1
|
Massold T, Ibrahim F, Niemann V, Steckel B, Becker K, Schrader J, Stegbauer J, Temme S, Grandoch M, Flögel U, Bouvain P. CD73 deficiency does not aggravate angiotensin II-induced aortic inflammation in mice. Sci Rep 2023; 13:17125. [PMID: 37816827 PMCID: PMC10564884 DOI: 10.1038/s41598-023-44361-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 10/06/2023] [Indexed: 10/12/2023] Open
Abstract
Vascular inflammation plays a key role in the development of aortic diseases. A potential novel target for treatment might be CD73, an ecto-5'-nucleotidase that generates anti-inflammatory adenosine in the extracellular space. Here, we investigated whether a lack of CD73 results in enhanced aortic inflammation. To this end, angiotensin II was infused into wildtype and CD73-/- mice over 10 days. Before and after infusion, mice were analyzed using magnetic resonance imaging, ultrasound, flow cytometry, and histology. The impact of age and gender was investigated using female and male mice of three and six months of age, respectively. Angiotensin II infusion led to increased immune cell infiltration in both genotypes' aortae, but depletion of CD73 had no impact on immune cell recruitment. These findings were not modified by age or sex. No substantial difference in morphological or functional characteristics could be detected between wildtype and CD73-/- mice. Interestingly, the expression of CD73 on neutrophils decreased significantly in wildtype mice during treatment. In summary, we have found no evidence that CD73 deficiency affects the onset of aortic inflammation. However, as CD73 expression decreased during disease induction, an increase in CD73 by pharmaceutical intervention might result in lower vascular inflammation and less vascular disease.
Collapse
Affiliation(s)
- Timo Massold
- Experimental Cardiovascular Imaging, Department of Molecular Cardiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Fady Ibrahim
- Experimental Cardiovascular Imaging, Department of Molecular Cardiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Viola Niemann
- Institute for Translational Pharmacology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Bodo Steckel
- Department of Molecular Cardiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Katrin Becker
- Department of Cardiology, Pulmonology, and Angiology, University Hospital Düsseldorf, Düsseldorf, Germany
- Institute for Cardiovascular Sciences, Endothelial Signaling and Metabolism, University Hospital Bonn, Bonn, Germany
| | - Jürgen Schrader
- Department of Molecular Cardiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Johannes Stegbauer
- Department of Nephrology, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, 40225, Düsseldorf, Germany
| | - Sebastian Temme
- Department of Anesthesiology, University Hospital Düsseldorf, Düsseldorf, Germany
- CARID, Cardiovascular Research Institute Düsseldorf, Düsseldorf, Germany
| | - Maria Grandoch
- Institute for Translational Pharmacology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- CARID, Cardiovascular Research Institute Düsseldorf, Düsseldorf, Germany
| | - Ulrich Flögel
- Experimental Cardiovascular Imaging, Department of Molecular Cardiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
- CARID, Cardiovascular Research Institute Düsseldorf, Düsseldorf, Germany.
| | - Pascal Bouvain
- Experimental Cardiovascular Imaging, Department of Molecular Cardiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|
2
|
Kelestemur T, Németh ZH, Pacher P, Beesley J, Robson SC, Eltzschig HK, Haskó G. Adenosine metabolized from extracellular ATP ameliorates organ injury by triggering A 2BR signaling. Respir Res 2023; 24:186. [PMID: 37438813 PMCID: PMC10339538 DOI: 10.1186/s12931-023-02486-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 06/29/2023] [Indexed: 07/14/2023] Open
Abstract
BACKGROUND Trauma and a subsequent hemorrhagic shock (T/HS) result in insufficient oxygen delivery to tissues and multiple organ failure. Extracellular adenosine, which is a product of the extracellular degradation of adenosine 5' triphosphate (ATP) by the membrane-embedded enzymes CD39 and CD73, is organ protective, as it participates in signaling pathways, which promote cell survival and suppress inflammation through adenosine receptors including the A2BR. The aim of this study was to evaluate the role of CD39 and CD73 delivering adenosine to A2BRs in regulating the host's response to T/HS. METHODS T/HS shock was induced by blood withdrawal from the femoral artery in wild-type, global knockout (CD39, CD73, A2BR) and conditional knockout (intestinal epithelial cell-specific deficient VillinCre-A2BRfl/fl) mice. At 3 three hours after resuscitation, blood and tissue samples were collected to analyze organ injury. RESULTS T/HS upregulated the expression of CD39, CD73, and the A2BR in organs. ATP and adenosine levels increased after T/HS in bronchoalveolar lavage fluid. CD39, CD73, and A2BR mimics/agonists alleviated lung and liver injury. Antagonists or the CD39, CD73, and A2BR knockout (KO) exacerbated lung injury, inflammatory cytokines, and chemokines as well as macrophage and neutrophil infiltration and accumulation in the lung. Agonists reduced the levels of the liver enzymes aspartate transferase and alanine transaminase in the blood, whereas antagonist administration or CD39, CD73, and A2BR KO enhanced enzyme levels. In addition, intestinal epithelial cell-specific deficient VillinCre-A2BRfl/fl mice showed increased intestinal injury compared to their wild-type VillinCre controls. CONCLUSION In conclusion, the CD39-CD73-A2BR axis protects against T/HS-induced multiple organ failure.
