151
|
Altered foetoplacental vascular endothelial signalling to insulin in diabesity. Mol Aspects Med 2019; 66:40-48. [DOI: 10.1016/j.mam.2019.02.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 02/19/2019] [Accepted: 02/28/2019] [Indexed: 12/26/2022]
|
152
|
Involvement of A2B adenosine receptors as anti-inflammatory in gestational diabesity. Mol Aspects Med 2019; 66:31-39. [DOI: 10.1016/j.mam.2019.01.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Revised: 11/23/2018] [Accepted: 01/17/2019] [Indexed: 02/07/2023]
|
153
|
CD73 Promotes Glioblastoma Pathogenesis and Enhances Its Chemoresistance via A 2B Adenosine Receptor Signaling. J Neurosci 2019; 39:4387-4402. [PMID: 30926752 DOI: 10.1523/jneurosci.1118-18.2019] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 03/19/2019] [Accepted: 03/22/2019] [Indexed: 12/18/2022] Open
Abstract
Glioblastoma (GB) is one of the deadliest brain cancers to afflict humans, and it has a very poor survival rate even with treatment. The extracellular adenosine-generating enzyme CD73 is involved in many cellular functions that can be usurped by tumors, including cell adhesion, proliferation, invasion, and angiogenesis. We set out to determine the role of CD73 in GB pathogenesis. To do this, we established a unique GB mouse model (CD73-FLK) in which we spatially expressed CD73 on endothelial cells in CD73-/- mice. This allowed us to elucidate the mechanism of host CD73 versus GB-expressed CD73 by comparing GB pathogenesis in WT, CD73-/-, and CD73-FLK mice. GB in CD73-/- mice had decreased tumor size, decreased tumor vessel density, and reduced tumor invasiveness compared with GB in WT mice. Interestingly, GBs in CD73-FLK mice were much more invasive and caused complete distortion of the brain morphology. We showed a 20-fold upregulation of A2B AR on GB compared with sham, and its activation induced matrix metalloproteinase-2, which enhanced GB pathogenesis. Inhibition of A2B AR signaling decreased multidrug resistance transporter protein expression, including permeability glycoprotein (P-gp) and multidrug resistance-associated protein 1 (MRP1). Further, we showed that blockade of A2B AR signaling potently increased GB cell death induced by the chemotherapeutic drug temozolomide. Together, these findings suggest that CD73 and A2B AR play a multifaceted role in GB pathogenesis and progression and that targeting the CD73-A2B AR axis can benefit GB patients and inform new approaches for therapy to treat GB patients.SIGNIFICANCE STATEMENT Glioblastoma (GB) is the most devastating primary brain tumor. GB patients' median survival is 16 months even with treatment. It is critical that we develop prophylaxes to advance GB treatment and improve patient survival. CD73-generated adenosine has been implicated in cancer pathogenesis, but its role in GB was not ascertained. Here, we demonstrated that host CD73 plays a prominent role in multiple areas of glioblastoma pathogenesis, including promoting GB growth, its angiogenesis, and its invasiveness. We found a 20-fold increase in A2B adenosine receptor (AR) expression on GB compared with sham, and its inhibition increased GB chemosensitivity to temozolomide. These findings strongly indicate that blockade or inhibition of CD73 and the A2B AR are prime targets for future GB therapy.
Collapse
|
154
|
Wan TC, Tampo A, Kwok WM, Auchampach JA. Ability of CP-532,903 to protect mouse hearts from ischemia/reperfusion injury is dependent on expression of A 3 adenosine receptors in cardiomyoyctes. Biochem Pharmacol 2019; 163:21-31. [PMID: 30710517 DOI: 10.1016/j.bcp.2019.01.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 01/28/2019] [Indexed: 10/27/2022]
Abstract
A3 adenosine receptor (A3AR) agonists are effective at limiting injury caused by ischemia/reperfusion injury of the heart in experimental animal models. However, understanding of their mechanism of action, which is likely multifactorial, remains incomplete. In prior studies, it has been demonstrated that A3AR-mediated ischemic protection is blocked by glibenclamide and is absent in Kir6.2 gene ablated mice that lack the pore-forming subunit of the ATP-sensitive potassium (KATP) channel, suggesting one contributing mechanism may involve accelerated activation of KATP channels. However, presence of A3ARs in the myocardium has yet to be established. Utilizing a whole-cell recording technique, in this study we confirm functional expression of the A3AR in adult mouse ventricular cardiomyocytes, coupled to activation of ATP-dependent potassium (KATP) channels via Gi inhibitory proteins. We further show that ischemic protection provided by the selective A3AR agonist CP-532,903 in an isolated, buffer-perfused heart model is lost completely in Adora3LoxP/LoxP;Myh6-Cre mice, which is a newly developed model developed and comprehensively described herein whereby the A3AR gene (Adora3) is deleted exclusively in cardiomyocytes. Our findings, taken together with previously published work, are consistent with the hypothesis that A3AR agonists provide ischemic tolerance, at least in part, by facilitating opening of myocardial KATP channels.
Collapse
Affiliation(s)
- Tina C Wan
- Department of Pharmacology & Toxicology, Medical College of Wisconsin, Milwaukee, WI 53226, United States; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, United States
| | - Akihito Tampo
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI 53226, United States; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, United States
| | - Wai-Meng Kwok
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI 53226, United States; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, United States
| | - John A Auchampach
- Department of Pharmacology & Toxicology, Medical College of Wisconsin, Milwaukee, WI 53226, United States; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, United States.
| |
Collapse
|
155
|
Caiazzo E, Ialenti A, Cicala C. The relatively selective cyclooxygenase-2 inhibitor nimesulide: What's going on? Eur J Pharmacol 2019; 848:105-111. [PMID: 30689999 DOI: 10.1016/j.ejphar.2019.01.044] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 01/21/2019] [Accepted: 01/24/2019] [Indexed: 12/17/2022]
Abstract
Nimesulide is a relatively selective cyclooxygenase (COX)-2 inhibitor, non-steroidal anti-inflammatory drug; it has been discovered in 1971 and firstly commercialized in Italy in 1985. There is much evidence that the pharmacological profile of nimesulide is peculiar and not shared with the other COX-2 selective inhibitors, suggesting that other molecular mechanisms besides inhibition of COX-2 derived prostaglandins are involved. Similarly, experimental data suggest that the gastrointestinal safety of nimesulide cannot be ascribed only to a COX-1 sparing effect. On the inflammatory process, the efficacy of nimesulide is dependent upon a wide spectrum of actions, due to the combination of effects on immune and non-immune cells. Early data demonstrated a central role for cyclic AMP (cAMP) in the anti-inflammatory effect of nimesulide; more recently, we have shown the involvement of the pathway ecto-5'-nucleotidase/adenosine A2A receptor. To date, the molecular mechanism(s) that confers uniqueness to nimesulide have not yet been defined. To go inside the mechanism of action of an existing drug, such as nimesulide, would be helpful to refine its therapeutic use but also to identify new targets for novel therapeutic anti-inflammatory approach. Here, we focus on accumulated evidence for a peculiar pharmacological profile of nimesulide.
Collapse
Affiliation(s)
- Elisabetta Caiazzo
- Department of Pharmacy, School of Medicine, University of Naples Federico II, via Domenico Montesano, 49, 80131 Naples, Italy
| | - Armando Ialenti
- Department of Pharmacy, School of Medicine, University of Naples Federico II, via Domenico Montesano, 49, 80131 Naples, Italy
| | - Carla Cicala
- Department of Pharmacy, School of Medicine, University of Naples Federico II, via Domenico Montesano, 49, 80131 Naples, Italy.
| |
Collapse
|
156
|
Vecchio EA, White PJ, May LT. The adenosine A 2B G protein-coupled receptor: Recent advances and therapeutic implications. Pharmacol Ther 2019; 198:20-33. [PMID: 30677476 DOI: 10.1016/j.pharmthera.2019.01.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The adenosine A2B receptor (A2BAR) is one of four adenosine receptor subtypes belonging to the Class A family of G protein-coupled receptors (GPCRs). Until recently, the A2BAR remained poorly characterised, in part due to its relatively low affinity for the endogenous agonist adenosine and therefore presumed minor physiological significance. However, the substantial increase in extracellular adenosine concentration, the sensitisation of the receptor and the upregulation of A2BAR expression under conditions of hypoxia and inflammation, suggest the A2BAR as an exciting therapeutic target in a variety of pathological disease states. Here we discuss the pharmacology of the A2BAR and outline its role in pathophysiology including ischaemia-reperfusion injury, fibrosis, inflammation and cancer.
Collapse
Affiliation(s)
- Elizabeth A Vecchio
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia; Heart Failure Pharmacology, Baker Heart & Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Paul J White
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Lauren T May
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia.
| |
Collapse
|
157
|
Jiang Z, Zhang R, Sun M, Liu Q, Wang S, Wang W, Zhao Q, Zhang H, Wang Y, Hou J, Yu B. Effect of Clopidogrel vs Ticagrelor on Platelet Aggregation and Inflammation Markers After Percutaneous Coronary Intervention for ST-Elevation Myocardial Infarction. Can J Cardiol 2018; 34:1606-1612. [DOI: 10.1016/j.cjca.2018.08.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 08/14/2018] [Accepted: 08/14/2018] [Indexed: 01/19/2023] Open
|
158
|
Li C, Yan S, He W, Yang S, Sun J, Gu N. Real-time Temperature Measurements of HMEC-1 Cells during Inflammation Production and Repair detected by Wireless Thermometry. IEEE Trans Biomed Eng 2018; 66:1898-1904. [PMID: 30418879 DOI: 10.1109/tbme.2018.2880740] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Cell inflammation process is reflected through real-time in situ cellular temperature changes. METHODS A wireless thermometry system for in situ cellular temperature measurements was used in an incubator to detect the HMEC-1 cellular temperature under lipopolysaccharide inflammation production and norepinephrine for inflammation repair. Combining the changes in cell viability, inflammatory factor levels and ATP content caused by different lipopolysaccharide or norepinephrine doses, an obvious inflammatory response and repair effect was obtained. Temperature variations were correlated with ATP content. RESULTS An obvious inflammatory response with a lipopolysaccharide concentration of 0.1 mg/L and an optimal repair effect with 1 μM norepinephrine were obtained. The relationship between temperature changes and ATP content were quite different during the production of inflammation in HMEC-1 cells, having an approximately linear relationship, while under conditions of inflammation repair in HMEC-1 cells, there was an obvious nonlinear relationship. CONCLUSION During cell damage, cell thermogenesis has a linear correlation with intracellular energy. While during cell repair, there is a gradual saturation relationship between the temperature (small range) and ATP, which may be because the thermogenesis capacity of the cell is enhanced compared to conditions during cell energy storage. Additionally, there is an optimal drug concentration for cell action during cell injury and cell repair, which is not dose-dependent. SIGNIFICANCE Whether in inflammation production or treatment, there is an optimal drug concentration. The relationship between cell thermogenesis and intracellular energy reserves is related to cell processes. Quick analysis of the energy changes in different physiological process can be realized.
Collapse
|
159
|
Poernbacher I, Vincent JP. Epithelial cells release adenosine to promote local TNF production in response to polarity disruption. Nat Commun 2018; 9:4675. [PMID: 30405122 PMCID: PMC6220285 DOI: 10.1038/s41467-018-07114-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 10/15/2018] [Indexed: 12/14/2022] Open
Abstract
Disruption of epithelial integrity contributes to chronic inflammatory disorders through persistent activation of stress signalling. Here we uncover a mechanism whereby disruption of apico-basal polarity promotes stress signalling. We show that depletion of Scribbled (Scrib), a baso-lateral determinant, causes epithelial cells to release adenosine through equilibrative channels into the extracellular space. Autocrine activation of the adenosine receptor leads to transcriptional upregulation of TNF, which in turn boosts the activity of JNK signalling. Thus, disruption of cell polarity feeds into a well-established stress pathway through the intermediary of an adenosine signalling branch. Although this regulatory input could help ensuring an effective response to acute polarity stress, we suggest that it becomes deleterious in situations of low-grade chronic disruption by provoking a private inflammatory-like TNF-driven response within the polarity-deficient epithelium.
