulting in the metabolic complications of obesity. This study examined the relative expression of macrophage phenotypic surface markers, cholesterol efflux proteins, scavenger receptors, and adenosine receptors in human circulating peripheral blood mononuclear cells (PBMCs), isolated from patients with type 2 diabetes mellitus (T2DM), with the aim to phenotypically characterize and identify biomarkers for these ill-defined cells. Materials and Methodology: PBMCs were isolated from four groups of adults: Normal-weight non-diabetic, obese non-diabetic, newly diagnosed with T2DM, and T2DM on metformin. The mRNA expression levels of macrophage phenotypic surface markers (interleukin-12 (IL-12), C-X-C motif chemokine ligand 10 (CXCL10), C-C motif chemokine ligand 17 (CCL17), and C-C motif receptor 7 (CCR7)), cholesterol efflux proteins (ATP-binding cassette transporter-1 (ABCA1), ATP binding cassette subfamily G member 1 (ABCG1), and sterol 27-hydroxylase (CYP27A)), scavenger receptors (scavenger receptor-A (SR-A), C-X-C motif ligand 16 (CXCL16), and lectin-like oxidized LDL receptor-1 (LOX-1)), and adenosine receptors (adenosine A2A receptor (A2AR) and adenosine A3 receptor (A3R)) were measured using qRT-PCR. Results: In PBMCs from T2DM patients, the expression of IL-12, CCR7, ABCA1, and SR-A1 was increased, whereas the expression of CXCL10, CCL17, ABCG1,27-hydroxylase, LOX-1, A2AR and A3R was decreased. On the other hand, treatment with the antidiabetic drug, metformin, reduced the expression of IL-12 and increased the expression of 27-hydroxylase, LOX-1, CXCL16 and A2AR. Conclusions: PBMCs in the circulation of patients with T2DM express phenotypic markers that are different from those typically present in adipose tissue M1 and M2 macrophages and could be representative of metabolically activated macrophages (MMe)-like cells. Our findings suggest that metformin alters phenotypic markers of MMe-like cells in circulation.
Collapse
Affiliation(s)
- Hana M. A. Fakhoury
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
- Correspondence: (H.M.A.F.); (A.A.)
| | - Muhammad Affan Elahi
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Saud Al Sarheed
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh 11481, Saudi Arabia
| | - Mohammed Al Dubayee
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh 11481, Saudi Arabia
- King Abdullah International Medical Research Center (KAIMRC), Riyadh 11481, Saudi Arabia
- Department of Medicine, Ministry of National Guard Health Affairs (MNG-HA), Riyadh 11426, Saudi Arabia
| | - Awad Alshahrani
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh 11481, Saudi Arabia
- King Abdullah International Medical Research Center (KAIMRC), Riyadh 11481, Saudi Arabia
- Department of Medicine, Ministry of National Guard Health Affairs (MNG-HA), Riyadh 11426, Saudi Arabia
| | - Mahmoud Zhra
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Arwa Almassri
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Ahmad Aljada
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
- Correspondence: (H.M.A.F.); (A.A.)
| |
Collapse
|
40
|
Nucleoside transporters and immunosuppressive adenosine signaling in the tumor microenvironment: Potential therapeutic opportunities. Pharmacol Ther 2022; 240:108300. [PMID: 36283452 DOI: 10.1016/j.pharmthera.2022.108300] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 10/17/2022] [Accepted: 10/19/2022] [Indexed: 11/30/2022]
Abstract
Adenosine compartmentalization has a profound impact on immune cell function by regulating adenosine localization and, therefore, extracellular signaling capabilities, which suppresses immune cell function in the tumor microenvironment. Nucleoside transporters, responsible for the translocation and cellular compartmentalization of hydrophilic adenosine, represent an understudied yet crucial component of adenosine disposition in the tumor microenvironment. In this review article, we will summarize what is known regarding nucleoside transporter's function within the purinome in relation to currently devised points of intervention (i.e., ectonucleotidases, adenosine receptors) for cancer immunotherapy, alterations in nucleoside transporter expression reported in cancer, and potential avenues for targeting of nucleoside transporters for the desired modulation of adenosine compartmentalization and action. Further, we put forward that nucleoside transporters are an unexplored therapeutic opportunity, and modulation of nucleoside transport processes could attenuate the pathogenic buildup of immunosuppressive adenosine in solid tumors, particularly those enriched with nucleoside transport proteins.