Collapse
Affiliation(s)
- Taha Kelestemur
- Department of Anesthesiology, Columbia University, 630 W 168th Street, New York City, NY, 10032, USA
- Department of Physiology, Faculty of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Zoltán H Németh
- Department of Anesthesiology, Columbia University, 630 W 168th Street, New York City, NY, 10032, USA
- Department of Surgery, Morristown Medical Center, Morristown, NJ, 07960, USA
| | - Pal Pacher
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Jennet Beesley
- Daresbury Proteins Ltd, Sci-Tech Daresbury, Warrington, UK
| | - Simon C Robson
- Department of Anesthesia, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Holger K Eltzschig
- Department of Anesthesiology, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - György Haskó
- Department of Anesthesiology, Columbia University, 630 W 168th Street, New York City, NY, 10032, USA.
| |
Collapse
|
3
|
Yuan X, Mills T, Doursout MF, Evans SE, Vidal Melo MF, Eltzschig HK. Alternative adenosine Receptor activation: The netrin-Adora2b link. Front Pharmacol 2022; 13:944994. [PMID: 35910389 PMCID: PMC9334855 DOI: 10.3389/fphar.2022.944994] [Citation(s) in RCA: 10] [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: 05/16/2022] [Accepted: 06/28/2022] [Indexed: 11/25/2022] Open
Abstract
During hypoxia or inflammation, extracellular adenosine levels are elevated. Studies using pharmacologic approaches or genetic animal models pertinent to extracellular adenosine signaling implicate this pathway in attenuating hypoxia-associated inflammation. There are four distinct adenosine receptors. Of these, it is not surprising that the Adora2b adenosine receptor functions as an endogenous feedback loop to control hypoxia-associated inflammation. First, Adora2b activation requires higher adenosine concentrations compared to other adenosine receptors, similar to those achieved during hypoxic inflammation. Second, Adora2b is transcriptionally induced during hypoxia or inflammation by hypoxia-inducible transcription factor HIF1A. Studies seeking an alternative adenosine receptor activation mechanism have linked netrin-1 with Adora2b. Netrin-1 was originally discovered as a neuronal guidance molecule but also functions as an immune-modulatory signaling molecule. Similar to Adora2b, netrin-1 is induced by HIF1A, and has been shown to enhance Adora2b signaling. Studies of acute respiratory distress syndrome (ARDS), intestinal inflammation, myocardial or hepatic ischemia and reperfusion implicate the netrin-Adora2b link in tissue protection. In this review, we will discuss the potential molecular linkage between netrin-1 and Adora2b, and explore studies demonstrating interactions between netrin-1 and Adora2b in attenuating tissue inflammation.