Collapse
|
160
|
Hackett TA. Adenosine A 1 Receptor mRNA Expression by Neurons and Glia in the Auditory Forebrain. Anat Rec (Hoboken) 2018; 301:1882-1905. [PMID: 30315630 PMCID: PMC6282551 DOI: 10.1002/ar.23907] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 12/05/2017] [Accepted: 01/10/2018] [Indexed: 12/30/2022]
Abstract
In the brain, purines such as ATP and adenosine can function as neurotransmitters and co‐transmitters, or serve as signals in neuron–glial interactions. In thalamocortical (TC) projections to sensory cortex, adenosine functions as a negative regulator of glutamate release via activation of the presynaptic adenosine A1 receptor (A1R). In the auditory forebrain, restriction of A1R‐adenosine signaling in medial geniculate (MG) neurons is sufficient to extend LTP, LTD, and tonotopic map plasticity in adult mice for months beyond the critical period. Interfering with adenosine signaling in primary auditory cortex (A1) does not contribute to these forms of plasticity, suggesting regional differences in the roles of A1R‐mediated adenosine signaling in the forebrain. To advance understanding of the circuitry, in situ hybridization was used to localize neuronal and glial cell types in the auditory forebrain that express A1R transcripts (Adora1), based on co‐expression with cell‐specific markers for neuronal and glial subtypes. In A1, Adora1 transcripts were concentrated in L3/4 and L6 of glutamatergic neurons. Subpopulations of GABAergic neurons, astrocytes, oligodendrocytes, and microglia expressed lower levels of Adora1. In MG, Adora1 was expressed by glutamatergic neurons in all divisions, and subpopulations of all glial classes. The collective findings imply that A1R‐mediated signaling broadly extends to all subdivisions of auditory cortex and MG. Selective expression by neuronal and glial subpopulations suggests that experimental manipulations of A1R‐adenosine signaling could impact several cell types, depending on their location. Strategies to target Adora1 in specific cell types can be developed from the data generated here. Anat Rec, 301:1882–1905, 2018. © 2018 The Authors. The Anatomical Record published by Wiley Periodicals, Inc. on behalf of American Association of Anatomists.
Collapse
Affiliation(s)
- Troy A Hackett
- Department of Hearing and Speech Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Department of Psychology, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
161
|
Karmouty-Quintana H, Molina JG, Philip K, Bellocchi C, Gudenkauf B, Wu M, Chen NY, Collum SD, Ko J, Agarwal SK, Assassi S, Zhong H, Blackburn MR, Weng T. The Antifibrotic Effect of A 2B Adenosine Receptor Antagonism in a Mouse Model of Dermal Fibrosis. Arthritis Rheumatol 2018; 70:1673-1684. [PMID: 29771006 PMCID: PMC10077881 DOI: 10.1002/art.40554] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 05/08/2018] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Systemic sclerosis (SSc; scleroderma) is a chronic disease that affects the skin and various internal organs. Dermal fibrosis is a major component of this disease. The mechanisms that promote dermal fibrosis remain elusive. Elevations in tissue adenosine levels and the subsequent engagement of the profibrotic A2B adenosine receptor (ADORA2B) have been shown to regulate fibrosis in multiple organs including the lung, kidney, and penis; however, the role of ADORA2B in dermal fibrosis has not been investigated. We undertook this study to test our hypothesis that elevated expression of ADORA2B in the skin drives the development of dermal fibrosis. METHODS We assessed the involvement of ADORA2B in the regulation of dermal fibrosis using a well-established mouse model of dermal fibrosis. Using an orally active ADORA2B antagonist, we demonstrated how inhibition of ADORA2B results in reduced dermal fibrosis in 2 distinct experimental models. Finally, using human dermal fibroblasts, we characterized the expression of adenosine receptors. RESULTS We demonstrated that levels of ADORA2B were significantly elevated in dermal fibrosis and that the therapeutic blockade of this receptor in vivo using an ADORA2B antagonist could reduce the production of profibrotic mediators in the skin and attenuate dermal fibrosis. Antagonism of ADORA2B resulted in reduced numbers of arginase-expressing macrophages and myofibroblasts and in reduced levels of the extracellular matrix proteins fibronectin, collagen, and hyaluronan. CONCLUSION These findings identify ADORA2B as a potential profibrotic regulator in dermal fibrosis and suggest that ADORA2B antagonism may be a useful approach for the treatment of SSc.
Collapse
Affiliation(s)
| | | | | | - Chiara Bellocchi
- McGovern Medical School, Houston, Texas, Referral Center for Systemic Autoimmune Diseases, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico di Milano, Milan, Italy
| | - Brent Gudenkauf
- McGovern Medical School, Houston, Texas, Texas Tech University Health Sciences Center, Lubbock
| | | | | | | | - Junsuk Ko
- McGovern Medical School, Houston, Texas
| | | | | | | | | | | |
Collapse
|
162
|
Hypoxia-inducible factor 1-dependent expression of adenosine receptor 2B promotes breast cancer stem cell enrichment. Proc Natl Acad Sci U S A 2018; 115:E9640-E9648. [PMID: 30242135 DOI: 10.1073/pnas.1809695115] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Breast cancer stem cells (BCSCs), which are characterized by a capacity for unlimited self-renewal and for generation of the bulk cancer cell population, play a critical role in cancer relapse and metastasis. Hypoxia is a common feature of the cancer microenvironment that stimulates the specification and maintenance of BCSCs. In this study, we found that hypoxia increased expression of adenosine receptor 2B (A2BR) in human breast cancer cells through the transcriptional activity of hypoxia-inducible factor 1. The binding of adenosine to A2BR promoted BCSC enrichment by activating protein kinase C-δ, which phosphorylated and activated the transcription factor STAT3, leading to increased expression of interleukin 6 and NANOG, two key mediators of the BCSC phenotype. Genetic or pharmacological inhibition of A2BR expression or activity decreased hypoxia- or adenosine-induced BCSC enrichment in vitro, and dramatically impaired tumor initiation and lung metastasis after implantation of MDA-MB-231 human breast cancer cells into the mammary fat pad of immunodeficient mice. These data provide evidence that targeting A2BR might be an effective strategy to eradicate BCSCs.
Collapse
|
163
|
Hatakeyama H, Fujiwara T, Sato H, Terui A, Hisaka A. Investigation of Metabolomic Changes in Sunitinib-Resistant Human Renal Carcinoma 786-O Cells by Capillary Electrophoresis-Time of Flight Mass Spectrometry. Biol Pharm Bull 2018; 41:619-627. [PMID: 29607935 DOI: 10.1248/bpb.b17-00992] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Acquired resistance to sunitinib is a challenge in the treatment of renal cell carcinoma (RCC). The dysregulation of cellular metabolism is prevalent during resistance acquisition. It is known that in sunitinib-resistant RCC 786-O (786-O Res) cells sunitinib is mainly sequestered in the intracellular lysosomes. However, the relevance between sunitinib resistance and cellular metabolism has not been examined. In this study, we examined the metabolic changes in 786-O Res by using capillary electrophoresis-time of flight mass spectrometry. The cell line 786-O Res was established via persistent treatment with sunitinib, where increase in intracellular sunitinib, and sizes of lysosomes and nuclei were enhanced as compared with those in the parental 786-O (786-O Par) cells. Metabolic analyses revealed that out of the 110 metabolites examined, 13 were up-regulated and 4 were down-regulated in the 786-O Res cells. The glycolysis, tricarboxylic acid cycle and pentose phosphate pathway (PPP) were identified as being altered in the sunitinib-resistant cells, which resulted in the enhanced metabolisms of energy, nucleic acids, and glutathione redox cycle. As sunitinib was sequestered in the enlarged lysosomes in 786-O Res, the enriched energy metabolism might contribute to the maintenance of luminal pH in lysosomes via the H+ ATPase. The changes in the PPP could contribute to nuclei enlargement through up-regulation of nucleic acid biosynthesis and protect 786-O Res from cytotoxicity induced by sunitinib through up-regulation of reduced glutathione. Though the direct link between sunitinib resistance and metabolic alternation remains to be elucidated, this metabolomics study provides fundamental insights into acquisition of sunitinib resistance.
Collapse
Affiliation(s)
- Hiroto Hatakeyama
- Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University
| | - Takuya Fujiwara
- Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University
| | - Hiromi Sato
- Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University
| | - Ayu Terui
- Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University
| | - Akihiro Hisaka
- Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University
| |
Collapse
|
164
|
Bahreyni A, Avan A, Shabani M, Ryzhikov M, Fiuji H, Soleimanpour S, Khazaei M, Hassanian SM. Therapeutic potential of A2 adenosine receptor pharmacological regulators in the treatment of cardiovascular diseases, recent progress, and prospective. J Cell Physiol 2018; 234:1295-1299. [PMID: 30146778 DOI: 10.1002/jcp.27161] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Accepted: 07/10/2018] [Indexed: 12/16/2022]
Abstract
Adenosine and its analogs are of particular interest as potential therapeutic agents for treatment of cardiovascular diseases (CVDs). A2 adenosine receptor subtypes (A2a and A2b) are extensively expressed in cardiovascular system, and modulation of these receptors using A2 adenosine receptor agonists or antagonists regulates heart rate, blood pressure, heart rate variability, and cardiovascular toxicity during both normoxia and hypoxia conditions. Regulation of A2 adenosine receptor signaling via specific and novel pharmacological regulators is a potentially novel therapeutic approach for a better understanding and hence a better management of CVDs. This review summarizes the role of pharmacological A2 adenosine receptor regulators in the pathogenesis of CVDs.
Collapse
Affiliation(s)
- Amirhossein Bahreyni
- Department of Clinical Biochemistry and Immunogenetic Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.,Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Modern Sciences and Technologies, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Shabani
- Department of Medical Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mikhail Ryzhikov
- Division of Pulmonary and Critical Care Medicine, School of Medicine, Washington University, St. Louis, Missouri
| | - Hamid Fiuji
- Department of Biochemistry, Payam-e-Noor University, Mashhad, Iran
| | - Saman Soleimanpour
- Department of Microbiology and Virology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Khazaei
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Microanatomy Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
165
|
Adefegha SA, Okeke BM, Oboh G, Ijomone OM, Oyeleye SI. Modulatory effect of eugenol on arginase, nucleotidase, and adenosine deaminase activities of platelets in a carrageenan-induced arthritis rat model: A possible anti-arthritic mechanism of eugenol. Biomed Pharmacother 2018; 106:1616-1623. [PMID: 30119237 DOI: 10.1016/j.biopha.2018.07.143] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 07/24/2018] [Accepted: 07/25/2018] [Indexed: 12/20/2022] Open
Abstract
This study investigated the effect of eugenol on arginase, nucleotidase and adenosine deaminase activities in platelets of carrageenan-induced arthritic rat model to explain a possible anti-arthritic mechanism of eugenol. Fifty adult female rats (140-250 g) were divided into ten (10) groups (n = 5). Group I received oral administration of corn oil, group II received 2.50 mg/kg of eugenol, group III and IV rats received oral administration of 5.0 and 10.0 mg/kg of eugenol respectively, group V received 0.20 mg/kg of dexamethasone orally, group VI rats was injected with 1% carrageenan (arthritic rats) and received saline solution orally (arthritic control rat group), group VII, VIII and IX: arthritic rats received 2.50, 5.0 or 10 mg/kg of eugenol orally respectively, group X: arthritic rats was administered with 0.20 mg/kg of dexamethasone orally. The animals were treated for 21 days, thereafter, tibiofemoral histological examination, thiobabituric acid reactive substances level, arginase, nucleoside triphosphate diphosphohydrolase, 5´-nucleotidase and adenosine deaminase activities were assessed. Tibiofemoral histological examination result showed that infiltration of inflammatory cells was significantly decreased with an increase in eugenol dose. Activities of arginase, adenosine triphosphate and adenosine monophosphate hydrolyses were significantly decreased while adenosine diphosphate hydrolysis and adenosine deaminase activities were significantly increased in arthritic rat groups administered with different doses of eugenol. Therefore, eugenol might be a natural complement and alternative promising anti-arthritic agent. These possible anti-arthritic mechanisms may be partly through the modulation of arginase and adenosine nucleotides hydrolyzing enzyme activities as well as the antioxidative action of eugenol.
Collapse
Affiliation(s)
- Stephen Adeniyi Adefegha
- Functional Foods, Nutraceuticals and Phytomedicine Laboratory, Department of Biochemistry, Federal University of Technology, P.M.B. 704, Akure 340001, Nigeria.
| | - Bathlomew Maduka Okeke
- Functional Foods, Nutraceuticals and Phytomedicine Laboratory, Department of Biochemistry, Federal University of Technology, P.M.B. 704, Akure 340001, Nigeria
| | - Ganiyu Oboh
- Functional Foods, Nutraceuticals and Phytomedicine Laboratory, Department of Biochemistry, Federal University of Technology, P.M.B. 704, Akure 340001, Nigeria
| | - Omamuyovwi M Ijomone
- Department of Human Anatomy, Federal University of Technology, P.M.B. 704, Akure 340001, Nigeria
| | - Sunday Idowu Oyeleye
- Department of Biomedical Technology, Federal University of Technology, P.M.B. 704, Akure 340001, Nigeria
| |
Collapse
|
166
|
Moulias A, Xanthopoulou I, Alexopoulos D. Does Ticagrelor Improve Endothelial Function? J Cardiovasc Pharmacol Ther 2018; 24:11-17. [DOI: 10.1177/1074248418786936] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Ticagrelor is a P2Y12 receptor antagonist with proven clinical benefit in patients with acute coronary syndrome. Apart from its principal antiplatelet action, pleiotropic effects have been implicated in the clinical profile of ticagrelor, including a potentially beneficial impact on endothelial function. In light of the common presence and prognostic value of endothelial dysfunction in patients with coronary artery disease, several clinical studies have investigated the postulated effect of ticagrelor on endothelial function, yielding conflicting results. Limitations of the relevant studies as well as substantial differences in patient population, study design, and methods may account for these controversial findings. Most of these studies, however, support a beneficial impact of ticagrelor on endothelial function, which seems to be significant in the higher risk patients. In order to elucidate this effect, further research efforts should aim to clarify how quickly does endothelial function respond to ticagrelor, how sustained this response is during the dosing intervals and in the long term, which mechanisms are implicated, and whether this pleiotropic action is clinically significant. Future studies should include larger and diverse populations of patients, assess endothelial function at several time points after treatment initiation, and use multiple methods of endothelial function measurement, while implementing strict methodology. Nevertheless, the extent of the clinical benefit of ticagrelor attributable to actions beyond its potent and consistent antiplatelet effect remains uncertain.
