1
|
Tasca CI, Zuccarini M, Di Iorio P, Ciruela F. Lessons from the physiological role of guanosine in neurodegeneration and cancer: Toward a multimodal mechanism of action? Purinergic Signal 2025; 21:133-148. [PMID: 39004650 PMCID: PMC11958862 DOI: 10.1007/s11302-024-10033-y] [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: 03/07/2024] [Accepted: 06/18/2024] [Indexed: 07/16/2024] Open
Abstract
Neurodegenerative diseases and brain tumours represent important health challenges due to their severe nature and debilitating consequences that require substantial medical care. Interestingly, these conditions share common physiological characteristics, namely increased glutamate, and adenosine transmission, which are often associated with cellular dysregulation and damage. Guanosine, an endogenous nucleoside, is safe and exerts neuroprotective effects in preclinical models of excitotoxicity, along with cytotoxic effects on tumour cells. However, the lack of well-defined mechanisms of action for guanosine hinders a comprehensive understanding of its physiological effects. In fact, the absence of specific receptors for guanosine impedes the development of structure-activity research programs to develop guanosine derivatives for therapeutic purposes. Alternatively, given its apparent interaction with the adenosinergic system, it is plausible that guanosine exerts its neuroprotective and anti-tumorigenic effects by modulating adenosine transmission through undisclosed mechanisms involving adenosine receptors, transporters, and purinergic metabolism. Here, several potential molecular mechanisms behind the protective actions of guanosine will be discussed. First, we explore its potential interaction with adenosine receptors (A1R and A2AR), including the A1R-A2AR heteromer. In addition, we consider the impact of guanosine on extracellular adenosine levels and the role of guanine-based purine-converting enzymes. Collectively, the diverse cellular functions of guanosine as neuroprotective and antiproliferative agent suggest a multimodal and complementary mechanism of action.
Collapse
Affiliation(s)
- Carla Inês Tasca
- Department of Biochemistry, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, SC, Brazil.
- Laboratory of Neurochemistry-4, Neuroscience Program/Biochemistry Program, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, SC, Brazil.
| | - Mariachiara Zuccarini
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, 66100, Chieti, Italy
- Center for Advanced Studies and Technologies (CAST), University of Chieti-Pescara, 66100, Chieti, Italy
| | - Patrizia Di Iorio
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, 66100, Chieti, Italy
- Center for Advanced Studies and Technologies (CAST), University of Chieti-Pescara, 66100, Chieti, Italy
| | - Francisco Ciruela
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, 08907 L'Hospitalet de Llobregat, Barcelona, Spain
- Neuropharmacology & Pain Group, Neuroscience Program, Bellvitge Institute for Biomedical Research, 08907L'Hospitalet de Llobregat, Bellvitge, Spain
| |
Collapse
|
2
|
Jain A, Das R, Giri M, Mane P, Shard A. Carbohydrate kinase inhibition: a promising strategy in cancer treatment. Drug Discov Today 2025; 30:104308. [PMID: 39912130 DOI: 10.1016/j.drudis.2025.104308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/21/2025] [Accepted: 01/30/2025] [Indexed: 02/07/2025]
Abstract
Carbohydrate kinases (CKs) are pivotal in various biological processes, including energy consumption, cell signaling, and biosynthesis. They are a group of enzymes that facilitate the phosphorylation of carbohydrates, playing a crucial role in cellular metabolism. These enzymes facilitate the transfer of a phosphate group from a high-energy donor like ATP to a specified location on a carbohydrate substrate. Dysregulated kinase activity drives tumor growth and progression. Inhibitors targeting these enzymes have been developed and used in cancer therapy. The CK family encompasses three major types: hexokinases, ribokinases, and phosphatidylinositol kinases, with inhibitors of paramount importance in cancer treatment. This review explores the role of CKs in cancer and its inhibitors, providing insights into improving existing inhibitors and designing new ones.