Collapse
Affiliation(s)
- Xiaoyi Yuan
- Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Tingting Mills
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Marie-Francoise Doursout
- Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Scott E. Evans
- Department of Pulmonology, MD Anderson Cancer Center, 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
| |
Collapse
|
4
|
Yang JH, Choi HP, Niu W, Azadzoi KM. Cellular Stress and Molecular Responses in Bladder Ischemia. Int J Mol Sci 2021; 22:ijms222111862. [PMID: 34769293 PMCID: PMC8584445 DOI: 10.3390/ijms222111862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/28/2021] [Accepted: 10/29/2021] [Indexed: 11/16/2022] Open
Abstract
The concept of bladder ischemia as a contributing factor to detrusor overactivity and lower urinary tract symptoms (LUTS) is evolving. Bladder ischemia as a consequence of pelvic arterial atherosclerosis was first documented in experimental models and later in elderly patients with LUTS. It was shown that early-stage moderate ischemia produces detrusor overactivity, while prolonged severe ischemia provokes changes consistent with detrusor underactivity. Recent studies imply a central role of cellular energy sensors, cellular stress sensors, and stress response molecules in bladder responses to ischemia. The cellular energy sensor adenosine monophosphate-activated protein kinase was shown to play a role in detrusor overactivity and neurodegeneration in bladder ischemia. The cellular stress sensors apoptosis signal-regulating kinase 1 and caspase-3 along with heat shock proteins were characterized as important contributing factors to smooth muscle structural modifications and apoptotic responses in bladder ischemia. Downstream pathways seem to involve hypoxia-inducible factor, transforming growth factor beta, vascular endothelial growth factor, and nerve growth factor. Molecular responses to bladder ischemia were associated with differential protein expression, the accumulation of non-coded amino acids, and post-translational modifications of contractile proteins and stress response molecules. Further insight into cellular stress responses in bladder ischemia may provide novel diagnostic and therapeutic targets against LUTS.
Collapse
Affiliation(s)
- Jing-Hua Yang
- Department of Surgery, Boston University School of Medicine, Boston, MA 02118, USA;
- Proteomics Laboratory, VA Boston Healthcare System, Boston, MA 02130, USA;
| | - Han-Pil Choi
- Proteomics Laboratory, VA Boston Healthcare System, Boston, MA 02130, USA;
| | - Wanting Niu
- Research Section, VA Boston Healthcare System, Boston, MA 02130, USA;
| | - Kazem M. Azadzoi
- Departments of Urology and Pathology, VA Boston Healthcare System and Boston University School of Medicine, Boston, MA 02130, USA
- Correspondence: ; Tel.: +1-(857)-364-5602
| |
Collapse
|
5
|
Role of Purinergic Signalling in Endothelial Dysfunction and Thrombo-Inflammation in Ischaemic Stroke and Cerebral Small Vessel Disease. Biomolecules 2021; 11:biom11070994. [PMID: 34356618 PMCID: PMC8301873 DOI: 10.3390/biom11070994] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 06/28/2021] [Accepted: 07/01/2021] [Indexed: 12/20/2022] Open
Abstract
The cerebral endothelium is an active interface between blood and the central nervous system. In addition to being a physical barrier between the blood and the brain, the endothelium also actively regulates metabolic homeostasis, vascular tone and permeability, coagulation, and movement of immune cells. Being part of the blood–brain barrier, endothelial cells of the brain have specialized morphology, physiology, and phenotypes due to their unique microenvironment. Known cardiovascular risk factors facilitate cerebral endothelial dysfunction, leading to impaired vasodilation, an aggravated inflammatory response, as well as increased oxidative stress and vascular proliferation. This culminates in the thrombo-inflammatory response, an underlying cause of ischemic stroke and cerebral small vessel disease (CSVD). These events are further exacerbated when blood flow is returned to the brain after a period of ischemia, a phenomenon termed ischemia-reperfusion injury. Purinergic signaling is an endogenous molecular pathway in which the enzymes CD39 and CD73 catabolize extracellular adenosine triphosphate (eATP) to adenosine. After ischemia and CSVD, eATP is released from dying neurons as a damage molecule, triggering thrombosis and inflammation. In contrast, adenosine is anti-thrombotic, protects against oxidative stress, and suppresses the immune response. Evidently, therapies that promote adenosine generation or boost CD39 activity at the site of endothelial injury have promising benefits in the context of atherothrombotic stroke and can be extended to current CSVD known pathomechanisms. Here, we have reviewed the rationale and benefits of CD39 and CD39 therapies to treat endothelial dysfunction in the brain.