Collapse
|
41
|
Elaskalani O, Gilmore G, Hagger M, Baker RI, Metharom P. Adenosine 2A Receptor Activation Amplifies Ibrutinib Antiplatelet Effect; Implications in Chronic Lymphocytic Leukemia. Cancers (Basel) 2022; 14:cancers14235750. [PMID: 36497231 PMCID: PMC9741389 DOI: 10.3390/cancers14235750] [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: 10/24/2022] [Revised: 11/20/2022] [Accepted: 11/21/2022] [Indexed: 11/24/2022] Open
Abstract
Chronic lymphocytic leukemia patients have an increased bleeding risk with the introduction of Bruton tyrosine kinase (BTK) inhibitors. BTK is a signaling effector downstream of the platelet GPVI receptor. Innate platelet dysfunction in CLL patients and the contribution of the leukemia microenvironment to the anti-platelet effect of BTK inhibitors are still not well defined. Herein, we investigated platelet function in stable, untreated CLL patients in comparison to age-matched healthy subjects as control. Secondly, we proposed a novel mechanism of platelet dysfunction via the adenosinergic pathway during BTK inhibitor therapy. Our data indicate that the nucleotidase that produces adenosine, CD73, was expressed on one-third of B-cells in CLL patients. Inhibition of CD73 improved platelet response to ADP in the blood of CLL patients ex vivo. Using healthy platelets, we show that adenosine 2A (A2A) receptor activation amplifies the anti-platelet effect of ibrutinib (10 nM). Ibrutinib plus an A2A agonist-but not ibrutinib as a single agent-significantly inhibited collagen (10 µg/mL)-induced platelet aggregation. Mechanistically, A2A activation attenuated collagen-mediated inhibition of p-VASP and synergized with ibrutinib to inhibit the phosphorylation of AKT, ERK and SYK kinases. This manuscript highlights the potential role of adenosine generated by the microenvironment in ibrutinib-associated bleeding in CLL patients.
Collapse
Affiliation(s)
- Omar Elaskalani
- Telethon Kids Institute, Cancer Centre, Nedlands, WA 6009, Australia
- Centre for Child Health Research, University of Western Australia, Crawley, WA 6009, Australia
| | - Grace Gilmore
- Perth Blood Institute (PBI), Perth, WA 6005, Australia
- Western Australian Centre for Thrombosis and Haemostasis (WACTH), Health Futures Institute, Murdoch University, Murdoch, WA 6150, Australia
| | - Madison Hagger
- Platelet Research Laboratory, School of Pharmacy and Biomedical Sciences, Curtin Health and Innovation Research Institute (CHIRI), Faculty of Health Sciences, Curtin University, Bentley, WA 6102, Australia
| | - Ross I. Baker
- Perth Blood Institute (PBI), Perth, WA 6005, Australia
- Western Australian Centre for Thrombosis and Haemostasis (WACTH), Health Futures Institute, Murdoch University, Murdoch, WA 6150, Australia
- Correspondence: (R.I.B.); (P.M.)
| | - Pat Metharom
- Platelet Research Laboratory, School of Pharmacy and Biomedical Sciences, Curtin Health and Innovation Research Institute (CHIRI), Faculty of Health Sciences, Curtin University, Bentley, WA 6102, Australia
- Correspondence: (R.I.B.); (P.M.)