Collapse
Affiliation(s)
| | | | - Dimitrios Alexopoulos
- Second Department of Cardiology, Attikon University Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| |
Collapse
|
167
|
Abstract
Extracellular adenosine nucleoside is a potent, endogenous mediator that signals through specific G protein-coupled receptors, and exerts pleiotropic effects on liver physiology, in health and disease. Particularly, adenosinergic or adenosine-mediated signaling pathways impact the progression of hepatic fibrosis, a common feature of chronic liver diseases, through regulation of matrix deposition by liver myofibroblasts. This review examines the current lines of evidence on adenosinergic regulation of liver fibrosis and myofibroblasts, identifies unanswered research questions, and proposes important future areas of investigation.
Collapse
Affiliation(s)
- Michel Fausther
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Arkansas for Medical Sciences , Little Rock, Arkansas
| |
Collapse
|
168
|
Vecchio EA, Baltos JA, Nguyen ATN, Christopoulos A, White PJ, May LT. New paradigms in adenosine receptor pharmacology: allostery, oligomerization and biased agonism. Br J Pharmacol 2018; 175:4036-4046. [PMID: 29679502 DOI: 10.1111/bph.14337] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 03/29/2018] [Accepted: 04/04/2018] [Indexed: 12/17/2022] Open
Abstract
Adenosine receptors are a family of GPCRs containing four subtypes (A1 , A2A , A2B and A3 receptors), all of which bind the ubiquitous nucleoside adenosine. These receptors play an important role in physiology and pathophysiology and therefore represent attractive drug targets for a range of conditions. The theoretical framework surrounding drug action at adenosine receptors now extends beyond the notion of prototypical agonism and antagonism to encompass more complex pharmacological concepts. New paradigms include allostery, in which ligands bind a topographically distinct receptor site from that of the endogenous agonist, homomeric or heteromeric interactions across receptor oligomers and biased agonism, that is, ligand-dependent differential intracellular signalling. This review provides a concise overview of allostery, oligomerization and biased agonism at adenosine receptors and outlines how these paradigms may enhance future drug discovery endeavours focussed on the development of novel therapeutic agents acting at adenosine receptors. LINKED ARTICLES This article is part of a themed section on Molecular Pharmacology of GPCRs. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.21/issuetoc.
Collapse
Affiliation(s)
- Elizabeth A Vecchio
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia.,Department of Pharmacology, Monash University, Melbourne, VIC, Australia
| | - Jo-Anne Baltos
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia.,Department of Pharmacology, Monash University, Melbourne, VIC, Australia
| | - Anh T N Nguyen
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia.,Department of Pharmacology, Monash University, Melbourne, VIC, Australia
| | - Arthur Christopoulos
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia.,Department of Pharmacology, Monash University, Melbourne, VIC, Australia
| | - Paul J White
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | - Lauren T May
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia.,Department of Pharmacology, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
169
|
Poppe D, Doerr J, Schneider M, Wilkens R, Steinbeck JA, Ladewig J, Tam A, Paschon DE, Gregory PD, Reik A, Müller CE, Koch P, Brüstle O. Genome Editing in Neuroepithelial Stem Cells to Generate Human Neurons with High Adenosine-Releasing Capacity. Stem Cells Transl Med 2018; 7:477-486. [PMID: 29589874 PMCID: PMC5980162 DOI: 10.1002/sctm.16-0272] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 01/29/2018] [Indexed: 12/20/2022] Open
Abstract
As a powerful regulator of cellular homeostasis and metabolism, adenosine is involved in diverse neurological processes including pain, cognition, and memory. Altered adenosine homeostasis has also been associated with several diseases such as depression, schizophrenia, or epilepsy. Based on its protective properties, adenosine has been considered as a potential therapeutic agent for various brain disorders. Since systemic application of adenosine is hampered by serious side effects such as vasodilatation and cardiac suppression, recent studies aim at improving local delivery by depots, pumps, or cell-based applications. Here, we report on the characterization of adenosine-releasing human embryonic stem cell-derived neuroepithelial stem cells (long-term self-renewing neuroepithelial stem [lt-NES] cells) generated by zinc finger nuclease (ZFN)-mediated knockout of the adenosine kinase (ADK) gene. ADK-deficient lt-NES cells and their differentiated neuronal and astroglial progeny exhibit substantially elevated release of adenosine compared to control cells. Importantly, extensive adenosine release could be triggered by excitation of differentiated neuronal cultures, suggesting a potential activity-dependent regulation of adenosine supply. Thus, ZFN-modified neural stem cells might serve as a useful vehicle for the activity-dependent local therapeutic delivery of adenosine into the central nervous system. Stem Cells Translational Medicine 2018;7:477-486.
Collapse
Affiliation(s)
- Daniel Poppe
- Institute of Reconstructive Neurobiology, University of Bonn and Hertie FoundationBonnGermany
| | - Jonas Doerr
- Institute of Reconstructive Neurobiology, University of Bonn and Hertie FoundationBonnGermany
| | - Marion Schneider
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Chemistry I, University of BonnBonnGermany
| | - Ruven Wilkens
- Institute of Reconstructive Neurobiology, University of Bonn and Hertie FoundationBonnGermany
| | - Julius A. Steinbeck
- Institute of Reconstructive Neurobiology, University of Bonn and Hertie FoundationBonnGermany
| | - Julia Ladewig
- Institute of Reconstructive Neurobiology, University of Bonn and Hertie FoundationBonnGermany
- Central Institute of Mental Health, University of Heidelberg/Medical Faculty MannheimMannheimGermany
- Hector Institute for Translational Brain Research (HITBR gGmbH)MannheimGermany
- German Cancer Research Center (DKFZ)HeidelbergGermany
| | | | | | | | | | - Christa E. Müller
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Chemistry I, University of BonnBonnGermany
| | - Philipp Koch
- Institute of Reconstructive Neurobiology, University of Bonn and Hertie FoundationBonnGermany
- Central Institute of Mental Health, University of Heidelberg/Medical Faculty MannheimMannheimGermany
- Hector Institute for Translational Brain Research (HITBR gGmbH)MannheimGermany
- German Cancer Research Center (DKFZ)HeidelbergGermany
| | - Oliver Brüstle
- Institute of Reconstructive Neurobiology, University of Bonn and Hertie FoundationBonnGermany
| |
Collapse
|
170
|
Mertens TCJ, Hanmandlu A, Tu L, Phan C, Collum SD, Chen NY, Weng T, Davies J, Liu C, Eltzschig HK, Jyothula SSK, Rajagopal K, Xia Y, Guha A, Bruckner BA, Blackburn MR, Guignabert C, Karmouty-Quintana H. Switching-Off Adora2b in Vascular Smooth Muscle Cells Halts the Development of Pulmonary Hypertension. Front Physiol 2018; 9:555. [PMID: 29910735 PMCID: PMC5992271 DOI: 10.3389/fphys.2018.00555] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 04/30/2018] [Indexed: 11/26/2022] Open
Abstract
Background: Pulmonary hypertension (PH) is a devastating and progressive disease characterized by excessive proliferation of pulmonary artery smooth muscle cells (PASMCs) and remodeling of the lung vasculature. Adenosine signaling through the ADORA2B receptor has previously been implicated in disease progression and tissue remodeling in chronic lung disease. In experimental models of PH associated with chronic lung injury, pharmacological or genetic inhibition of ADORA2B improved markers of chronic lung injury and hallmarks of PH. However, the contribution of ADORA2B expression in the PASMC was not fully evaluated. Hypothesis: We hypothesized that adenosine signaling through the ADORA2B receptor in PASMC mediates the development of PH. Methods: PASMCs from controls and patients with idiopathic pulmonary arterial hypertension (iPAH) were characterized for expression levels of all adenosine receptors. Next, we evaluated the development of PH in ADORA2Bf/f-Transgelin (Tagln)cre mice. These mice or adequate controls were exposed to a combination of SUGEN (SU5416, 20 mg/kg/b.w. IP) and hypoxia (10% O2) for 28 days (HX-SU) or to chronic low doses of bleomycin (BLM, 0.035U/kg/b.w. IP). Cardiovascular readouts including right ventricle systolic pressures (RVSPs), Fulton indices and vascular remodeling were determined. Using PASMCs we identified ADORA2B-dependent mediators involved in vascular remodeling. These mediators: IL-6, hyaluronan synthase 2 (HAS2) and tissue transglutaminase (Tgm2) were determined by RT-PCR and validated in our HX-SU and BLM models. Results: Increased levels of ADORA2B were observed in PASMC from iPAH patients. ADORA2Bf/f-Taglncre mice were protected from the development of PH following HX-SU or BLM exposure. In the BLM model of PH, ADORA2Bf/f- Taglncre mice were not protected from the development of fibrosis. Increased expression of IL-6, HAS2 and Tgm2 was observed in PASMC in an ADORA2B-dependent manner. These mediators were also reduced in ADORA2Bf/f- Taglncre mice exposed to HX-SU or BLM. Conclusions: Our studies revealed ADORA2B-dependent increased levels of IL-6, hyaluronan and Tgm2 in PASMC, consistent with reduced levels in ADORA2Bf/f- Taglncre mice exposed to HX-SU or BLM. Taken together, our data indicates that ADORA2B on PASMC mediates the development of PH through the induction of IL-6, hyaluronan and Tgm2. These studies point at ADORA2B as a therapeutic target to treat PH.
Collapse
Affiliation(s)
- Tinne C J Mertens
- Department of Biochemistry and Molecular Biology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Ankit Hanmandlu
- Department of Biochemistry and Molecular Biology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Ly Tu
- Institut National de la Santé et de la Recherche Médicale UMR_S 999, Le Plessis-Robinson, France.,Université Paris-Sud and Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Carole Phan
- Institut National de la Santé et de la Recherche Médicale UMR_S 999, Le Plessis-Robinson, France.,Université Paris-Sud and Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Scott D Collum
- Department of Biochemistry and Molecular Biology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Ning-Yuan Chen
- Department of Biochemistry and Molecular Biology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Tingting Weng
- Department of Biochemistry and Molecular Biology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Jonathan Davies
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
| | - Chen Liu
- Department of Biochemistry and Molecular Biology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Holger K Eltzschig
- Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Soma S K Jyothula
- Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Keshava Rajagopal
- Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Yang Xia
- Department of Biochemistry and Molecular Biology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Ashrith Guha
- Methodist Debakey Heart and Vascular Center, Houston Methodist Hospital, Houston, TX, United States
| | - Brian A Bruckner
- Methodist Debakey Heart and Vascular Center, Houston Methodist Hospital, Houston, TX, United States
| | - Michael R Blackburn
- Department of Biochemistry and Molecular Biology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Christophe Guignabert
- Institut National de la Santé et de la Recherche Médicale UMR_S 999, Le Plessis-Robinson, France.,Université Paris-Sud and Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Harry Karmouty-Quintana
- Department of Biochemistry and Molecular Biology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
171
|
Rivera-Oliver M, Moreno E, Álvarez-Bagnarol Y, Ayala-Santiago C, Cruz-Reyes N, Molina-Castro GC, Clemens S, Canela EI, Ferré S, Casadó V, Díaz-Ríos M. Adenosine A 1-Dopamine D 1 Receptor Heteromers Control the Excitability of the Spinal Motoneuron. Mol Neurobiol 2018; 56:797-811. [PMID: 29797183 DOI: 10.1007/s12035-018-1120-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 05/11/2018] [Indexed: 10/16/2022]
Abstract
While the role of the ascending dopaminergic system in brain function and dysfunction has been a subject of extensive research, the role of the descending dopaminergic system in spinal cord function and dysfunction is just beginning to be understood. Adenosine plays a key role in the inhibitory control of the ascending dopaminergic system, largely dependent on functional complexes of specific subtypes of adenosine and dopamine receptors. Combining a selective destabilizing peptide strategy with a proximity ligation assay and patch-clamp electrophysiology in slices from male mouse lumbar spinal cord, the present study demonstrates the existence of adenosine A1-dopamine D1 receptor heteromers in the spinal motoneuron by which adenosine tonically inhibits D1 receptor-mediated signaling. A1-D1 receptor heteromers play a significant control of the motoneuron excitability, represent main targets for the excitatory effects of caffeine in the spinal cord and can constitute new targets for the pharmacological therapy after spinal cord injury, motor aging-associated disorders and restless legs syndrome.