Collapse
|
6
|
Wang K, Zhang Z, Meng D, Li J. Investigating genetic drivers of juvenile dermatomyositis pathogenesis using bioinformatics methods. J Dermatol 2021; 48:1007-1020. [PMID: 33891717 DOI: 10.1111/1346-8138.15856] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 03/03/2021] [Accepted: 03/04/2021] [Indexed: 11/30/2022]
Abstract
Juvenile dermatomyositis (JDM) is a chronic autoimmune disease. The pathogenic mechanisms remain ill-defined. The purpose of this study was to identify key genes related to JDM. Microarray datasets were downloaded from the Gene Expression Omnibus database. The differentially expressed genes (DEG) were identified. Then, Gene Ontology, Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis, and protein-protein interaction (PPI) network were carried out. In addition, the hub genes were selected by cytoHubba. The expression profile and diagnostic capacity (receiver-operator curve [ROC]) of interested hub genes were verified. Gene set enrichment analysis (GSEA) was also carried out. Moreover, the signature of hub genes was then used as a search query to explore the Connectivity Map (CMAP). A total of 128 DEG were identified. The enriched functions and pathways of the DEG include response to virus, negative regulation of cell migration, cadmium ion transmembrane transport, defense response to Gram-negative bacterium, positive regulation of megakaryocyte differentiation, and negative regulation of angiogenesis. Twenty-one hub genes were identified. The expression levels of the interested genes were also confirmed. ROC analysis confirmed that the expression of these genes can distinguish JDM from controls. GSEA showed that these genes are mainly related to "inflammatory response", "complement", "interferon-α response", "IL6/JAK/STAT3 signaling", "TGF-β signaling", "IL2/STAT5 signaling" and "TNF-α signaling via NF-κB". The CMAP research found some compounds with the potential to counteract the effects of the dysregulated molecular signature in JDM. In this study, bioinformatics methods were used to identify DEG, which helps us understand the molecular mechanisms of JDM and provide candidate targets for diagnosis and treatment of JDM.
Collapse
Affiliation(s)
- Kai Wang
- Department of Rheumatology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huaian, China
| | - Zhongyuan Zhang
- Department of Rheumatology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huaian, China
| | - Deqian Meng
- Department of Rheumatology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huaian, China
| | - Ju Li
- Department of Rheumatology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huaian, China
| |
Collapse
|
7
|
Paganelli F, Mottola G, Fromonot J, Marlinge M, Deharo P, Guieu R, Ruf J. Hyperhomocysteinemia and Cardiovascular Disease: Is the Adenosinergic System the Missing Link? Int J Mol Sci 2021; 22:1690. [PMID: 33567540 PMCID: PMC7914561 DOI: 10.3390/ijms22041690] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 01/30/2021] [Accepted: 02/04/2021] [Indexed: 12/17/2022] Open
Abstract
The influence of hyperhomocysteinemia (HHCy) on cardiovascular disease (CVD) remains unclear. HHCy is associated with inflammation and atherosclerosis, and it is an independent risk factor for CVD, stroke and myocardial infarction. However, homocysteine (HCy)-lowering therapy does not affect the inflammatory state of CVD patients, and it has little influence on cardiovascular risk. The HCy degradation product hydrogen sulfide (H2S) is a cardioprotector. Previous research proposed a positive role of H2S in the cardiovascular system, and we discuss some recent data suggesting that HHCy worsens CVD by increasing the production of H2S, which decreases the expression of adenosine A2A receptors on the surface of immune and cardiovascular cells to cause inflammation and ischemia, respectively.
Collapse
Affiliation(s)
- Franck Paganelli
- C2VN, INSERM, INRAE, Aix-Marseille University, F-13005 Marseille, France; (F.P.); (G.M.); (J.F.); (M.M.); (P.D.); (R.G.)
- Department of Cardiology, North Hospital, F-13015 Marseille, France
| | - Giovanna Mottola
- C2VN, INSERM, INRAE, Aix-Marseille University, F-13005 Marseille, France; (F.P.); (G.M.); (J.F.); (M.M.); (P.D.); (R.G.)
- Laboratory of Biochemistry, Timone Hospital, F-13005 Marseille, France
| | - Julien Fromonot
- C2VN, INSERM, INRAE, Aix-Marseille University, F-13005 Marseille, France; (F.P.); (G.M.); (J.F.); (M.M.); (P.D.); (R.G.)
- Laboratory of Biochemistry, Timone Hospital, F-13005 Marseille, France
| | - Marion Marlinge
- C2VN, INSERM, INRAE, Aix-Marseille University, F-13005 Marseille, France; (F.P.); (G.M.); (J.F.); (M.M.); (P.D.); (R.G.)
- Laboratory of Biochemistry, Timone Hospital, F-13005 Marseille, France
| | - Pierre Deharo
- C2VN, INSERM, INRAE, Aix-Marseille University, F-13005 Marseille, France; (F.P.); (G.M.); (J.F.); (M.M.); (P.D.); (R.G.)
- Department of Cardiology, Timone Hospital, F-13005 Marseille, France
| | - Régis Guieu
- C2VN, INSERM, INRAE, Aix-Marseille University, F-13005 Marseille, France; (F.P.); (G.M.); (J.F.); (M.M.); (P.D.); (R.G.)