Collapse
Affiliation(s)
- Archit Jain
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Rudradip Das
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Muskan Giri
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Pranita Mane
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Amit Shard
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad, Gandhinagar, Gujarat 382355, India.
| |
Collapse
|
3
|
Cheu JWS, Chiu DKC, Kwan KKL, Yang C, Yuen VWH, Goh CC, Chui NNQ, Shen W, Law CT, Li Q, Zhang MS, Bao MHR, Wong BPY, Chan CYK, Liu CX, Sit GFW, Ooi ZY, Deng H, Tse APW, Ng IOL, Wong CCL. Hypoxia-inducible factor orchestrates adenosine metabolism to promote liver cancer development. SCIENCE ADVANCES 2023; 9:eade5111. [PMID: 37146141 PMCID: PMC10162666 DOI: 10.1126/sciadv.ade5111] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Hypoxia-induced adenosine creates an immunosuppressive tumor microenvironment (TME) and dampens the efficacy of immune checkpoint inhibitors (ICIs). We found that hypoxia-inducible factor 1 (HIF-1) orchestrates adenosine efflux through two steps in hepatocellular carcinoma (HCC). First, HIF-1 activates transcriptional repressor MXI1, which inhibits adenosine kinase (ADK), resulting in the failure of adenosine phosphorylation to adenosine monophosphate. This leads to adenosine accumulation in hypoxic cancer cells. Second, HIF-1 transcriptionally activates equilibrative nucleoside transporter 4, pumping adenosine into the interstitial space of HCC, elevating extracellular adenosine levels. Multiple in vitro assays demonstrated the immunosuppressive role of adenosine on T cells and myeloid cells. Knockout of ADK in vivo skewed intratumoral immune cells to protumorigenic and promoted tumor progression. Therapeutically, combination treatment of adenosine receptor antagonists and anti-PD-1 prolonged survival of HCC-bearing mice. We illustrated the dual role of hypoxia in establishing an adenosine-mediated immunosuppressive TME and offered a potential therapeutic approach that synergizes with ICIs in HCC.
Collapse
Affiliation(s)
- Jacinth Wing-Sum Cheu
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
- Centre for Oncology and Immunology, Hong Kong Science Park, Hong Kong
| | - David Kung-Chun Chiu
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Kenneth Kin-Leung Kwan
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
- Centre for Oncology and Immunology, Hong Kong Science Park, Hong Kong
| | - Chunxue Yang
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Vincent Wai-Hin Yuen
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
- Centre for Oncology and Immunology, Hong Kong Science Park, Hong Kong
| | - Chi Ching Goh
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Noreen Nog-Qin Chui
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Wei Shen
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
- Centre for Oncology and Immunology, Hong Kong Science Park, Hong Kong
| | - Cheuk-Ting Law
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Qidong Li
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Misty Shuo Zhang
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
- Centre for Oncology and Immunology, Hong Kong Science Park, Hong Kong
| | - Macus Hao-Ran Bao
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
- Centre for Oncology and Immunology, Hong Kong Science Park, Hong Kong
| | - Bowie Po-Yee Wong
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Cerise Yuen-Ki Chan
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
- Centre for Oncology and Immunology, Hong Kong Science Park, Hong Kong
| | - Cindy Xinqi Liu
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Grace Fu-Wan Sit
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Zher Yee Ooi
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Haijing Deng
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Aki Pui-Wah Tse
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
- Centre for Oncology and Immunology, Hong Kong Science Park, Hong Kong
| | - Irene Oi-Lin Ng
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong
| | - Carmen Chak-Lui Wong
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
- Centre for Oncology and Immunology, Hong Kong Science Park, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong
- Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen University, Guangzhou, China 510120
- Shenzhen Hospital, The University of Hong Kong, Shenzhen, China
| |
Collapse
|
4
|
Zhulai GA, Shibaev MI. Relationship between the Gene Expression of Adenosine Kinase Isoforms and the Expression of CD39 and CD73 Ectonucleotidases in Colorectal Cancer. Acta Naturae 2023; 15:42-49. [PMID: 37538807 PMCID: PMC10395772 DOI: 10.32607/actanaturae.11871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 04/03/2023] [Indexed: 08/05/2023] Open
Abstract