Collapse
Affiliation(s)
- Marla Rivera-Oliver
- Department of Anatomy and Neurobiology and Institute of Neurobiology, University of Puerto Rico, Medical Sciences, Rio Piedras and Cayey Campuses, San Juan, 00936, Puerto Rico
| | - Estefanía Moreno
- Center for Biomedical Research in Neurodegenerative Diseases Network (CIBERNED) and Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Institute of Biomedicine of the University of Barcelona, University of Barcelona, 08028, Barcelona, Spain
| | - Yocasta Álvarez-Bagnarol
- Department of Anatomy and Neurobiology and Institute of Neurobiology, University of Puerto Rico, Medical Sciences, Rio Piedras and Cayey Campuses, San Juan, 00936, Puerto Rico
| | - Christian Ayala-Santiago
- Department of Anatomy and Neurobiology and Institute of Neurobiology, University of Puerto Rico, Medical Sciences, Rio Piedras and Cayey Campuses, San Juan, 00936, Puerto Rico
| | - Nicole Cruz-Reyes
- Department of Anatomy and Neurobiology and Institute of Neurobiology, University of Puerto Rico, Medical Sciences, Rio Piedras and Cayey Campuses, San Juan, 00936, Puerto Rico
| | - Gian Carlo Molina-Castro
- Department of Anatomy and Neurobiology and Institute of Neurobiology, University of Puerto Rico, Medical Sciences, Rio Piedras and Cayey Campuses, San Juan, 00936, Puerto Rico
| | - Stefan Clemens
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA
| | - Enric I Canela
- Center for Biomedical Research in Neurodegenerative Diseases Network (CIBERNED) and Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Institute of Biomedicine of the University of Barcelona, University of Barcelona, 08028, Barcelona, Spain
| | - Sergi Ferré
- Integrative Neurobiology Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Triad Technology Building, 333 Cassell Drive, Baltimore, MD, 21224, USA.
| | - Vicent Casadó
- Center for Biomedical Research in Neurodegenerative Diseases Network (CIBERNED) and Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Institute of Biomedicine of the University of Barcelona, University of Barcelona, 08028, Barcelona, Spain
| | - Manuel Díaz-Ríos
- Department of Anatomy and Neurobiology and Institute of Neurobiology, University of Puerto Rico, Medical Sciences, Rio Piedras and Cayey Campuses, San Juan, 00936, Puerto Rico
| |
Collapse
|
172
|
Beach KM, Hung LF, Arumugam B, Smith EL, Ostrin LA. Adenosine receptor distribution in Rhesus monkey ocular tissue. Exp Eye Res 2018; 174:40-50. [PMID: 29792846 DOI: 10.1016/j.exer.2018.05.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 04/26/2018] [Accepted: 05/20/2018] [Indexed: 02/06/2023]
Abstract
Adenosine receptor (ADOR) antagonists, such as 7-methylxanthine (7-MX), have been shown to slow myopia progression in humans and animal models. Adenosine receptors are found throughout the body, and regulate the release of neurotransmitters such as dopamine and glutamate. However, the role of adenosine in eye growth is unclear. Evidence suggests that 7-MX increases scleral collagen fibril diameter, hence preventing axial elongation. This study used immunohistochemistry (IHC) and reverse-transcription quantitative polymerase chain reaction (RT-qPCR) to examine the distribution of the four ADORs in the normal monkey eye to help elucidate potential mechanisms of action. Eyes were enucleated from six Rhesus monkeys. Anterior segments and eyecups were separated into components and flash-frozen for RNA extraction or fixed in 4% paraformaldehyde and processed for immunohistochemistry against ADORA1, ADORA2a, ADORA2b, and ADORA3. RNA was reverse-transcribed, and qPCR was performed using custom primers. Relative gene expression was calculated using the ΔΔCt method normalizing to liver expression, and statistical analysis was performed using Relative Expression Software Tool. ADORA1 immunostaining was highest in the iris sphincter muscle, trabecular meshwork, ciliary epithelium, and retinal nerve fiber layer. ADORA2a immunostaining was highest in the corneal epithelium, trabecular meshwork, ciliary epithelium, retinal nerve fiber layer, and scleral fibroblasts. ADORA2b immunostaining was highest in corneal basal epithelium, limbal stem cells, iris sphincter, ciliary muscle, ciliary epithelium, choroid, isolated retinal ganglion cells and scattered scleral fibroblasts. ADORA3 immunostaining was highest in the iris sphincter, ciliary muscle, ciliary epithelium, choroid, isolated retinal ganglion cells, and scleral fibroblasts. Compared to liver mRNA, ADORA1 mRNA was significantly higher in the brain, retina and choroid, and significantly lower in the iris/ciliary body. ADORA2a expression was higher in brain and retina, ADORA2b expression was higher in retina, and ADORA3 was higher in the choroid. In conclusion, immunohistochemistry and RT-qPCR indicated differential patterns of expression of the four adenosine receptors in the ocular tissues of the normal non-human primate. The presence of ADORs in scleral fibroblasts and the choroid may support mechanisms by which ADOR antagonists prevent myopia. The potential effects of ADOR inhibition on both anterior and posterior ocular structures warrant investigation.
Collapse
Affiliation(s)
- Krista M Beach
- University of Houston College of Optometry, 4901 Calhoun Rd, Houston, TX 77204, USA
| | - Li-Fang Hung
- University of Houston College of Optometry, 4901 Calhoun Rd, Houston, TX 77204, USA
| | - Baskar Arumugam
- University of Houston College of Optometry, 4901 Calhoun Rd, Houston, TX 77204, USA
| | - Earl L Smith
- University of Houston College of Optometry, 4901 Calhoun Rd, Houston, TX 77204, USA
| | - Lisa A Ostrin
- University of Houston College of Optometry, 4901 Calhoun Rd, Houston, TX 77204, USA.
| |
Collapse
|
173
|
Sung Y, Spagou K, Kafeza M, Kyriakides M, Dharmarajah B, Shalhoub J, Diaz JA, Wakefield TW, Holmes E, Davies AH. Deep Vein Thrombosis Exhibits Characteristic Serum and Vein Wall Metabolic Phenotypes in the Inferior Vena Cava Ligation Mouse Model. Eur J Vasc Endovasc Surg 2018. [DOI: 10.1016/j.ejvs.2018.01.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
174
|
Critical roles of TRPV2 channels, histamine H1 and adenosine A1 receptors in the initiation of acupoint signals for acupuncture analgesia. Sci Rep 2018; 8:6523. [PMID: 29695862 PMCID: PMC5916903 DOI: 10.1038/s41598-018-24654-y] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 04/03/2018] [Indexed: 12/31/2022] Open
Abstract
Acupuncture is one of the most promising modalities in complimentary medicine. However, the underlying mechanisms are not well understood yet. We found that in TRPV2 knockout male mice, acupuncture-induced analgesia was suppressed with a decreased activation of mast cells in the acupoints stimulated. The mast cell stabilizer sodium cromolyn could suppress the release of adenosine in the acupoints on male rats. A direct injection of adenosine A1 receptor agonist or histamine H1 receptor agonist increased β-endorphin in the cerebral-spinal fluid in the acute adjuvant arthritis male rats and thus replicated the analgesic effect of acupuncture. These observations suggest that the mast cell is the central structure of acupoints and is activated by acupuncture through TRPV2 channels. The mast cell transduces the mechanical stimuli to acupuncture signal by activating either H1 or A1 receptors, therefore triggering the acupuncture effect in the subject. These findings might open new frontiers for acupuncture research.
Collapse
|
175
|
Adenosine pretreatment attenuates angiotensin II-mediated p38 MAPK activation in a protein kinase A dependent manner. ASIAN BIOMED 2018. [DOI: 10.2478/abm-2010-0094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Abstract
Background: Adenosine is known as a protective and anti-inflammatory nucleoside. Angiotensin II is the main hormone of the renin-angiotensin system. It is associated with endothelial permeability, recruitment, and activation of the immune cells through induction of inflammatory mediators. Matrix metalloproteinase-9 (MMP-9) plays an important role in inflammatory processes mediated by macrophages. Objectives: Investigate whether adenosine pretreatment modulates angiotensin II-induced MMP-9 expression and activation of signaling molecules. Methods: Human monocytic U-937 cells were treated with either adenosine or angiotensin II alone or angiotensin II following a pretreatment with adenosine. Supernatants were analyzed for MMP-9 activity by zymography method. MMP-9 gene expression was analyzed using real-time PCR. Activation of inflammatory mediators IκB-α, NF-κB, JNK, p38 MAPK, and STAT3 were analyzed by a multi-target ELISA kit. Association of Protein kinase A (PKA) in adenosine effects was studied by pre-incubation with H89, a selective PKA inhibitor. Results: Treatment of the cells with angiotensin II significantly increased MMP-9 production (p <0.05). Adenosine pretreatment did not attenuate this angiotensin II effect. Angiotensin II treatment induced NF-κB, JNK and p38 activation. Pretreatment with adenosine prior to angiotensin II stimulation showed a 40% inhibitory effect on p38 induction (p <0.05). This effect was reversed by PKA inhibition. Conclusion: The present data confirmed that monocytic MMP-9 was a target gene for angiotensin II. Adenosine pretreatment did not inhibit MMP-9 increase in response to angiotensin II. However, it showed a potential inhibitory effect on angiotensin II inflammatory signaling.
Collapse
|
176
|
Baldissera MD, Souza CF, Bottari NB, Verdi CM, Santos RCV, Vizzotto BS, Baldisserotto B. Purinergic signalling displays an anti-inflammatory profile in the spleen of fish experimentally infected with Aeromonas caviae: Modulation of the immune response. JOURNAL OF FISH DISEASES 2018; 41:683-687. [PMID: 29265378 DOI: 10.1111/jfd.12773] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 11/24/2017] [Indexed: 06/07/2023]
Abstract
Extracellular adenosine triphosphate (ATP) and its metabolite adenosine (Ado) are recognized as key mediators of immune and inflammatory responses. Depending on its concentration, ATP may act as an immunostimulant or immunodepressant, while Ado levels display an anti-inflammatory profile. The aim of this study was to evaluate whether splenic purinergic signalling is capable of modulating immune and inflammatory responses in fish experimentally infected with Aeromonas caviae. Triphosphate diphosphohydrolase (NTPDase) and 5'-nucleotidase activities increased in the spleen of silver catfish (Rhamdia quelen) experimentally infected with A. caviae compared with the uninfected control group. Moreover, splenic Ado levels increased in the infected animals relative to the uninfected control group. Based on these lines of evidence, our findings revealed that adenine nucleotide hydrolysis is modified in the spleen of fish infected with A. caviae attempting to restrict the inflammatory process through the upregulation of NTPDase and 5'-nucleotidase activities, which occurs in an attempt to hydrolyse the excessive ATP in the extracellular environment and rapidly hydrolyse AMP to form Ado. In summary, purinergic signalling can modulate immune and inflammatory responses during A. caviae infection.
Collapse
Affiliation(s)
- M D Baldissera
- Department of Microbiology and Parasitology, Universidade Federal de Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - C F Souza
- Department of Physiology and Pharmacology, Universidade Federal de Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - N B Bottari
- Department of Biochemistry and Molecular Biology, Universidade Federal de Santa Maria (UFSM), Santa Maria, Rio Grande do Sul, Brazil
| | - C M Verdi
- Department of Microbiology and Parasitology, Universidade Federal de Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - R C V Santos
- Department of Microbiology and Parasitology, Universidade Federal de Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - B S Vizzotto
- Laboratory of Molecular Biology, Centro Universitário Franciscano, Santa Maria, Rio Grande do Sul, Brazil
| | - B Baldisserotto
- Department of Physiology and Pharmacology, Universidade Federal de Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| |
Collapse
|
177
|
Ankylosing spondylitis monocyte-derived macrophages express increased level of A2A adenosine receptor and decreased level of ectonucleoside triphosphate diphosphohydrolase-1 (CD39), A1 and A2B adenosine receptors. Clin Rheumatol 2018. [DOI: 10.1007/s10067-018-4055-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
178
|
Chen PZ, He WJ, Zhu ZR, E GJ, Xu G, Chen DW, Gao YQ. Adenosine A 2A receptor involves in neuroinflammation-mediated cognitive decline through activating microglia under acute hypobaric hypoxia. Behav Brain Res 2018; 347:99-107. [PMID: 29501623 DOI: 10.1016/j.bbr.2018.02.038] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 02/26/2018] [Accepted: 02/26/2018] [Indexed: 01/05/2023]
Abstract
Hypobaric hypoxia (HH) at high altitudes leads to a wide range of cognitive impairments which can handicap human normal activities and performances. However, the underlying mechanism is still unclear. Adenosine A2A receptors (A2ARs) of the brain are pivotal to synaptic plasticity and cognition. Besides, insult-induced up-regulation of A2AR regulates neuroinflammation and therefore induces brain damages in various neuropathological processes. The present study was designed to determine whether A2AR-mediate neuroinflammation involves in cognitive impairments under acute HH. A2AR knock-out and wild-type male mice were exposed to a simulated altitude of 8000 m for 7 consecutive days in a hypobaric chamber and simultaneously received behavioral tests including Morris water maze test and open filed test. A2AR expression, the activation of microglia and the production of TNF-α were evaluated in the hippocampus by immunohistochemistry and ELISA, respectively. Behavioral tests showed that acute HH exposure caused the dysfunction of spatial memory and mood, while genetic inactivation of A2AR attenuated the impairment of spatial memory but not that of mood. Double-labeled immunofluorescence showed that A2ARs were mainly expressed on microglia and up-regulated in the hippocampus of acute HH model mice. Acute HH also induced the accumulation of microglia and increased production of TNF-α in the hippocampus, which could be markedly inhibited by A2AR inactivation. These findings indicate that microglia-mediated neuroinflammation triggered by A2AR activation involves in acute HH-induced spatial memory impairment and that A2AR could be a new target for the pharmacotherapy of cognitive dysfunction at high altitudes.