- Laboratory of Biochemistry, Timone Hospital, F-13005 Marseille, France
| | - Jean Ruf
- C2VN, INSERM, INRAE, Aix-Marseille University, F-13005 Marseille, France; (F.P.); (G.M.); (J.F.); (M.M.); (P.D.); (R.G.)
| |
Collapse
|
8
|
Zeng J, Ning Z, Wang Y, Xiong H. Implications of CD39 in immune-related diseases. Int Immunopharmacol 2020; 89:107055. [PMID: 33045579 DOI: 10.1016/j.intimp.2020.107055] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 09/10/2020] [Accepted: 09/26/2020] [Indexed: 02/07/2023]
Abstract
Extracellular adenosine triphosphate (eATP) mediates pro-inflammatory responses by recruiting and activating inflammatory cells. CD39 can hydrolyze eATP into adenosine monophosphate (AMP), while CD73 can convert AMP into the immunosuppressive nucleoside adenosine (ADO). CD39 is a rate-limiting enzyme in this cascade, which is regarded as an immunological switch shifting the ATP-mediated pro-inflammatory environment to the ADO- mediated anti-inflammatory status. The CD39 expression can be detected in a wide spectrum of immunocytes, which is under the influence of environmental and genetic factors. It is increasingly suggested that, CD39 participates in some pathophysiological processes, like inflammatory bowel disease (IBD), sepsis, multiple sclerosis (MS), allergic diseases, ischemia-reperfusion (I/R) injury, systemic lupus erythematosus (SLE), diabetes and cancer. Here, we focus on the current understanding of CD39 in immunity, and comprehensively illustrate the diverse CD39 functions within a variety of disorders.
Collapse
Affiliation(s)
- Jianrui Zeng
- Institute of Immunology and Molecular Medicine, Jining Medical University, Shandong 272067, China
| | - Zhaochen Ning
- Institute of Immunology and Molecular Medicine, Jining Medical University, Shandong 272067, China
| | - Yuzhong Wang
- Department of Neurology and Central Laboratory, Affiliated Hospital of Jining Medical University, Shandong 272000, China.
| | - Huabao Xiong
- Institute of Immunology and Molecular Medicine, Jining Medical University, Shandong 272067, China.
| |
Collapse
|
9
|
Han SJ, Kim M, Novitsky E, D'Agati V, Lee HT. Intestinal TLR9 deficiency exacerbates hepatic IR injury via altered intestinal inflammation and short-chain fatty acid synthesis. FASEB J 2020; 34:12083-12099. [PMID: 32738096 DOI: 10.1096/fj.202000314r] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 06/22/2020] [Accepted: 06/28/2020] [Indexed: 12/13/2022]
Abstract
Mice deficient in intestinal epithelial TLR9 develop small intestinal Paneth cell hyperplasia and higher Paneth cell IL-17A levels. Since small intestinal Paneth cells and IL-17A play critical roles in hepatic ischemia reperfusion (IR) injury, we tested whether mice lacking intestinal TLR9 have increased hepatic IR injury. Mice lacking intestinal TLR9 had profoundly increased liver injury after hepatic IR compared to WT mice with exacerbated hepatocyte necrosis, apoptosis, neutrophil infiltration, and inflammatory cytokine generation. Moreover, we observed increased small intestinal inflammation and apoptosis after hepatic IR in intestinal TLR9 deficient mice. As a potential explanation for increased hepatic IR injury, fecal short-chain fatty acids butyrate and propionate levels were lower in intestinal TLR9 deficient mice. Suggesting a potential therapy for hepatic IR, exogenous administration of butyrate or propionate protected against hepatic IR injury in intestinal TLR9 deficient mice. Mechanistically, butyrate induced small intestinal IL-10 expression and downregulated the claudin-2 expression. Finally, IL-10 neutralization abolished the protective effects of butyrate against hepatic IR injury. Our studies show intestinal TLR9 deficiency results in exacerbated hepatic IR injury with increased small intestinal apoptosis and inflammation. Furthermore, short-chain fatty acids butyrate and propionate protect against hepatic IR injury and intestinal apoptosis/inflammation in intestinal TLR9 deficient mice.