Tumor cells have the capacity to create an adenosine-rich immunosuppressive environment, which can interfere with antitumor immunotherapy. Approaches are currently being developed with a view to suppressing the production of adenosine or its signals. Such approaches include the use of antibodies to inhibit CD39, CD73, and adenosine-receptor antagonists. However, the abundance of enzymatic pathways that control the ATP-adenosine balance, as well as the still poorly understood intracellular adenosine regulation, makes the hoped-for success unlikely. In the present study, the enzyme adenosine kinase (ADK) needed to convert adenosine to adenosine monophosphate, thereby regulating its levels, was investigated. To do so, peripheral blood samples from patients with colorectal cancer (CRC) (n = 31) were collected with blood samples from healthy donors (n = 17) used as controls. ADK gene expression levels and those of its long (ADK-L) and short (ADK-S) isoforms were measured. The relationship between the levels of ADK gene expression and that of CD39, CD73, and A2aR genes was analyzed. It turned out that in the group of CRC patients (stages III-IV), the level of ADK-L mRNA was lower (p < 0.0011) when compared to that of the control. For the first time, an average correlation was found between the level of expression of CD39 and ADK-S (r = -0.468 at p = 0.043) and between CD73 and ADK-L (r = 0.518 at p = 0.0232) in CRC patients. Flow cytometry was used to assess the content of CD39/CD73-expressing CD8+, CD4+ and Treg lymphocytes, as well as their relationship with the level of ADK gene expression in CRC patients. But no significant correlations were found.
Collapse
Affiliation(s)
- G A Zhulai
- Institute of Biology, Karelian Research Centre, Russian Academy of Sciences, Petrozavodsk, 185910 Russian Federation
| | - M I Shibaev
- Endoscopic Department, Baranov Republican Hospital, Petrozavodsk, 185910 Russian Federation
| |
Collapse
|
5
|
Vascular Remodeling Is a Crucial Event in the Early Phase of Hepatocarcinogenesis in Rodent Models for Liver Tumorigenesis. Cells 2022; 11:cells11142129. [PMID: 35883572 PMCID: PMC9320355 DOI: 10.3390/cells11142129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/26/2022] [Accepted: 07/01/2022] [Indexed: 02/05/2023] Open
Abstract
Simple Summary Hepatocellular carcinoma (HCC) is a highly vascularized tumor and remodeling of the tumor vasculature is one of the hallmarks of tumor progression. Mouse models are elegant tools to study the onset and progression of liver tumors. However, only few data exist on the vasculature and vascular remodeling processes especially in the early phase of hepatocarcinogenesis. The aim of this study was therefore to perform a comprehensive characterization and comparison of the vasculature in mouse models used for hepatocarcinogenesis studies. For this purpose, we characterized the preneoplastic foci of cellular alteration (FCA) and hepatocellular carcinoma (HCC) by using tissue-based techniques and computer-assisted analysis to better understand if and how vascular remodeling appears in rodent models for liver tumorigenesis. Our findings demonstrated crucial differences in the number and size of the vessels, degree of maturation and intratumoral localization of the vasculature in FCA and HCC, clearly indicating that vascular remodeling is an important step in the early phase of liver tumorigenesis of rodent models. Abstract The investigation of hepatocarcinogenesis is a major field of interest in oncology research and rodent models are commonly used to unravel the pathophysiology of onset and progression of hepatocellular carcinoma. HCC is a highly vascularized tumor and vascular remodeling is one of the hallmarks of tumor progression. To date, only a few detailed data exist about the vasculature and vascular remodeling in rodent models used for hepatocarcinogenesis. In this study, the vasculature of HCC and the preneoplastic foci of alteration (FCA) of different mouse models with varying genetic backgrounds were comprehensively characterized by using immunohistochemistry (CD31, Collagen IV, αSMA, Desmin and LYVE1) and RNA in situ hybridization (VEGF-A). Computational image analysis was performed to evaluate selected parameters including microvessel density, pericyte coverage, vessel size, intratumoral vessel distribution and architecture using the Aperio ImageScope and Definiens software programs. HCC presented with a significantly lower number of vessels, but larger vessel size and increased coverage, leading to a higher degree of maturation, whereas FCA lesions presented with a higher microvessel density and a higher amount of smaller but more immature vessels. Our results clearly demonstrate that vascular remodeling is present and crucial in early stages of experimental hepatocarcinogenesis. In addition, our detailed characterization provides a strong basis for further angiogenesis studies in these experimental models.