Collapse
Affiliation(s)
- Peng-Zhi Chen
- Institute of Medicine and Hygienic Equipment for High Altitude Region, College of High Altitude Military Medicine, Third Military Medical University, Chongqing, China; Key Laboratory of High Altitude Environmental Medicine, Third Military Medical University, Ministry of Education, Chongqing, China; Key Laboratory of High Altitude Medicine, PLA, Chongqing, China
| | - Wen-Juan He
- Department of Pathophysiology and High Altitude Pathology, College of High Altitude Military Medicine, Third Military Medical University, Chongqing, China
| | - Zhi-Ru Zhu
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing, China
| | - Guo-Ji E
- Institute of Medicine and Hygienic Equipment for High Altitude Region, College of High Altitude Military Medicine, Third Military Medical University, Chongqing, China; Key Laboratory of High Altitude Environmental Medicine, Third Military Medical University, Ministry of Education, Chongqing, China; Key Laboratory of High Altitude Medicine, PLA, Chongqing, China
| | - Gang Xu
- Institute of Medicine and Hygienic Equipment for High Altitude Region, College of High Altitude Military Medicine, Third Military Medical University, Chongqing, China; Key Laboratory of High Altitude Environmental Medicine, Third Military Medical University, Ministry of Education, Chongqing, China; Key Laboratory of High Altitude Medicine, PLA, Chongqing, China
| | - De-Wei Chen
- Department of Pathophysiology and High Altitude Pathology, College of High Altitude Military Medicine, Third Military Medical University, Chongqing, China
| | - Yu-Qi Gao
- Institute of Medicine and Hygienic Equipment for High Altitude Region, College of High Altitude Military Medicine, Third Military Medical University, Chongqing, China; Key Laboratory of High Altitude Environmental Medicine, Third Military Medical University, Ministry of Education, Chongqing, China; Key Laboratory of High Altitude Medicine, PLA, Chongqing, China.
| |
Collapse
|
179
|
Costa MA, Paiva AE, Andreotti JP, Cardoso MV, Cardoso CD, Mintz A, Birbrair A. Pericytes constrict blood vessels after myocardial ischemia. J Mol Cell Cardiol 2018; 116:1-4. [PMID: 29371134 PMCID: PMC6089363 DOI: 10.1016/j.yjmcc.2018.01.014] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 01/21/2018] [Indexed: 12/22/2022]
Abstract
No-reflow phenomenon is defined as the reduced blood flow after myocardial ischemia. If prolonged it leads to profound damages in the myocardium. The lack of a detailed knowledge about the cells mediating no-reflow restricts the design of effective therapies. Recently, O'Farrell et al. (2017) by using state-of-the-art technologies, including high-resolution confocal imaging in combination with myocardial ischemia/reperfusion mouse model, reveal that pericytes contribute to the no-reflow phenomenon post-ischemia in the heart. Strikingly, intravenous adenosine increased vascular diameter at pericyte site after cardiac ischemia. This study provides a novel therapeutic target to inhibit no-reflow phenomenon after myocardial ischemia.
Collapse
Affiliation(s)
- Matheus A Costa
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ana E Paiva
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Julia P Andreotti
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Marcus V Cardoso
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Carlos D Cardoso
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Akiva Mintz
- Department of Radiology, Columbia University Medical Center, New York, NY, USA
| | - Alexander Birbrair
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil; Department of Radiology, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
180
|
Baeta-Corral R, Johansson B, Giménez-Llort L. Long-term Treatment with Low-Dose Caffeine Worsens BPSD-Like Profile in 3xTg-AD Mice Model of Alzheimer's Disease and Affects Mice with Normal Aging. Front Pharmacol 2018; 9:79. [PMID: 29497377 PMCID: PMC5818407 DOI: 10.3389/fphar.2018.00079] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 01/24/2018] [Indexed: 12/26/2022] Open
Abstract
Coffee or caffeine has recently been suggested as prophylaxis for dementia. Although memory problems are hallmarks of Alzheimer's disease, this dementia is also characterized by neuropsychiatric symptoms called Behavioral and Psychological Symptoms of Dementia (BPSD). The impact of preventive/therapeutic strategies on both cognitive and non-cognitive symptoms can be addressed in the 3xTg-AD mice, since they exhibit cognitive but also BPSD-like profiles. Here, we studied the long-term effects of a low dose of caffeine in male 3xTg-AD mice and as compared to age-matched non-transgenic (NTg) counterparts with normal aging. Animals were treated (water or caffeine in drinking water) from adulthood (6 months of age) until middle-aged (13 months of age), that in 3xTg-AD mice correspond to onset of cognitive impairment and advanced stages, respectively. The low caffeine dosing used (0.3 mg/ml) was previously found to give a plasma concentration profile in mice roughly equivalent to that of a human coffee drinker. There were significant effects of caffeine on most behavioral variables, especially those related to neophobia and other anxiety-like behaviors, emotionality, and cognitive flexibility. The 3xTg-AD and NTg mice were differently influenced by caffeine. Overall, the increase of neophobia and other anxiety-related behaviors resulted in an exacerbation of BPSD-like profile in 3xTg-AD mice. Learning and memory, strongly influenced by anxiety in 3xTg-AD mice, got little benefit from caffeine, only shown after a detailed analysis of navigation strategies. The worsened pattern in NTg mice and the use of search strategies in 3xTg-AD mice make both groups more similar. Circadian motor activity showed genotype differences, which were found to be enhanced by caffeine. Selective effects of caffeine on NTg were found in the modulation of behaviors related to emotional profile and risk assessment. Caffeine normalized splenomegaly of 3xTg-AD mice, a physical indicator of their impaired peripheral immune system, and trended to increase their corticosterone levels. Our observations of adverse caffeine effects in an Alzheimer's disease model together with previous clinical observations suggest that an exacerbation of BPSD-like symptoms may partly interfere with the beneficial cognitive effects of caffeine. These results are relevant when coffee-derived new potential treatments for dementia are to be devised and tested.
Collapse
Affiliation(s)
- Raquel Baeta-Corral
- Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Psychiatry and Forensic Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Björn Johansson
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Solna, Sweden
- Department of Geriatrics, Karolinska University Hospital, Solna, Sweden
| | - Lydia Giménez-Llort
- Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Psychiatry and Forensic Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
181
|
Garg C, Seo JH, Ramachandran J, Loh JM, Calderon F, Contreras JE. Trovafloxacin attenuates neuroinflammation and improves outcome after traumatic brain injury in mice. J Neuroinflammation 2018; 15:42. [PMID: 29439712 PMCID: PMC5812039 DOI: 10.1186/s12974-018-1069-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 01/17/2018] [Indexed: 12/20/2022] Open
Abstract
Background Trovafloxacin is a broad-spectrum antibiotic, recently identified as an inhibitor of pannexin-1 (Panx1) channels. Panx1 channels are important conduits for the adenosine triphosphate (ATP) release from live and dying cells that enhances the inflammatory response of immune cells. Elevated extracellular levels ATP released upon injury activate purinergic pathways in inflammatory cells that promote migration, proliferation, phagocytosis, and apoptotic signals. Here, we tested whether trovafloxacin administration attenuates the neuroinflammatory response and improves outcomes after brain trauma. Methods The murine controlled cortical impact (CCI) model was used to determine whether in vivo delivery of trovafloxacin has anti-inflammatory and neuroprotective actions after brain trauma. Locomotor deficit was assessed using the rotarod test. Levels of tissue damage markers and inflammation were measured using western blot, qPCR, and immunofluorescence. In vitro assays were used to evaluate whether trovafloxacin blocks ATP release and cell migration in a chemotactic-stimulated microglia cell line. Results Trovafloxacin treatment of CCI-injured mice significantly reduced tissue damage markers and improved locomotor deficits. In addition, trovafloxacin treatment significantly reduced mRNA levels of several pro-inflammatory cytokines (IL-1β, IL-6, and TNF-α), which correlates with an overall reduction in the accumulation of inflammatory cell types (neutrophils, microglia/macrophages, and astroglia) at the injury zone. To determine whether trovafloxacin exerted these effects by direct action on immune cells, we evaluated its effect on ATP release and cell migration using a chemotactic-stimulated microglial cell line. We found that trovafloxacin significantly inhibited both ATP release and migration of these cells. Conclusion Our results show that trovafloxacin administration has pronounced anti-inflammatory and neuroprotective effects following brain injury. These findings lay the foundation for future studies to directly test a role for Panx1 channels in pathological inflammation following brain trauma. Electronic supplementary material The online version of this article (10.1186/s12974-018-1069-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Charu Garg
- Department of Pharmacology, Physiology and Neurosciences, New Jersey Medical School, Rutgers University, 185 South Orange Ave, Newark, NJ, 07103, USA
| | - Joon Ho Seo
- Department of Pharmacology, Physiology and Neurosciences, New Jersey Medical School, Rutgers University, 185 South Orange Ave, Newark, NJ, 07103, USA
| | - Jayalakshmi Ramachandran
- Department of Pharmacology, Physiology and Neurosciences, New Jersey Medical School, Rutgers University, 185 South Orange Ave, Newark, NJ, 07103, USA
| | - Ji Meng Loh
- Department of Mathematical Sciences, New Jersey Institute of Technology, University Heights, Newark, NJ, 07102, USA
| | - Frances Calderon
- Department of Pharmacology, Physiology and Neurosciences, New Jersey Medical School, Rutgers University, 185 South Orange Ave, Newark, NJ, 07103, USA.
| | - Jorge E Contreras
- Department of Pharmacology, Physiology and Neurosciences, New Jersey Medical School, Rutgers University, 185 South Orange Ave, Newark, NJ, 07103, USA.
| |
Collapse
|
182
|
Ialenti A, Caiazzo E, Morello S, Carnuccio R, Cicala C. Adenosine A 2A Receptor Agonist, 2- p-(2-Carboxyethyl)phenethylamino-5'- N-ethylcarboxamidoadenosine Hydrochloride Hydrate, Inhibits Inflammation and Increases Fibroblast Growth Factor-2 Tissue Expression in Carrageenan-Induced Rat Paw Edema. J Pharmacol Exp Ther 2018; 364:221-228. [PMID: 29212832 DOI: 10.1124/jpet.117.244319] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 11/27/2017] [Indexed: 03/08/2025] Open
Abstract
Adenosine is the final product of ATP metabolism, mainly derived from the action of 5'-nucleotidase cleavage of AMP. Cellular production of adenosine is greatly enhanced in inflamed tissues, ischemic tissues, and under hypoxia, where ATP is released from damaged cells. Much evidence has been accumulated on adenosine anti-inflammatory effects mediated through A2A receptor activation; A2A adenosine receptor has also been shown to play a role in matrix deposition and wound healing in a damaged tissue, contributing to dermal tissue protection and repair. Fibroblast growth factor-2 (FGF-2) is a powerful mitogen for fibroblast; it is expressed by several inflammatory cell types and plays a pivotal role in angiogenesis, wound healing, gastric ulcer protection. Human recombinant FGF-2 has been shown to have anti-inflammatory effects. The purpose of the present work was to investigate on the anti-inflammatory effect of systemic administration of the adenosine A2A agonist 2-p-(2-carboxyethyl)phenethylamino-5'-N-ethylcarboxamidoadenosine hydrochloride hydrate (CGS21680) in the rat model of carrageenan-induced paw edema. We found that CGS21680 inhibits inflammation induced by carrageenan injection into the rat paw, and this effect is associated to the local reduction of cytokine levels and dermal increase of FGF-2 expression. Our results suggest that FGF-2 might be involved in the anti-inflammatory and tissue protective effect due to A2A receptor activation.