Collapse
Affiliation(s)
- Sang Jun Han
- Anesthesiology Research Laboratories, Department of Anesthesiology, Columbia University, New York, NY, USA
| | - Mihwa Kim
- Anesthesiology Research Laboratories, Department of Anesthesiology, Columbia University, New York, NY, USA
| | - Ella Novitsky
- Anesthesiology Research Laboratories, Department of Anesthesiology, Columbia University, New York, NY, USA
| | - Vivette D'Agati
- Department of Pathology, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - H Thomas Lee
- Anesthesiology Research Laboratories, Department of Anesthesiology, Columbia University, New York, NY, USA
| |
Collapse
|
10
|
Gao RY, Wang M, Liu Q, Feng D, Wen Y, Xia Y, Colgan SP, Eltzschig HK, Ju C. Hypoxia-Inducible Factor-2α Reprograms Liver Macrophages to Protect Against Acute Liver Injury Through the Production of Interleukin-6. Hepatology 2020; 71:2105-2117. [PMID: 31529728 PMCID: PMC7075728 DOI: 10.1002/hep.30954] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 09/04/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND AIMS Acetaminophen (APAP) overdose represents the most frequent cause of acute liver failure, resulting in death or liver transplantation in more than one third of patients in the United States. The effectiveness of the only antidote, N-acetylcysteine, declines rapidly after APAP ingestion, long before patients are admitted to the clinic with symptoms of severe liver injury. The direct hepatotoxicity of APAP triggers a cascade of innate immune responses that may exacerbate or limit the progression of tissue damage. A better understanding of this complex mechanism will help uncover targets for therapeutic interventions. APPROACH AND RESULTS We observed that APAP challenge caused stabilization of hypoxia-inducible factors (HIFs) in the liver and hepatic macrophages (MΦs), particularly HIF-2α. Genetic deletion of the HIF-2α gene in myeloid cells (HIF-2αmye/- ) markedly exacerbated APAP-induced liver injury (AILI) without affecting APAP bioactivation and detoxification. In contrast, hepatic and serum levels of the hepatoprotective cytokine interleukin 6 (IL-6), its downstream signal transducer and transcription factor 3 activation in hepatocytes, as well as hepatic MΦ IL-6 expression were markedly reduced in HIF-2αmye/- mice compared to wild-type mice post-APAP challenge. In vitro experiments revealed that hypoxia induced IL-6 production in hepatic MΦs and that such induction was abolished in HIF-2α-deleted hepatic MΦs. Restoration of IL-6 by administration of exogenous IL-6 ameliorated AILI in HIF-2αmye/- mice. Finally, IL-6-mediated hepatoprotection against AILI was abolished in hepatocyte-specific IL-6 receptor knockout mice. CONCLUSIONS The data demonstrate that APAP treatment leads to HIF-2α stabilization in hepatic MΦs and that HIF-2α subsequently reprograms hepatic MΦs to produce the hepatoprotective cytokine IL-6, thereby ameliorating AILI.
Collapse
Affiliation(s)
- Rachel Y. Gao
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.,Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Meng Wang
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.,Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Qihui Liu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.,Current address: Biomedical Institute, Zhuhai People’s Hospital, Jinan University, Zhuhai, Guangdong, China
| | - Dechun Feng
- Laboratory of Liver Disease, NIAAA, NIH, Bethesda, MD, USA
| | - Yankai Wen
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Yang Xia
- Department of Biochemistry and Molecular Biology, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Sean P. Colgan
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Holger K. Eltzschig
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Cynthia Ju
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.,Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA.,Correspondence should be addressed to C. J. ()
| |
Collapse
|
11
|
Zhang X, Sun Y, Song D, Diao Y. κ-opioid receptor agonists may alleviate intestinal damage in cardiopulmonary bypass rats by inhibiting the NF-κB/HIF-1α pathway. Exp Ther Med 2020; 20:325-334. [PMID: 32509012 PMCID: PMC7271736 DOI: 10.3892/etm.2020.8685] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 03/05/2020] [Indexed: 02/07/2023] Open
Abstract