Collapse
|
6
|
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: 50] [Impact Index Per Article: 16.7] [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
|
7
|
Rasmussen HB, Jürgens G, Thomsen R, Taboureau O, Zeth K, Hansen PE, Hansen PR. Cellular Uptake and Intracellular Phosphorylation of GS-441524: Implications for Its Effectiveness against COVID-19. Viruses 2021; 13:v13071369. [PMID: 34372575 PMCID: PMC8310262 DOI: 10.3390/v13071369] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/27/2021] [Accepted: 07/07/2021] [Indexed: 12/22/2022] Open
Abstract
GS-441524 is an adenosine analog and the parent nucleoside of the prodrug remdesivir, which has received emergency approval for treatment of COVID-19. Recently, GS-441524 has been proposed to be effective in the treatment of COVID-19, perhaps even being superior to remdesivir for treatment of this disease. Evaluation of the clinical effectiveness of GS-441524 requires understanding of its uptake and intracellular conversion to GS-441524 triphosphate, the active antiviral substance. We here discuss the potential impact of these pharmacokinetic steps of GS-441524 on the formation of its active antiviral substance and effectiveness for treatment of COVID-19. Available protein expression data suggest that several adenosine transporters are expressed at only low levels in the epithelial cells lining the alveoli in the lungs, i.e., the alveolar cells or pneumocytes from healthy lungs. This may limit uptake of GS-441524. Importantly, cellular uptake of GS-441524 may be reduced during hypoxia and inflammation due to decreased expression of adenosine transporters. Similarly, hypoxia and inflammation may lead to reduced expression of adenosine kinase, which is believed to convert GS-441524 to GS-441524 monophosphate, the perceived rate-limiting step in the intracellular formation of GS-441524 triphosphate. Moreover, increases in extracellular and intracellular levels of adenosine, which may occur during critical illnesses, has the potential to competitively decrease cellular uptake and phosphorylation of GS-441524. Taken together, tissue hypoxia and severe inflammation in COVID-19 may lead to reduced uptake and phosphorylation of GS-441524 with lowered therapeutic effectiveness as a potential outcome. Hypoxia may be particularly critical to the ability of GS-441524 to eliminate SARS-CoV-2 from tissues with low basal expression of adenosine transporters, such as alveolar cells. This knowledge may also be relevant to treatments with other antiviral adenosine analogs and anticancer adenosine analogs as well.
Collapse
Affiliation(s)
- Henrik Berg Rasmussen
- Institute of Biological Psychiatry, Mental Health Centre Sct. Hans, DK-4000 Roskilde, Denmark
- Department of Science and Environment, Roskilde University Center, DK-4000 Roskilde, Denmark; (K.Z.); (P.E.H.)
- Correspondence:
| | - Gesche Jürgens
- Clinical Pharmacology Unit, Zealand University Hospital, DK-4000 Roskilde, Denmark;
| | - Ragnar Thomsen
- Section of Forensic Chemistry, Department of Forensic Medicine, Faculty of Health Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark;
| | - Olivier Taboureau
- INSERM U1133, CNRS UMR 8251, Université de Paris, F-75013 Paris, France;
| | - Kornelius Zeth
- Department of Science and Environment, Roskilde University Center, DK-4000 Roskilde, Denmark; (K.Z.); (P.E.H.)
| | - Poul Erik Hansen
- Department of Science and Environment, Roskilde University Center, DK-4000 Roskilde, Denmark; (K.Z.); (P.E.H.)