Collapse
Affiliation(s)
- Armando Ialenti
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy (A.I., E.C., R.C., C.C.) and Department of Pharmacy, University of Salerno, Fisciano, Salerno, Italy (S.M.)
| | - Elisabetta Caiazzo
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy (A.I., E.C., R.C., C.C.) and Department of Pharmacy, University of Salerno, Fisciano, Salerno, Italy (S.M.)
| | - Silvana Morello
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy (A.I., E.C., R.C., C.C.) and Department of Pharmacy, University of Salerno, Fisciano, Salerno, Italy (S.M.)
| | - Rosa Carnuccio
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy (A.I., E.C., R.C., C.C.) and Department of Pharmacy, University of Salerno, Fisciano, Salerno, Italy (S.M.)
| | - Carla Cicala
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy (A.I., E.C., R.C., C.C.) and Department of Pharmacy, University of Salerno, Fisciano, Salerno, Italy (S.M.)
| |
Collapse
|
183
|
Koupenova M, Ravid K. Biology of Platelet Purinergic Receptors and Implications for Platelet Heterogeneity. Front Pharmacol 2018; 9:37. [PMID: 29441021 PMCID: PMC5797577 DOI: 10.3389/fphar.2018.00037] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 01/12/2018] [Indexed: 12/29/2022] Open
Abstract
Platelets are small anucleated cells present only in mammals. Platelets mediate intravascular hemostatic balance, prevent interstitial bleeding, and have a major role in thrombosis. Activation of platelet purinergic receptors is instrumental in initiation of hemostasis and formation of the hemostatic plug, although this activation process becomes problematic in pathological settings of thrombosis. This review briefly outlines the roles and function of currently known platelet purinergic receptors (P1 and P2) in the setting of hemostasis and thrombosis. Additionally, we discuss recent novel studies on purinergic receptor distribution according to heterogeneous platelet size, and the possible implication of this distribution on hemostatic function.
Collapse
Affiliation(s)
- Milka Koupenova
- Department of Medicine, Division of Cardiovascular Medicine, University of Massachusetts Medical School, Worcester, MA, United States
| | - Katya Ravid
- Departments of Medicine and Biochemistry and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
184
|
Mei HF, Poonit N, Zhang YC, Ye CY, Cai HL, Yu CY, Zhou YH, Wu BB, Cai J, Cai XH. Activating adenosine A1 receptor accelerates PC12 cell injury via ADORA1/PKC/KATP pathway after intermittent hypoxia exposure. Mol Cell Biochem 2018; 446:161-170. [PMID: 29380238 DOI: 10.1007/s11010-018-3283-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2017] [Accepted: 01/18/2018] [Indexed: 12/19/2022]
Abstract
Obstructive sleep apnea hypopnea syndrome (OSAHS) is associated with the neurocognitive deficits as a result of the neuronal cell injury. Previous studies have shown that adenosine A1 receptor (ADORA1) played an important role against hypoxia exposure, such as controlling the metabolic recovery in rat hippocampal slices and increasing the resistance in the combined effects of hypoxia and hypercapnia. However, little is known about whether ADORA1 takes part in the course of neuronal cell injury after intermittent hypoxia exposure which was the main pathological characteristic of OSAHS. The present study is performed to explore the underlying mechanism of neuronal cell injury which was induced by intermittent hypoxia exposure in PC12 cells. In our research, we find that the stimulation of the ADORA1 by CCPA accelerated the injury of PC12 cells as well as upregulated the expression of PKC, inwardly rectifying potassium channel 6.2(Kir6.2) and sulfonylurea receptor 1(SUR1) while inhibition of the ADORA1 by DPCPX alleviated the injury of PC12 cells as well as downregulated the expression of PKC, Kir6.2, and SUR1. Moreover, inhibition of the PKC by CHE, also mitigated the injury of PC12 cells, suppressed the Kir6.2 and SUR1 expressions induced by PKC. Taken together, our findings indicate that ADORA1 accelerated PC12 cells injury after intermittent hypoxia exposure via ADORA1/PKC/KATP signaling pathway.
Collapse
Affiliation(s)
- Hong-Fang Mei
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325027, China.,Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China
| | - Neha Poonit
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325027, China
| | - Yi-Chun Zhang
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325027, China
| | - Chu-Yuan Ye
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325027, China
| | - Hui-Lin Cai
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325027, China
| | - Chen-Yi Yu
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325027, China
| | - Yong-Hai Zhou
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325027, China
| | - Bei-Bei Wu
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325027, China
| | - Jun Cai
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325027, China. .,Department of Pediatrics, Children's Hospital Research Institute, The University of Louisville, Louisville, KY, USA.
| | - Xiao-Hong Cai
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325027, China.
| |
Collapse
|
185
|
Zeng M, Zhang L, Li M, Zhang B, Zhou N, Ke Y, Feng W, Zheng X. Estrogenic Effects of the Extracts from the Chinese Yam (Dioscorea opposite Thunb.) and Its Effective Compounds in Vitro and in Vivo. Molecules 2018; 23:E11. [PMID: 29360751 PMCID: PMC6017084 DOI: 10.3390/molecules23020011] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 01/16/2018] [Accepted: 01/16/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The aim of this study was to explore the estrogenic effects of the extracts from Chinese yam and its effective compounds. METHODS The activity of the yam was investigated by the uterine weight gain of mice and a proliferation assay of breast cancer cell lines (MCF-7 cell); the estrogenic activity was comprehensively evaluated by a serum pharmacology experiment. The levels of estradiol (E2), follicle stimulating hormone (FSH), and luteinizing hormone (LH) were also measured. Western blot analysis and antagonist assays with faslodex (ICI182,780), methylpiperidino-pyrazole (MPP), Delta (9) -tetrahydrocannabinol (THC), and G-15 were used to explore the mechanism of the effects of the yam. To find the effective compounds of the yam which play a role in its estrogen-like effects, we used the same methods to study the effects of adenosine and arbutin. RESULTS The Chinese yam and two main compounds, adenosine and arbutin, have estrogen-like effects. The mechanism of the yam which plays a role in its estrogen-like effects was mainly mediated by the estrogen receptors ERα, ERβ, and GPR30; that of adenosine was mainly mediated by estrogen receptors ERα and ERβ, and that of arbutin was mainly mediated by estrogen receptors ERβ and GPR30. CONCLUSIONS The Chinese yam has estrogen-like effects; adenosine and arbutin are two of the effective compounds in the yam which play a role in its estrogen-like effects.
Collapse
Affiliation(s)
- Mengnan Zeng
- Department of Medicine, Henan University of Chinese Medicine, China.
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, China.
| | - Li Zhang
- Department of Medicine, Henan University of Chinese Medicine, China.
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, China.
| | - Miao Li
- Department of Medicine, Henan University of Chinese Medicine, China.
| | - Beibei Zhang
- Department of Medicine, Henan University of Chinese Medicine, China.
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, China.
| | - Ning Zhou
- Department of Medicine, Henan University of Chinese Medicine, China.
| | - Yingying Ke
- Department of Medicine, Henan University of Chinese Medicine, China.
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, China.
| | - Weisheng Feng
- Department of Medicine, Henan University of Chinese Medicine, China.
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, China.
| | - Xiaoke Zheng
- Department of Medicine, Henan University of Chinese Medicine, China.
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, China.
| |
Collapse
|
186
|
Lee JS, Yilmaz Ö. Unfolding Role of a Danger Molecule Adenosine Signaling in Modulation of Microbial Infection and Host Cell Response. Int J Mol Sci 2018; 19:E199. [PMID: 29315226 PMCID: PMC5796148 DOI: 10.3390/ijms19010199] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 12/10/2017] [Accepted: 01/04/2018] [Indexed: 02/06/2023] Open
Abstract
Ectonucleotidases CD39 and CD73, specific nucleotide metabolizing enzymes located on the surface of the host, can convert a pro-inflammatory environment driven by a danger molecule extracellular-ATP to an adenosine-mediated anti-inflammatory milieu. Accordingly, CD39/CD73 signaling have has strongly implicated in modulating the intensity, duration, and composition of purinergic danger signals delivered to host. Recent studies have eluted potential roles for CD39 and CD73 in selective triggering of a variety of host immune cells and molecules in the presence of pathogenic microorganisms or microbial virulence molecules. Growing evidence also suggests that CD39 and CD73 present complimentary, but likely differential, actions against pathogens to shape the course and severity of microbial infection as well as the associated immune response. Similarly, adenosine receptors A2A and A2B have been proposed to be major immunomodulators of adenosine signaling during chronic inflammatory conditions induced by opportunistic pathogens, such as oral colonizer Porphyromonas gingivalis. Therefore, we here review the recent studies that demonstrate how complex network of molecules in the extracellular adenosine signaling machinery and their interactions can reshape immune responses and may also be targeted by opportunistic pathogens to establish successful colonization in human mucosal tissues and modulate the host immune response.
Collapse
Affiliation(s)
- Jaden S Lee
- Department of Oral Health Sciences, College of Dental Medicine, Medical University of South Carolina, 29425 Charleston, SC 29425, USA.
| | - Özlem Yilmaz
- Department of Oral Health Sciences, College of Dental Medicine, Medical University of South Carolina, 29425 Charleston, SC 29425, USA.
- Department of Microbiology and Immunology, Medical University of South Carolina, 29425 Charleston, SC 29425, USA.
| |
Collapse
|
187
|
Corciulo C, Irrera N, Cronstein BN. Adenosine Receptors Regulate Bone Remodeling and Cartilage Physiology. THE ADENOSINE RECEPTORS 2018:515-527. [DOI: 10.1007/978-3-319-90808-3_21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
188
|
Lavoie EG, Fausther M, Goree JR, Dranoff JA. The Cholangiocyte Adenosine-IL-6 Axis Regulates Survival During Biliary Cirrhosis. Gene Expr 2017; 17:327-340. [PMID: 28893353 PMCID: PMC5885153 DOI: 10.3727/105221617x15042723767876] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Epithelial response to injury is critical to the pathogenesis of biliary cirrhosis, and IL-6 has been suggested as a mediator of this phenomenon. Several liver cell types can secrete IL-6 following activation by various signaling molecules including circulating adenosine. The aims of this study were to assess whether adenosine can induce IL-6 secretion by cholangiocytes via the A2b adenosine receptor (A2bAR) and to determine the effect of A2bAR-sensitive IL-6 release on injury response in biliary cirrhosis. Human normal cholangiocyte H69 cells were used for in vitro studies to determine the mechanism by which adenosine and the A2bAR induce release of IL-6. In vivo, control and A2bAR-deficient mice were used to determine the roles of A2bAR-sensitive IL-6 release in biliary cirrhosis induced by common bile duct ligation (BDL). Additionally, the response to exogenous IL-6 was assessed in C57BL/6 and A2bAR-deficient mice. Adenosine induced IL-6 mRNA expression and protein secretion via A2bAR activation. Although activation of A2bAR induced cAMP and intracellular Ca2+ signals, only the Ca2+ signals were linked to IL-6 upregulation. After BDL, A2bAR-deficient mice have impaired survival, which is further impaired by exogenous IL-6; however, decreased survival is not due to changes in fibrosis and no changes in inflammatory cells. Exogenous IL-6 is associated with the increased presence of bile infarcts. Extracellular adenosine induces cholangiocyte IL-6 release via the A2bAR. This signaling pathway is important in the pathogenesis of injury response in biliary cirrhosis but does not alter fibrosis. Adenosine upregulates IL-6 release by cholangiocytes via the A2bAR in a calcium-sensitive fashion. Mice deficient in A2bAR experience impaired survival after biliary cirrhosis induced by common bile duct ligation independent of changes in fibrosis.