| |
Collapse
|
42
|
Wang J, Du L, Chen X. Adenosine signaling: Optimal target for gastric cancer immunotherapy. Front Immunol 2022; 13:1027838. [PMID: 36189223 PMCID: PMC9523428 DOI: 10.3389/fimmu.2022.1027838] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 09/05/2022] [Indexed: 11/21/2022] Open
Abstract
Gastric cancer (GC) is one of the most common malignancy and leading cause of cancer-related deaths worldwide. Due to asymptomatic or only nonspecific early symptoms, GC patients are usually in the advanced stage at first diagnosis and miss the best opportunity of treatment. Immunotherapies, especially immune checkpoint inhibitors (ICIs), have dramatically changed the landscape of available treatment options for advanced-stage cancer patients. However, with regards to existing ICIs, the clinical benefit of monotherapy for advanced gastric cancer (AGC) is quite limited. Therefore, it is urgent to explore an optimal target for the treatment of GC. In this review, we summarize the expression profiles and prognostic value of 20 common immune checkpoint-related genes in GC from Gene Expression Profiling Interactive Analysis (GEPIA) database, and then find that the adenosinergic pathway plays an indispensable role in the occurrence and development of GC. Moreover, we discuss the pathophysiological function of adenosinergic pathway in cancers. The accumulation of extracellular adenosine inhibits the normal function of immune effector cells and facilitate the effect of immunosuppressive cells to foster GC cells proliferation and migration. Finally, we provide insights into potential clinical application of adenosinergic-targeting therapies for GC patients.
Collapse
Affiliation(s)
- Junqing Wang
- School of the 1St Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Linyong Du
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, China
- *Correspondence: Xiangjian Chen, ; Linyong Du,
| | - Xiangjian Chen
- School of the 1St Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
- *Correspondence: Xiangjian Chen, ; Linyong Du,
| |
Collapse
|
43
|
Park S, Ahn Y, Kim Y, Roh EJ, Lee Y, Han C, Yoo HM, Yu J. Design, Synthesis and Biological Evaluation of 1,3,5-Triazine Derivatives Targeting hA1 and hA3 Adenosine Receptor. Molecules 2022; 27:molecules27134016. [PMID: 35807265 PMCID: PMC9268102 DOI: 10.3390/molecules27134016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/16/2022] [Accepted: 06/20/2022] [Indexed: 12/04/2022] Open
Abstract
Adenosine mediates various physiological activities in the body. Adenosine receptors (ARs) are widely expressed in tumors and the tumor microenvironment (TME), and they induce tumor proliferation and suppress immune cell function. There are four types of human adenosine receptor (hARs): hA1, hA2A, hA2B, and hA3. Both hA1 and hA3 AR play an important role in tumor proliferation. We designed and synthesized novel 1,3,5-triazine derivatives through amination and Suzuki coupling, and evaluated them for binding affinities to each hAR subtype. Compounds 9a and 11b showed good binding affinity to both hA1 and hA3 AR, while 9c showed the highest binding affinity to hA1 AR. In this study, we discovered that 9c inhibits cell viability, leading to cell death in lung cancer cell lines. Flow cytometry analysis revealed that 9c caused an increase in intracellular reactive oxygen species (ROS) and a depolarization of the mitochondrial membrane potential. The binding mode of 1,3,5-triazine derivatives to hA1 and hA3 AR were predicted by a molecular docking study.
Collapse
Affiliation(s)
- Sujin Park
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea; (S.P.); (C.H.)
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; (Y.K.); (E.J.R.)
| | - Yujin Ahn
- Biometrology Group, Korea Research Institute of Standards and Science (KRISS), Daejeon 34113, Korea;
- Department of Precision Measurement, University of Science and Technology (UST), Daejeon 34113, Korea
| | - Yongchan Kim
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; (Y.K.); (E.J.R.)
| | - Eun Joo Roh
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; (Y.K.); (E.J.R.)
| | - Yoonji Lee
- College of Pharmacy, Chung-Ang University, Seoul 06974, Korea;
| | - Chaebin Han
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea; (S.P.); (C.H.)
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; (Y.K.); (E.J.R.)
| | - Hee Min Yoo
- Biometrology Group, Korea Research Institute of Standards and Science (KRISS), Daejeon 34113, Korea;
- Department of Precision Measurement, University of Science and Technology (UST), Daejeon 34113, Korea
- Correspondence: (H.M.Y.); (J.Y.)
| | - Jinha Yu
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea; (S.P.); (C.H.)