The aims of the present study were to investigate the protective effect of a κ-opioid receptor (KOR) agonist on intestinal barrier dysfunction in rats during cardiopulmonary bypass (CPB), as well as to examine the role of NF-κB and the transcription factor hypoxia-inducible factor-1α (HIF-1α) signaling pathway in the regulatory mechanism. A total of 50 rats were randomly divided into five groups, with 10 rats in each group: Sham surgery group (group Sham), CPB surgery group (group CPB), KOR agonist + CPB (group K), KOR agonist + specific KOR antagonist + CBP (group NK) and KOR agonist + NF-κB pathway specific inhibitor + CPB (group NF). Intestinal microcirculation was evaluated to determine intestinal barrier dysfunction in rats following CPB surgery. Hematoxylin and eosin (H&E) staining was used to observe intestinal tissue injury in the rats. ELISA was used to detect the inflammatory factors interleukin (IL)-1β, IL-6, IL10 and tumor necrosis factor-α, and the oxidative stress factors superoxidase dismutase, malondialdehyde and nitric oxide in serum. In addition, ELISA was used to investigate the serum levels of the intestinal damage markers D-lactic acid, diamine oxidase and intestinal fatty acid-binding protein. Western blotting was used to investigate the protein expression levels of tight junction proteins zonula occludens-1 and claudin-1. Furthermore, immunohistochemistry was used to examine intestinal injuries and western blotting was used to detect expression levels of NF-κB/HIF-1α signaling pathway-related proteins. H&E staining results suggested that the KOR agonist alleviated intestinal damage in the CPB model rats. This effect was reversed by the addition of a KOR antagonist. Further investigation of inflammatory and oxidative stress factors using ELISA revealed that the KOR agonist reduced the inflammatory and oxidative stress responses in the intestinal tissues of the CPB model rats. The ELISA results of intestinal damage markers and western blotting results of tight junction protein expression suggested that KOR agonist treatment may alleviate intestinal injury in CPB model rats. In addition, the western blotting and immunohistochemistry results suggested that KOR agonists may decrease the expression levels of NF-κB, p65 and HIF-1α in CPB. Collectively, the present results suggested that KOR agonists are able to ameliorate the intestinal barrier dysfunction in rats undergoing CPB by inhibiting the expression levels of NF-κB/HIF-1α signaling pathway-related proteins.
Collapse
Affiliation(s)
- Xiaoyan Zhang
- Postgraduate Training Base of The General Hospital of Northern Theater Command, Jinzhou Medical University, Jinzhou, Liaoning 121013, P.R. China.,Department of Anesthesia, The General Hospital of Northern Theater Command, Shenyang, Liaoning 110016, P.R. China
| | - Yingjie Sun
- Department of Anesthesia, The General Hospital of Northern Theater Command, Shenyang, Liaoning 110016, P.R. China
| | - Dandan Song
- Department of Anesthesia, The General Hospital of Northern Theater Command, Shenyang, Liaoning 110016, P.R. China
| | - Yugang Diao
- Department of Anesthesia, The General Hospital of Northern Theater Command, Shenyang, Liaoning 110016, P.R. China
| |
Collapse
|
12
|
Dal Ben D, Antonioli L, Lambertucci C, Spinaci A, Fornai M, D'Antongiovanni V, Pellegrini C, Blandizzi C, Volpini R. Approaches for designing and discovering purinergic drugs for gastrointestinal diseases. Expert Opin Drug Discov 2020; 15:687-703. [PMID: 32228110 DOI: 10.1080/17460441.2020.1743673] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Purines finely modulate physiological motor, secretory, and sensory functions in the gastrointestinal tract. Their activity is mediated by the purinergic signaling machinery, including receptors and enzymes regulating their synthesis, release, and degradation. Several gastrointestinal dysfunctions are characterized by alterations affecting the purinergic system. AREAS COVERED The authors provide an overview on the purinergic receptor signaling machinery, the molecules and proteins involved, and a summary of medicinal chemistry efforts aimed at developing novel compounds able to modulate the activity of each player involved in this machinery. The involvement of purinergic signaling in gastrointestinal motor, secretory, and sensory functions and dysfunctions, and the potential therapeutic applications of purinergic signaling modulators, are then described. EXPERT OPINION A number of preclinical and clinical studies demonstrate that the pharmacological manipulation of purinergic signaling represents a viable way to counteract several gastrointestinal diseases. At present, the paucity of purinergic therapies is related to the lack of receptor-subtype-specific agonists and antagonists that are effective in vivo. In this regard, the development of novel therapeutic strategies should be focused to include tools able to control the P1 and P2 receptor expression as well as modulators of the breakdown or transport of purines.