Collapse
Affiliation(s)
- Elise G. Lavoie
- *Division of Gastroenterology and Hepatology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- †Research Service, Central Arkansas VA Healthcare System, Little Rock, AR, USA
| | - Michel Fausther
- *Division of Gastroenterology and Hepatology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- †Research Service, Central Arkansas VA Healthcare System, Little Rock, AR, USA
| | - Jessica R. Goree
- *Division of Gastroenterology and Hepatology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- †Research Service, Central Arkansas VA Healthcare System, Little Rock, AR, USA
| | - Jonathan A. Dranoff
- *Division of Gastroenterology and Hepatology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- †Research Service, Central Arkansas VA Healthcare System, Little Rock, AR, USA
| |
Collapse
|
189
|
Rico EP, Rosemberg DB, Berteli JFA, da Silveira Langoni A, Souto AA, Bogo MR, Bonan CD, Souza DO. Adenosine deaminase activity and gene expression patterns are altered after chronic ethanol exposure in zebrafish brain. Neurotoxicol Teratol 2017; 65:14-18. [PMID: 29122710 DOI: 10.1016/j.ntt.2017.11.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 11/01/2017] [Accepted: 11/05/2017] [Indexed: 11/28/2022]
Abstract
Ethanol alters the homeostasis between excitatory and inhibitory neurotransmitters and its intoxication reveals adenosine as responsible to modify several responses including signal transduction. Zebrafish has been recently investigated for knowledge the prolonged effect of ethanol on behavioral and biochemical parameters. The aim of this study was to evaluate the soluble and membrane adenosine deaminase activities and gene expression in zebrafish brain. Animals were exposed to 0.5% ethanol for 7, 14, and 28days. There were no significant changes in ADA activity from soluble fraction after all treatments. However, we verified a decrease of ADA activity in membrane fraction after 28days (44%) of ethanol exposure. ADA1 was not altered whereas mRNA transcript levels for ADAL presented an increase after 28days of ethanol exposure (34%). ADA2-1 showed a decrease (26%) followed by an increase (17%) of transcripts after 14 and 28days of ethanol exposure, respectively. However, ADA2-1 truncated alternative splice isoform (ADA2-1/T) demonstrated a reduction after 28days (20%). ADA2-2 was decreased (22%) followed by an increase (109%) of transcripts after 14 and 18days of ethanol exposure, respectively. Altogether, the purine catabolism promoted by ADA may be an important target of the chronic toxicity induced for ethanol.
Collapse
Affiliation(s)
- Eduardo Pacheco Rico
- Programa de Pós-Graduação em Ciências da Saúde, Laboratório de Sinalização Neural e Psicofarmacologia, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Av. Universitária, 1105, Bloco S, Sala 6, Bairro Universitário, Criciúma, SC, Brazil; Instituto Nacional de Ciência e Tecnologia em Excitotoxicidade e Neuroproteção (INCT-EN), 90035-003 Porto Alegre, RS, Brazil.
| | - Denis Broock Rosemberg
- Programa de Pós-Graduação em Bioquímica Toxicológica, Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, RS, 97105-900 Santa Maria, RS, Brazil; Instituto Nacional de Ciência e Tecnologia em Excitotoxicidade e Neuroproteção (INCT-EN), 90035-003 Porto Alegre, RS, Brazil
| | - Jotele Fontana Agostini Berteli
- Programa de Pós-Graduação em Ciências da Saúde, Laboratório de Sinalização Neural e Psicofarmacologia, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Av. Universitária, 1105, Bloco S, Sala 6, Bairro Universitário, Criciúma, SC, Brazil
| | - Andrei da Silveira Langoni
- Programa de Pós-Graduação em Biologia Celular e Molecular, Laboratório de Neuroquímica e Psicofarmacologia, Brazil
| | - André Arigony Souto
- Faculdade de Química, Pontifícia Universidade Católica do Rio Grande do Sul. Avenida Ipiranga, 6681, 90619-900 Porto Alegre, RS, Brazil
| | - Maurício Reis Bogo
- Laboratório de Biologia Genômica e Molecular, Departamento de Biologia Celular e Molecular, Brazil; Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), 90035-003 Porto Alegre, RS, Brazil
| | - Carla Denise Bonan
- Programa de Pós-Graduação em Biologia Celular e Molecular, Laboratório de Neuroquímica e Psicofarmacologia, Brazil; Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), 90035-003 Porto Alegre, RS, Brazil
| | - Diogo Onofre Souza
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul. Rua Ramiro Barcelos 2600-Anexo, 90035-003 Porto Alegre, RS, Brazil; Instituto Nacional de Ciência e Tecnologia em Excitotoxicidade e Neuroproteção (INCT-EN), 90035-003 Porto Alegre, RS, Brazil
| |
Collapse
|
190
|
Li X, Xu R, Wei X, Hu H, Zhao S, Liu YM. Direct Analysis of Biofluids by Mass Spectrometry with Microfluidic Voltage-Assisted Liquid Desorption Electrospray Ionization. Anal Chem 2017; 89:12014-12022. [PMID: 29065681 DOI: 10.1021/acs.analchem.7b02398] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Signal suppression by sample matrix in direct electrospray ionization-mass spectrometric (ESI-MS) analysis hampers its clinical and biomedical applications. We report herein the development of a microfluidic voltage-assisted liquid desorption electrospray ionization (VAL-DESI) source to overcome this limitation. Liquid DESI is achieved for the first time in a microfluidic format. Direct analysis of urine, serum, and cell lysate samples by using the proposed microfluidic VAL-DESI-MS/MS method to detect chemical compounds of biomedical interest, including nucleosides, monoamines, amino acids, and peptides is demonstrated. Analyzing a set of urine samples spiked with dihydroxyphenylalanine (DOPA) showed that the assay had a linear calibration curve with r2 value of 0.997 and a limit of detection of 0.055 μM DOPA. The method was applied to simultaneous quantification of nucleosides, that is, cytidine, adenosine, uridine, thymidine, and guanosine in cell lysates using 8-bromoadenosine as internal standard. Adenosine was found most abundant at 26.5 ± 0.57 nmol/106 cells, while thymidine was least at 3.1 ± 0.31 nmol/106 cells. Interestingly, the ratio of adenosine to deoxyadenosine varied significantly from human red blood cells (1.07 ± 0.06) to cancerous cells, including lymphoblast TK6 (0.52 ± 0.02), skin melanoma C32 (0.82 ± 0.04), and promyelocytic leukemia NB4 cells (0.38 ± 0.06). These results suggest that the VAL-DESI-MS/MS technique has a good potential in direct analysis of biofluids. Further, because of the simplicity in its design and operation, the proposed microfluidic liquid DESI source can be fabricated as a disposable device for point-of-care measurements.
Collapse
Affiliation(s)
- Xiangtang Li
- Department of Chemistry and Biochemistry, Jackson State University , Jackson, Mississippi 39217, United States
| | - Rui Xu
- Department of Chemistry and Biochemistry, Jackson State University , Jackson, Mississippi 39217, United States
| | - Xin Wei
- Zhongnan Hospital of Wuhan University , Wuhan 430071, China
| | - Hankun Hu
- Zhongnan Hospital of Wuhan University , Wuhan 430071, China
| | - Shulin Zhao
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Guangxi Normal University , Guilin 541004, China
| | - Yi-Ming Liu
- Department of Chemistry and Biochemistry, Jackson State University , Jackson, Mississippi 39217, United States
| |
Collapse
|
191
|
Boison D, Steinhäuser C. Epilepsy and astrocyte energy metabolism. Glia 2017; 66:1235-1243. [PMID: 29044647 DOI: 10.1002/glia.23247] [Citation(s) in RCA: 160] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 08/29/2017] [Accepted: 10/02/2017] [Indexed: 12/17/2022]
Abstract
Epilepsy is a complex neurological syndrome characterized by neuronal hyperexcitability and sudden, synchronized electrical discharges that can manifest as seizures. It is now increasingly recognized that impaired astrocyte function and energy homeostasis play key roles in the pathogenesis of epilepsy. Excessive neuronal discharges can only happen, if adequate energy sources are made available to neurons. Conversely, energy depletion during seizures is an endogenous mechanism of seizure termination. Astrocytes control neuronal energy homeostasis through neurometabolic coupling. In this review, we will discuss how astrocyte dysfunction in epilepsy leads to distortion of key metabolic and biochemical mechanisms. Dysfunctional glutamate metabolism in astrocytes can directly contribute to neuronal hyperexcitability. Closure of astrocyte intercellular gap junction coupling as observed early during epileptogenesis limits activity-dependent trafficking of energy metabolites, but also impairs clearance of the extracellular space from accumulation of K+ and glutamate. Dysfunctional astrocytes also increase the metabolism of adenosine, a metabolic product of ATP degradation that broadly inhibits energy-consuming processes as an evolutionary adaptation to conserve energy. Due to the critical role of astroglial energy homeostasis in the control of neuronal excitability, metabolic therapeutic approaches that prevent the utilization of glucose might represent a potent antiepileptic strategy. In particular, high fat low carbohydrate "ketogenic diets" as well as inhibitors of glycolysis and lactate metabolism are of growing interest for the therapy of epilepsy.
Collapse
Affiliation(s)
- Detlev Boison
- R.S. Dow Neurobiology Laboratories, Legacy Research Institute, Portland, Oregon
| | | |
Collapse
|
192
|
Arin RM, Gorostidi A, Navarro-Imaz H, Rueda Y, Fresnedo O, Ochoa B. Adenosine: Direct and Indirect Actions on Gastric Acid Secretion. Front Physiol 2017; 8:737. [PMID: 29018360 PMCID: PMC5614973 DOI: 10.3389/fphys.2017.00737] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 09/11/2017] [Indexed: 12/12/2022] Open
Abstract
Composed by a molecule of adenine and a molecule of ribose, adenosine is a paradigm of recyclable nucleoside with a multiplicity of functions that occupies a privileged position in the metabolic and regulatory contexts. Adenosine is formed continuously in intracellular and extracellular locations of all tissues. Extracellular adenosine is a signaling molecule, able to modulate a vast range of physiologic responses in many cells and organs, including digestive organs. The adenosine A1, A2A, A2B, and A3 receptors are P1 purinergic receptors, G protein-coupled proteins implicated in tissue protection. This review is focused on gastric acid secretion, a process centered on the parietal cell of the stomach, which contains large amounts of H+/K+-ATPase, the proton pump responsible for proton extrusion during acid secretion. Gastric acid secretion is regulated by an extensive collection of neural stimuli and endocrine and paracrine agents, which act either directly at membrane receptors of the parietal cell or indirectly through other regulatory cells of the gastric mucosa, as well as mechanic and chemic stimuli. In this review, after briefly introducing these points, we condense the current body of knowledge about the modulating action of adenosine on the pathophysiology of gastric acid secretion and update its significance based on recent findings in gastric mucosa and parietal cells in humans and animal models.
Collapse
Affiliation(s)
- Rosa M Arin
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU)Leioa, Spain
| | - Adriana Gorostidi
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU)Leioa, Spain
| | - Hiart Navarro-Imaz
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU)Leioa, Spain
| | - Yuri Rueda
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU)Leioa, Spain
| | - Olatz Fresnedo
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU)Leioa, Spain
| | - Begoña Ochoa
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU)Leioa, Spain
| |
Collapse
|
193
|
Bahreyni A, Samani SS, Ghorbani E, Rahmani F, Khayami R, Toroghian Y, Behnam-Rassouli R, Khazaei M, Ryzhikov M, Parizadeh MR, Hasanzadeh M, Avan A, Hassanian SM. Adenosine: An endogenous mediator in the pathogenesis of gynecological cancer. J Cell Physiol 2017; 233:2715-2722. [PMID: 28617999 DOI: 10.1002/jcp.26056] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 06/14/2017] [Indexed: 12/15/2022]
Abstract
Extracellular concentration of adenosine increases in the hypoxic tumor microenvironment. Adenosine signaling regulates apoptosis, angiogenesis, metastasis, and immune suppression in cancer cells. Adenosine-induced cell responses depend upon different subtypes of adenosine receptors activation and type of cancer. Suppression of adenosine signaling via inhibition of adenosine receptors or adenosine generating enzymes including CD39 and CD73 on ovarian or cervical cancer cells is a potentially novel therapeutic approach for gynecological cancer patients. This review summarizes the role of adenosine in the pathogenesis of gynecological cancer for a better understanding and hence a better management of this disease.