- Correspondence: (H.M.Y.); (J.Y.)
| |
Collapse
|
44
|
Rotondo JC, Martini F, Maritati M, Caselli E, Gallenga CE, Guarino M, De Giorgio R, Mazziotta C, Tramarin ML, Badiale G, Tognon M, Contini C. Advanced Molecular and Immunological Diagnostic Methods to Detect SARS-CoV-2 Infection. Microorganisms 2022; 10:1193. [PMID: 35744711 PMCID: PMC9231257 DOI: 10.3390/microorganisms10061193] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/06/2022] [Accepted: 06/06/2022] [Indexed: 02/06/2023] Open
Abstract
COVID-19 emerged in late 2019 in China and quickly spread across the globe, causing over 521 million cases of infection and 6.26 million deaths to date. After 2 years, numerous advances have been made. First of all, the preventive vaccine, which has been implemented in record time, is effective in more than 95% of cases. Additionally, in the diagnostic field, there are numerous molecular and antigenic diagnostic kits that are equipped with high sensitivity and specificity. Real Time-PCR-based assays for the detection of viral RNA are currently considered the gold-standard method for SARS-CoV-2 diagnosis and can be used efficiently on pooled nasopharyngeal, or oropharyngeal samples for widespread screening. Moreover, additional, and more advanced molecular methods such as droplet-digital PCR (ddPCR), clustered regularly interspaced short palindromic repeats (CRISPR) and next-generation sequencing (NGS), are currently under development to detect the SARS-CoV-2 RNA. However, as the number of subjects infected with SARS-CoV-2 continuously increases globally, health care systems are being placed under increased stress. Thus, the clinical laboratory plays an important role, helping to select especially asymptomatic individuals who are actively carrying the live replicating virus, with fast and non-invasive molecular technologies. Recent diagnostic strategies, other than molecular methods, have been adopted to either detect viral antigens, i.e., antigen-based immunoassays, or human anti-SARS-CoV-2 antibodies, i.e., antibody-based immunoassays, in nasal or oropharyngeal swabs, as well as in blood or saliva samples. However, the role of mucosal sIgAs, which are essential in the control of viruses entering the body through mucosal surfaces, remains to be elucidated, and in particular the role of the immune response in counteracting SARS-CoV-2 infection, primarily at the site(s) of virus entry that appears to be promising.
Collapse
Affiliation(s)
- John Charles Rotondo
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (F.M.); (M.M.); (C.E.G.); (C.M.); (M.L.T.); (G.B.); (M.T.)
- Center for Studies on Gender Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Fernanda Martini
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (F.M.); (M.M.); (C.E.G.); (C.M.); (M.L.T.); (G.B.); (M.T.)
- Center for Studies on Gender Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
- Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy
| | - Martina Maritati
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (F.M.); (M.M.); (C.E.G.); (C.M.); (M.L.T.); (G.B.); (M.T.)
- Orthopaedic Ward, Casa di Cura Santa Maria Maddalena, 45030 Occhiobello, Italy
| | - Elisabetta Caselli
- Section of Microbiology, CIAS Research Center and LTTA, Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy;
| | - Carla Enrica Gallenga
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (F.M.); (M.M.); (C.E.G.); (C.M.); (M.L.T.); (G.B.); (M.T.)
| | - Matteo Guarino
- Department of Translational Medicine, St. Anna University Hospital of Ferrara, University of Ferrara, 44124 Ferrara, Italy; (M.G.); (R.D.G.)
| | - Roberto De Giorgio
- Department of Translational Medicine, St. Anna University Hospital of Ferrara, University of Ferrara, 44124 Ferrara, Italy; (M.G.); (R.D.G.)
| | - Chiara Mazziotta
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (F.M.); (M.M.); (C.E.G.); (C.M.); (M.L.T.); (G.B.); (M.T.)