| | - Peter Riis Hansen
- Department of Cardiology, Herlev and Gentofte Hospital, DK-2900 Hellerup, Denmark;
| |
Collapse
|
8
|
Murugan M, Fedele D, Millner D, Alharfoush E, Vegunta G, Boison D. Adenosine kinase: An epigenetic modulator in development and disease. Neurochem Int 2021; 147:105054. [PMID: 33961946 DOI: 10.1016/j.neuint.2021.105054] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 04/20/2021] [Accepted: 04/24/2021] [Indexed: 02/06/2023]
Abstract
Adenosine kinase (ADK) is the key regulator of adenosine and catalyzes the metabolism of adenosine to 5'-adenosine monophosphate. The enzyme exists in two isoforms: a long isoform (ADK-long, ADK-L) and a short isoform (ADK-short, ADK-S). The two isoforms are developmentally regulated and are differentially expressed in distinct subcellular compartments with ADK-L localized in the nucleus and ADK-S localized in the cytoplasm. The nuclear localization of ADK-L and its biochemical link to the transmethylation pathway suggest a specific role for gene regulation via epigenetic mechanisms. Recent evidence reveals an adenosine receptor-independent role of ADK in determining the global methylation status of DNA and thereby contributing to epigenomic regulation. Here we summarize recent progress in understanding the biochemical interactions between adenosine metabolism by ADK-L and epigenetic modifications linked to transmethylation reactions. This review will provide a comprehensive overview of ADK-associated changes in DNA methylation in developmental, as well as in pathological conditions including brain injury, epilepsy, vascular diseases, cancer, and diabetes. Challenges in investigating the epigenetic role of ADK for therapeutic gains are briefly discussed.
Collapse
Affiliation(s)
- Madhuvika Murugan
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA
| | - Denise Fedele
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA
| | - David Millner
- Department of Neurosurgery, New Jersey Medical School, Rutgers University, Newark, NJ 07102, USA
| | - Enmar Alharfoush
- Department of Cell Biology and Neuroscience, Rutgers University, New Brunswick, NJ 08901, USA
| | - Geetasravya Vegunta
- Department of Biology, Albert Dorman Honors College, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Detlev Boison
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA; Department of Neurosurgery, New Jersey Medical School, Rutgers University, Newark, NJ 07102, USA; Brain Health Institute, Rutgers University, Piscataway, NJ 08854, USA.
| |
Collapse
|
9
|
Boison D, Jarvis MF. Adenosine kinase: A key regulator of purinergic physiology. Biochem Pharmacol 2020; 187:114321. [PMID: 33161022 DOI: 10.1016/j.bcp.2020.114321] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/23/2020] [Accepted: 11/03/2020] [Indexed: 02/07/2023]
Abstract
Adenosine (ADO) is an essential biomolecule for life that provides critical regulation of energy utilization and homeostasis. Adenosine kinase (ADK) is an evolutionary ancient ribokinase derived from bacterial sugar kinases that is widely expressed in all forms of life, tissues and organ systems that tightly regulates intracellular and extracellular ADO concentrations. The facile ability of ADK to alter ADO availability provides a "site and event" specificity to the endogenous protective effects of ADO in situations of cellular stress. In addition to modulating the ability of ADO to activate its cognate receptors (P1 receptors), nuclear ADK isoform activity has been linked to epigenetic mechanisms based on transmethylation pathways. Previous drug discovery research has targeted ADK inhibition as a therapeutic approach to manage epilepsy, pain, and inflammation. These efforts generated multiple classes of highly potent and selective inhibitors. However, clinical development of early ADK inhibitors was stopped due to apparent mechanistic toxicity and the lack of suitable translational markers. New insights regarding the potential role of the nuclear ADK isoform (ADK-Long) in the epigenetic modulation of maladaptive DNA methylation offers the possibility of identifying novel ADK-isoform selective inhibitors and new interventional strategies that are independent of ADO receptor activation.
Collapse
Affiliation(s)
- Detlev Boison
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers University, United States.
| | | |
Collapse
|