Collapse
Affiliation(s)
- Amirhossein Bahreyni
- Faculty of Medicine, Department of Clinical Biochemistry and Immunogenetic Research Center, Mazandaran University of Medical Sciences, Sari, Mazandaran, Iran
| | - Seyed S Samani
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Elnaz Ghorbani
- Department of Microbiology, Al-Zahra University, Tehran, Iran
| | - Farzad Rahmani
- Faculty of Medicine, Department of Medical Biochemistry, Mashhad University of Medical Sciences, Mashhad, Iran.,Faculty of Medicine, Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reza Khayami
- Faculty of Medicine, Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Younes Toroghian
- Faculty of Medicine, Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Majid Khazaei
- Faculty of Medicine, Department of Medical Physiology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mikhail Ryzhikov
- Department of Molecular Microbiology and Immunology, St. Louis University School of Medicine, Saint Louis, Missouri
| | - Mohammad R Parizadeh
- Faculty of Medicine, Department of Medical Biochemistry, Mashhad University of Medical Sciences, Mashhad, Iran.,Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Malihe Hasanzadeh
- Faculty of Medicine, Department of Gynecology Oncology, Woman Health Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Faculty of Medicine, Department of Modern Sciences and Technologies, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed M Hassanian
- Faculty of Medicine, Department of Medical Biochemistry, Mashhad University of Medical Sciences, Mashhad, Iran.,Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Microanatomy Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
194
|
Ng WY, Migotto A, Ferreira TS, Lopes LB. Monoolein-alginate beads as a platform to promote adenosine cutaneous localization and wound healing. Int J Biol Macromol 2017; 102:1104-1111. [DOI: 10.1016/j.ijbiomac.2017.04.094] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Revised: 03/28/2017] [Accepted: 04/03/2017] [Indexed: 01/16/2023]
|
195
|
Xu Y, Wang Y, Yan S, Zhou Y, Yang Q, Pan Y, Zeng X, An X, Liu Z, Wang L, Xu J, Cao Y, Fulton DJ, Weintraub NL, Bagi Z, Hoda MN, Wang X, Li Q, Hong M, Jiang X, Boison D, Weber C, Wu C, Huo Y. Intracellular adenosine regulates epigenetic programming in endothelial cells to promote angiogenesis. EMBO Mol Med 2017; 9:1263-1278. [PMID: 28751580 PMCID: PMC5582416 DOI: 10.15252/emmm.201607066] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 06/26/2017] [Accepted: 06/30/2017] [Indexed: 01/07/2023] Open
Abstract
The nucleoside adenosine is a potent regulator of vascular homeostasis, but it remains unclear how expression or function of the adenosine-metabolizing enzyme adenosine kinase (ADK) and the intracellular adenosine levels influence angiogenesis. We show here that hypoxia lowered the expression of ADK and increased the levels of intracellular adenosine in human endothelial cells. Knockdown (KD) of ADK elevated intracellular adenosine, promoted proliferation, migration, and angiogenic sprouting in human endothelial cells. Additionally, mice deficient in endothelial ADK displayed increased angiogenesis as evidenced by the rapid development of the retinal and hindbrain vasculature, increased healing of skin wounds, and prompt recovery of arterial blood flow in the ischemic hindlimb. Mechanistically, hypomethylation of the promoters of a series of pro-angiogenic genes, especially for VEGFR2 in ADK KD cells, was demonstrated by the Infinium methylation assay. Methylation-specific PCR, bisulfite sequencing, and methylated DNA immunoprecipitation further confirmed hypomethylation in the promoter region of VEGFR2 in ADK-deficient endothelial cells. Accordingly, loss or inactivation of ADK increased VEGFR2 expression and signaling in endothelial cells. Based on these findings, we propose that ADK downregulation-induced elevation of intracellular adenosine levels in endothelial cells in the setting of hypoxia is one of the crucial intrinsic mechanisms that promote angiogenesis.
Collapse
Affiliation(s)
- Yiming Xu
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yong Wang
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Siyuan Yan
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Science, Beijing, China
| | - Yaqi Zhou
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA
- Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Qiuhua Yang
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA
- Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Yue Pan
- Georgia Prevention Institute, Augusta University, Augusta, GA, USA
| | - Xianqiu Zeng
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA
- Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Xiaofei An
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Zhiping Liu
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Lina Wang
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA
- Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Jiean Xu
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA
- Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Yapeng Cao
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA
- Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China
| | - David J Fulton
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Neal L Weintraub
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Zsolt Bagi
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Md Nasrul Hoda
- Departments of Medical Laboratory, Imaging & Radiologic Sciences, and Neurology, Augusta University, Augusta, GA, USA
| | - Xiaoling Wang
- Georgia Prevention Institute, Augusta University, Augusta, GA, USA
| | - Qinkai Li
- Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Mei Hong
- Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Xuejun Jiang
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Science, Beijing, China
| | - Detlev Boison
- Robert S. Dow Neurobiology Laboratories, Legacy Research Institute, Portland, OR, USA
| | - Christian Weber
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Chaodong Wu
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, USA
| | - Yuqing Huo
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA
| |
Collapse
|
196
|
Antonioli L, Pellegrini C, Fornai M, Tirotta E, Gentile D, Benvenuti L, Giron MC, Caputi V, Marsilio I, Orso G, Bernardini N, Segnani C, Ippolito C, Csóka B, Németh ZH, Haskó G, Scarpignato C, Blandizzi C, Colucci R. Colonic motor dysfunctions in a mouse model of high-fat diet-induced obesity: an involvement of A 2B adenosine receptors. Purinergic Signal 2017; 13:497-510. [PMID: 28808842 DOI: 10.1007/s11302-017-9577-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 08/01/2017] [Indexed: 12/13/2022] Open
Abstract
Adenosine A2B receptors (A2BR) regulate several enteric functions. However, their implication in the pathophysiology of intestinal dysmotility associated with high-fat diet (HFD)-induced obesity has not been elucidated. We investigated the expression of A2BR in mouse colon and their role in the mechanisms underlying the development of enteric dysmotility associated with obesity. Wild-type C57BL/6J mice were fed with HFD (60% kcal from fat) or normocaloric diet (NCD; 18% kcal from fat) for 8 weeks. Colonic A2BR localization was examined by immunofluorescence. The role of A2BR in the control of colonic motility was examined in functional experiments on longitudinal muscle preparations (LMPs). In NCD mice, A2BR were predominantly located in myenteric neurons; in HFD animals, their expression increased throughout the neuromuscular layer. Functionally, the A2BR antagonist MRS1754 enhanced electrically induced NK1-mediated tachykininergic contractions in LMPs from HFD mice, while it was less effective in tissues from NCD mice. The A2B receptor agonist BAY 60-6583 decreased colonic tachykininergic contractions in LMPs, with higher efficacy in preparations from obese mice. Both A2BR ligands did not affect contractions elicited by exogenous substance P. Obesity is related with a condition of colonic inflammation, leading to an increase of A2BR expression. A2BR, modulating the activity of excitatory tachykininergic nerves, participate to the enteric dysmotility associated with obesity.
Collapse
Affiliation(s)
- Luca Antonioli
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 55, 56126, Pisa, Italy.,Department of Surgery and Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA
| | - Carolina Pellegrini
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 55, 56126, Pisa, Italy
| | - Matteo Fornai
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 55, 56126, Pisa, Italy.
| | - Erika Tirotta
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 55, 56126, Pisa, Italy
| | - Daniela Gentile
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 55, 56126, Pisa, Italy
| | - Laura Benvenuti
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 55, 56126, Pisa, Italy
| | - Maria Cecilia Giron
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Valentina Caputi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy.,San Camillo Hospital, Treviso, Italy
| | - Ilaria Marsilio
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Genny Orso
- Scientific Institute IRCCS Eugenio Medea, Bosisio Parini-, Lecco, Italy
| | - Nunzia Bernardini
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 55, 56126, Pisa, Italy
| | - Cristina Segnani
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 55, 56126, Pisa, Italy
| | - Chiara Ippolito
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 55, 56126, Pisa, Italy
| | - Balázs Csóka
- Department of Surgery and Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA
| | - Zoltán H Németh
- Department of Surgery, Morristown Medical Center, Morristown, NJ, USA
| | - György Haskó
- Department of Surgery and Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA
| | | | - Corrado Blandizzi
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 55, 56126, Pisa, Italy
| | - Rocchina Colucci
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| |
Collapse
|
197
|
Robertson FP, Fuller BJ, Davidson BR. An Evaluation of Ischaemic Preconditioning as a Method of Reducing Ischaemia Reperfusion Injury in Liver Surgery and Transplantation. J Clin Med 2017; 6:jcm6070069. [PMID: 28708111 PMCID: PMC5532577 DOI: 10.3390/jcm6070069] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 06/22/2017] [Accepted: 07/04/2017] [Indexed: 12/16/2022] Open
Abstract
Liver Ischaemia Reperfusion (IR) injury is a major cause of post-operative liver dysfunction, morbidity and mortality following liver resection surgery and transplantation. There are no proven therapies for IR injury in clinical practice and new approaches are required. Ischaemic Preconditioning (IPC) can be applied in both a direct and remote fashion and has been shown to ameliorate IR injury in small animal models. Its translation into clinical practice has been difficult, primarily by a lack of knowledge regarding the dominant protective mechanisms that it employs. A review of all current studies would suggest that IPC/RIPC relies on creating a small tissue injury resulting in the release of adenosine and l-arginine which act through the Adenosine receptors and the haem-oxygenase and endothelial nitric oxide synthase systems to reduce hepatocyte necrosis and improve the hepatic microcirculation post reperfusion. The next key step is to determine how long the stimulus requires to precondition humans to allow sufficient injury to occur to release the potential mediators. This would open the door to a new therapeutic chapter in this field.
Collapse
Affiliation(s)
- Francis P Robertson
- Division of Surgery and Interventional Science, Royal Free Campus, University College London, 9th Floor, Royal Free Hospital, Pond Street, London NW3 2QG, UK.
| | - Barry J Fuller
- Division of Surgery and Interventional Science, Royal Free Campus, University College London, 9th Floor, Royal Free Hospital, Pond Street, London NW3 2QG, UK.
| | - Brian R Davidson
- Division of Surgery and Interventional Science, Royal Free Campus, University College London, 9th Floor, Royal Free Hospital, Pond Street, London NW3 2QG, UK.
- Department of Hepaticopancreatobiliary Surgery and Liver Transplantation, Royal Free Foundation Trust, 9th Floor, Royal Free Hospital, Pond Street, London NW3 2QG, UK.
| |
Collapse
|
198
|
Borea PA, Gessi S, Merighi S, Vincenzi F, Varani K. Pathological overproduction: the bad side of adenosine. Br J Pharmacol 2017; 174:1945-1960. [PMID: 28252203 PMCID: PMC6398520 DOI: 10.1111/bph.13763] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 02/22/2017] [Accepted: 02/23/2017] [Indexed: 12/12/2022] Open
Abstract
Adenosine is an endogenous ubiquitous purine nucleoside, which is increased by hypoxia, ischaemia and tissue damage and mediates a number of physiopathological effects by interacting with four GPCRs, identified as A1 , A2A , A2B and A3 . Physiological and acutely increased adenosine is mostly associated with beneficial effects that include vasodilatation and a decrease in inflammation. In contrast, chronic overproduction of adenosine occurs in important pathological states, where long-lasting increases in the nucleoside levels are responsible for the bad side of adenosine associated with chronic inflammation, fibrosis and organ damage. In this review, we describe and critically discuss the pathological overproduction of adenosine and analyse when, where and how adenosine exerts its detrimental effects throughout the body.
Collapse
Affiliation(s)
- Pier Andrea Borea
- Department of Medical SciencesUniversity of FerraraFerrara44121Italy
| | - Stefania Gessi
- Department of Medical SciencesUniversity of FerraraFerrara44121Italy
| | - Stefania Merighi
- Department of Medical SciencesUniversity of FerraraFerrara44121Italy
| | - Fabrizio Vincenzi
- Department of Medical SciencesUniversity of FerraraFerrara44121Italy
| | - Katia Varani
- Department of Medical SciencesUniversity of FerraraFerrara44121Italy
| |
Collapse
|
199
|
Li M, Dai Y, Lei J, Tang J, Zhou Y, Xia B, Xia Y, Yin G. Acute rejection after kidney transplantation promotes graft fibrosis with elevated adenosine level in rat. PLoS One 2017. [PMID: 28651015 PMCID: PMC5484506 DOI: 10.1371/journal.pone.0180211] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Aims Chronic allograft nephropathy is a worldwide issue with the major feature of progressive allograft fibrosis, eventually ending with graft loss. Adenosine has been demonstrated to play an important role in process of fibrosis. Our study aimed to investigate the relationship between adenosine and fibrosis in renal allograft acute rejection in rat. Materials and methods Wistar rats and SD rats were selected as experimental animals. Our study designed two groups. In the allograft transplantation group, kidneys of Wistar rats were orthotopically transplanted into SD rat recipients, the same species but not genetically identical, to induce acute rejection. Kidney transplantations of SD rats to SD rats which were genetically identical were served as the control. We established rat models and detected a series of indicators. All data were analyzed statistically. P<0.05 was considered statistically significant. Results Compared with the control group, levels of adenosine increased significantly in the allograft transplantation group, in which acute rejection was induced (P<0.05). Progressive allograft fibrosis as well as collagen deposition were observed. Conclusions These findings suggested that level of adenosine was upregulated in acute rejection after kidney allograft transplantation in rat. Acute rejection may promote renal allograft fibrosis via the adenosine signaling pathways.
Collapse
Affiliation(s)
- Mingliang Li
- Department of Urology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Department of Urology, The First People's Hospital of Xiangtan City, Xiangtan, China
| | - Yingbo Dai
- Department of Urology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Jun Lei
- Department of Urology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Jin Tang
- Department of Urology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yihong Zhou
- Department of Urology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Bing Xia
- Department of Urology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yang Xia
- Department of Biochemistry and Molecular Biology, The University of Texas–Houston Medical School, Houston, Texas, United States of America
| | - Guangming Yin
- Department of Urology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- * E-mail:
| |
Collapse
|
200
|
Salsoso R, Farías M, Gutiérrez J, Pardo F, Chiarello DI, Toledo F, Leiva A, Mate A, Vázquez CM, Sobrevia L. Adenosine and preeclampsia. Mol Aspects Med 2017; 55:126-139. [DOI: 10.1016/j.mam.2016.12.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Revised: 12/16/2016] [Accepted: 12/23/2016] [Indexed: 01/13/2023]
|