- Center for Studies on Gender Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Maria Letizia Tramarin
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (F.M.); (M.M.); (C.E.G.); (C.M.); (M.L.T.); (G.B.); (M.T.)
| | - Giada Badiale
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (F.M.); (M.M.); (C.E.G.); (C.M.); (M.L.T.); (G.B.); (M.T.)
| | - Mauro Tognon
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (F.M.); (M.M.); (C.E.G.); (C.M.); (M.L.T.); (G.B.); (M.T.)
| | - Carlo Contini
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (F.M.); (M.M.); (C.E.G.); (C.M.); (M.L.T.); (G.B.); (M.T.)
| |
Collapse
|
45
|
Fishman P. Drugs Targeting the A3 Adenosine Receptor: Human Clinical Study Data. Molecules 2022; 27:3680. [PMID: 35744805 PMCID: PMC9229414 DOI: 10.3390/molecules27123680] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 05/30/2022] [Accepted: 06/02/2022] [Indexed: 02/04/2023] Open
Abstract
The A3 adenosine receptor (A3AR) is overexpressed in pathological human cells. Piclidenoson and namodenoson are A3AR agonists with high affinity and selectivity to A3AR. Both induce apoptosis of cancer and inflammatory cells via a molecular mechanism entailing deregulation of the Wnt and the NF-κB signaling pathways. Our company conducted phase I studies showing the safety of these 2 molecules. In the phase II studies in psoriasis patients, piclidenoson was safe and demonstrated efficacy manifested in significant improvements in skin lesions. Namodenoson is currently being developed to treat liver cancer, where prolonged overall survival was observed in patients with advanced liver disease and a Child-Pugh B score of 7. A pivotal phase III study in this patient population has been approved by the FDA and the EMA and is currently underway. Namodenoson is also being developed to treat non-alcoholic steatohepatitis (NASH). A Phase IIa study has been successfully concluded and showed that namodenoson has anti-inflammatory, anti-fibrosis, and anti-steatosis effects. A phase IIb study in NASH is currently enrolling patients. In conclusion, A3AR agonists are promising drug candidates in advanced stages of clinical development and demonstrate safety and efficacy in their targeted indications.
Collapse
Affiliation(s)
- Pnina Fishman
- Can-Fite BioPharma Ltd., 10 Bareket St., Petah Tikva 49170, Israel
| |
Collapse
|
46
|
Adenosine Conjugated Docetaxel Nanoparticles—Proof of Concept Studies for Non-Small Cell Lung Cancer. Pharmaceuticals (Basel) 2022; 15:ph15050544. [PMID: 35631370 PMCID: PMC9144510 DOI: 10.3390/ph15050544] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/19/2022] [Accepted: 04/24/2022] [Indexed: 02/04/2023] Open
Abstract
Non-small cell lung cancer, a molecularly diverse disease, is the most prevalent cause of cancer mortality globally. Increasing understanding of the clinicopathology of the disease and mechanisms of tumor progression has facilitated early detection and multimodal care. Despite the advancements, survival rates are extremely low due to non-targeted therapeutics and correspondingly increased risk of metastasis. At some phases of cancer, patients need to face the ghost of chemotherapy. It is a difficult decision near the end of life. Such treatments have the capability to prolong survival or reduce symptoms, but can cause serious adverse effects, affecting quality of life of the patient. It is evident that many patients do not die from burden of the disease alone, but they die due to the toxic effect of treatment. Thus, increasing the efficacy is one aspect and decreasing the toxicity is another critical aspect of cancer formulation design. Through our current research, we tried to uncover both mentioned potentials of the formulation. Therefore, we designed actively targeted nanoparticles for improved therapeutics considering the overexpression of adenosine (ADN) receptors on non-small cell lung cancer (NSCLC) cells. Docetaxel (DTX), an essential therapeutic as part of combination therapy or as monotherapy for the treatment of NSCLC, was encapsulated in biodegradable poly(lactic-co-glycolic acid) nanoparticles. ADN was conjugated on the surface of nanoparticles using EDC-NHS chemistry. The particles were characterized in vitro for physicochemical properties, cellular uptake, and biocompatibility. The size and zeta potential of DTX nanoparticles (DPLGA) were found to be 138.4 ± 5.45 nm and −16.7 ± 2.3 mV which were found to change after ADN conjugation. The size was increased to 158.2 ± 6.3 nm, whereas zeta potential was decreased to −11.7 ± 1.4 mV for ADN-conjugated DTX nanoparticles (ADN-DPLGA) indicative of surface conjugation. As observed from transmission electron microscopy (TEM), the nanoparticles were spherical and showed no significant change in encapsulation efficiency even after surface conjugation. Careful and systematic optimization leads to ADN-conjugated PLGA nanoparticles having distinctive characteristic features such as particle size, surface potential, encapsulation efficacy, etc., that may play crucial roles in the fate of nanoparticles (NPs). Consequently, higher cellular uptake in the A549 lung cancer cell line was exhibited by ADN-DPLGA compared to DPLGA, illustrating the role of ADN receptors (ARs) in facilitating the uptake of NPs. Further in vivo pharmacokinetics and tissue distribution experiments revealed prolonged circulation in plasma and significantly higher lung tissue distribution than in other organs, dictating the targeting potential of the developed formulation over naïve drug and unconjugated formulations. Further, in vivo acute toxicity was examined using multiple parameters for non-toxic attributes of the developed formulation compared to other non-targeted organs. Further, it also supports the selection of biocompatible polymers in the formulation. The current study presents a proof-of-concept for a multipronged formulation technology strategy that might be used to maximize anticancer therapeutic responses in the lungs in the treatment of NSCLC. An improved therapeutic and safety profile would help achieve maximum efficacy at a reduced dose that would eventually help reduce the toxicity.
Collapse
|
47
|
Mazziotta C, Lanzillotti C, Gafà R, Touzé A, Durand MA, Martini F, Rotondo JC. The Role of Histone Post-Translational Modifications in Merkel Cell Carcinoma. Front Oncol 2022; 12:832047. [PMID: 35350569 PMCID: PMC8957841 DOI: 10.3389/fonc.2022.832047] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 02/01/2022] [Indexed: 12/12/2022] Open
Abstract
Merkel Cell Carcinoma (MCC) is a rare but highly aggressive form of non–melanoma skin cancer whose 5-year survival rate is 63%. Merkel cell polyomavirus (MCPyV), a small DNA tumor virus, is the etiological agent of MCC. Although representing a small proportion of MCC cases, MCPyV-negative MCCs have also been identified. The role of epigenetic mechanisms, including histone post-translational modifications (PTMs) in MCC, have been only partially determined. This review aims to describe the most recent progress on PTMs and their regulative factors in the context of MCC onset/development, providing an overview of current findings on both MCC subtypes. An outline of current knowledge on the potential employment of PTMs and related factors as diagnostic and prognostic markers, as well as novel treatment strategies targeting the reversibility of PTMs for MCC therapy is provided. Recent research shows that PTMs are emerging as important epigenetic players involved in MCC onset/development, and therefore may show a potential clinical significance. Deeper and integrated knowledge of currently known PTM dysregulations is of paramount importance in order to understand the molecular basis of MCC and improve the diagnosis, prognosis, and therapeutic options for this deadly tumor.
Collapse
Affiliation(s)
- Chiara Mazziotta
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy.,Center for Studies on Gender Medicine, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Carmen Lanzillotti
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy.,Center for Studies on Gender Medicine, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Roberta Gafà
- Department of Translational Medicine and for Romagna, University of Ferrara, Ferrara, Italy
| | - Antoine Touzé
- ISP "Biologie des infections à polyomavirus" Team, UMR INRA 1282, University of Tours, Tours, France
| | - Marie-Alice Durand
- ISP "Biologie des infections à polyomavirus" Team, UMR INRA 1282, University of Tours, Tours, France
| | - Fernanda Martini
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy.,Center for Studies on Gender Medicine, Department of Medical Sciences, University of Ferrara, Ferrara, Italy.,Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - John Charles Rotondo
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy.,Center for Studies on Gender Medicine, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| |
Collapse
|
48
|
Lemmerhirt JP, Isaak A, Liu R, Kock M, Daniliuc CG, Jacobson KA, Heitman LH, Junker A. Development of Bicyclo[3.1.0]hexane-Based A 3 Receptor Ligands: Closing the Gaps in the Structure-Affinity Relationships. Molecules 2022; 27:2283. [PMID: 35408685 PMCID: PMC9000336 DOI: 10.3390/molecules27072283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 03/24/2022] [Accepted: 03/30/2022] [Indexed: 11/17/2022] Open
Abstract
The adenosine A3 receptor is a promising target for treating and diagnosing inflammation and cancer. In this paper, a series of bicyclo[3.1.0]hexane-based nucleosides was synthesized and evaluated for their P1 receptor affinities in radioligand binding studies. The study focused on modifications at 1-, 2-, and 6-positions of the purine ring and variations of the 5'-position at the bicyclo[3.1.0]hexane moiety, closing existing gaps in the structure-affinity relationships. The most potent derivative 30 displayed moderate A3AR affinity (Ki of 0.38 μM) and high A3R selectivity. A subset of compounds varied at 5'-position was further evaluated in functional P2Y1R assays, displaying no off-target activity.
Collapse
Affiliation(s)
- Jan Phillip Lemmerhirt
- European Institute for Molecular Imaging (EIMI), University of Münster, Waldeyerstr. 15, 48149 Münster, Germany; (J.P.L.); (A.I.); (M.K.)
| | - Andreas Isaak
- European Institute for Molecular Imaging (EIMI), University of Münster, Waldeyerstr. 15, 48149 Münster, Germany; (J.P.L.); (A.I.); (M.K.)
| | - Rongfang Liu
- Leiden Academic Centre for Drug Research (LACDR), Division of Medicinal Chemistry, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands; (R.L.); (L.H.H.)
| | - Max Kock
- European Institute for Molecular Imaging (EIMI), University of Münster, Waldeyerstr. 15, 48149 Münster, Germany; (J.P.L.); (A.I.); (M.K.)
| | - Constantin G. Daniliuc
- Organisch-Chemisches Institut, University of Münster, Corrensstraße 40, 48149 Münster, Germany;
| | - Kenneth A. Jacobson
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA;
| | - Laura H. Heitman
- Leiden Academic Centre for Drug Research (LACDR), Division of Medicinal Chemistry, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands; (R.L.); (L.H.H.)
| | - Anna Junker
- European Institute for Molecular Imaging (EIMI), University of Münster, Waldeyerstr. 15, 48149 Münster, Germany; (J.P.L.); (A.I.); (M.K.)
| |
Collapse
|
49
|
Adenosine-Metabolizing Enzymes, Adenosine Kinase and Adenosine Deaminase, in Cancer. Biomolecules 2022; 12:biom12030418. [PMID: 35327609 PMCID: PMC8946555 DOI: 10.3390/biom12030418] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/05/2022] [Accepted: 03/06/2022] [Indexed: 12/17/2022] Open
Abstract
The immunosuppressive effect of adenosine in the microenvironment of a tumor is well established. Presently, researchers are developing approaches in immune therapy that target inhibition of adenosine or its signaling such as CD39 or CD73 inhibiting antibodies or adenosine A2A receptor antagonists. However, numerous enzymatic pathways that control ATP-adenosine balance, as well as understudied intracellular adenosine regulation, can prevent successful immunotherapy. This review contains the latest data on two adenosine-lowering enzymes: adenosine kinase (ADK) and adenosine deaminase (ADA). ADK deletes adenosine by its phosphorylation into 5′-adenosine monophosphate. Recent studies have revealed an association between a long nuclear ADK isoform and an increase in global DNA methylation, which explains epigenetic receptor-independent role of adenosine. ADA regulates the level of adenosine by converting it to inosine. The changes in the activity of ADA are detected in patients with various cancer types. The article focuses on the biological significance of these enzymes and their roles in the development of cancer. Perspectives of future studies on these enzymes in therapy for cancer are discussed.
Collapse
|
50
|
Rotondo JC, Mazziotta C, Lanzillotti C, Stefani C, Badiale G, Campione G, Martini F, Tognon M. The Role of Purinergic P2X7 Receptor in Inflammation and Cancer: Novel Molecular Insights and Clinical Applications. Cancers (Basel) 2022; 14:1116. [PMID: 35267424 PMCID: PMC8909580 DOI: 10.3390/cancers14051116] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/09/2022] [Accepted: 02/17/2022] [Indexed: 12/11/2022] Open
Abstract
The purinergic P2X7 receptor (P2X7R) is a transmembrane protein whose expression has been related to a variety of cellular processes, while its dysregulation has been linked to inflammation and cancer. P2X7R is expressed in cancer and immune system cell surfaces. ATP plays a key role in numerous metabolic processes due to its abundance in the tumour microenvironment. P2X7R plays an important role in cancer by interacting with ATP. The unusual property of P2X7R is that stimulation with low doses of ATP causes the opening of a permeable channel for sodium, potassium, and calcium ions, whereas sustained stimulation with high doses of ATP favours the formation of a non-selective pore. The latter effect induces a change in intracellular homeostasis that leads to cell death. This evidence suggests that P2X7R has both pro- and anti-tumour proprieties. P2X7R is increasingly recognised as a regulator of inflammation. In this review, we aimed to describe the most relevant characteristics of P2X7R function, activation, and its ligands, while also summarising the role of P2X7R activation in the context of inflammation and cancer. The currently used therapeutic approaches and clinical trials of P2X7R modulators are also described.
Collapse
Affiliation(s)
- John Charles Rotondo
- Laboratories of Cell Biology and Molecular Genetics, Section of Experimental Medicine, Department of Medical Sciences, School of Medicine, University of Ferrara, 44121 Ferrara, Italy; (J.C.R.); (C.M.); (C.L.); (C.S.); (G.B.); (G.C.); (F.M.)
- Centre for Studies on Gender Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Chiara Mazziotta
- Laboratories of Cell Biology and Molecular Genetics, Section of Experimental Medicine, Department of Medical Sciences, School of Medicine, University of Ferrara, 44121 Ferrara, Italy; (J.C.R.); (C.M.); (C.L.); (C.S.); (G.B.); (G.C.); (F.M.)
- Centre for Studies on Gender Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Carmen Lanzillotti
- Laboratories of Cell Biology and Molecular Genetics, Section of Experimental Medicine, Department of Medical Sciences, School of Medicine, University of Ferrara, 44121 Ferrara, Italy; (J.C.R.); (C.M.); (C.L.); (C.S.); (G.B.); (G.C.); (F.M.)
- Centre for Studies on Gender Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Chiara Stefani
- Laboratories of Cell Biology and Molecular Genetics, Section of Experimental Medicine, Department of Medical Sciences, School of Medicine, University of Ferrara, 44121 Ferrara, Italy; (J.C.R.); (C.M.); (C.L.); (C.S.); (G.B.); (G.C.); (F.M.)
| | - Giada Badiale
- Laboratories of Cell Biology and Molecular Genetics, Section of Experimental Medicine, Department of Medical Sciences, School of Medicine, University of Ferrara, 44121 Ferrara, Italy; (J.C.R.); (C.M.); (C.L.); (C.S.); (G.B.); (G.C.); (F.M.)
| | - Giulia Campione
- Laboratories of Cell Biology and Molecular Genetics, Section of Experimental Medicine, Department of Medical Sciences, School of Medicine, University of Ferrara, 44121 Ferrara, Italy; (J.C.R.); (C.M.); (C.L.); (C.S.); (G.B.); (G.C.); (F.M.)
| | - Fernanda Martini
- Laboratories of Cell Biology and Molecular Genetics, Section of Experimental Medicine, Department of Medical Sciences, School of Medicine, University of Ferrara, 44121 Ferrara, Italy; (J.C.R.); (C.M.); (C.L.); (C.S.); (G.B.); (G.C.); (F.M.)
- Centre for Studies on Gender Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
- Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy
| | - Mauro Tognon
- Laboratories of Cell Biology and Molecular Genetics, Section of Experimental Medicine, Department of Medical Sciences, School of Medicine, University of Ferrara, 44121 Ferrara, Italy; (J.C.R.); (C.M.); (C.L.); (C.S.); (G.B.); (G.C.); (F.M.)
| |
Collapse
|