Collapse
Affiliation(s)
- Diego Dal Ben
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino , Camerino, Italy
| | - Luca Antonioli
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa , Pisa, Italy
| | - Catia Lambertucci
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino , Camerino, Italy
| | - Andrea Spinaci
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino , Camerino, Italy
| | - Matteo Fornai
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa , Pisa, Italy
| | - Vanessa D'Antongiovanni
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa , Pisa, Italy
| | | | - Corrado Blandizzi
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa , Pisa, Italy
| | - Rosaria Volpini
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino , Camerino, Italy
| |
Collapse
|
13
|
Shih YRV, Liu M, Kwon SK, Iida M, Gong Y, Sangaj N, Varghese S. Dysregulation of ectonucleotidase-mediated extracellular adenosine during postmenopausal bone loss. SCIENCE ADVANCES 2019; 5:eaax1387. [PMID: 31457100 PMCID: PMC6703860 DOI: 10.1126/sciadv.aax1387] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 07/12/2019] [Indexed: 05/25/2023]
Abstract
Adenosine and its receptors play a key role in bone homeostasis and regeneration. Extracellular adenosine is generated from CD39 and CD73 activity in the cell membrane, through conversion of adenosine triphosphate to adenosine monophosphate (AMP) and AMP to adenosine, respectively. Despite the relevance of CD39/CD73 to bone health, the roles of these enzymes in bona fide skeletal disorders remain unknown. We demonstrate that CD39/CD73 expression and extracellular adenosine levels in the bone marrow are substantially decreased in animals with osteoporotic bone loss. Knockdown of estrogen receptors ESR1 and ESR2 in primary osteoprogenitors and osteoclasts undergoing differentiation showed decreased coexpression of membrane-bound CD39 and CD73 and lower extracellular adenosine. Targeting the adenosine A2B receptor using an agonist attenuated bone loss in ovariectomized mice. Together, these findings suggest a pathological association of purine metabolism with estrogen deficiency and highlight the potential of A2B receptor as a target to treat osteoporosis.
Collapse
Affiliation(s)
- Yu-Ru V. Shih
- Department of Orthopaedic Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Mengqian Liu
- Mechanical Engineering and Materials Science, Duke University, Durham, NC 27708, USA
| | - Seong Keun Kwon
- Department of Otorhinolaryngology–Head and Neck Surgery, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | | | - Ya Gong
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Nivedita Sangaj
- Department of Orthopaedic Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Shyni Varghese
- Department of Orthopaedic Surgery, Duke University Medical Center, Durham, NC 27710, USA
- Mechanical Engineering and Materials Science, Duke University, Durham, NC 27708, USA
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| |
Collapse
|
14
|
Be Aware of Aggregators in the Search for Potential Human ecto-5'-Nucleotidase Inhibitors. Molecules 2018; 23:molecules23081876. [PMID: 30060466 PMCID: PMC6222861 DOI: 10.3390/molecules23081876] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 07/22/2018] [Accepted: 07/26/2018] [Indexed: 01/09/2023] Open
Abstract
Promiscuous inhibition due to aggregate formation has been recognized as a major concern in drug discovery campaigns. Here, we report some aggregators identified in a virtual screening (VS) protocol to search for inhibitors of human ecto-5′-nucleotidase (ecto-5′-NT/CD73), a promising target for several diseases and pathophysiological events, including cancer, inflammation and autoimmune diseases. Four compounds (A, B, C and D), selected from the ZINC-11 database, showed IC50 values in the micromolar range, being at the same time computationally predicted as potential aggregators. To confirm if they inhibit human ecto-5′-NT via promiscuous mechanism, forming aggregates, enzymatic assays were done in the presence of 0.01% (v/v) Triton X-100 and an increase in the enzyme concentration by 10-fold. Under both experimental conditions, these four compounds showed a significant decrease in their inhibitory activities. To corroborate these findings, turbidimetric assays were performed, confirming that they form aggregate species. Additionally, aggregation kinetic studies were done by dynamic light scattering (DLS) for compound C. None of the identified aggregators has been previously reported in the literature. For the first time, aggregation and promiscuous inhibition issues were systematically studied and evaluated for compounds selected by VS as potential inhibitors for human ecto-5′-NT. Together, our results reinforce the importance of accounting for potential false-positive hits acting by aggregation in drug discovery campaigns to avoid misleading assay results.
Collapse
|
15
|
Haskó G, Antonioli L, Cronstein BN. Adenosine metabolism, immunity and joint health. Biochem Pharmacol 2018; 151:307-313. [PMID: 29427624 PMCID: PMC5899962 DOI: 10.1016/j.bcp.2018.02.002] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 02/02/2018] [Indexed: 12/19/2022]
Abstract
The purine nucleoside adenosine is a present in most body fluids where it regulates a wide variety of physiologic and pharmacologic processes. Adenosine mediates its effects through activating 4 G protein-coupled receptors expressed on the cell membrane: A1, A2A, A2B, and A3. The adenosine receptors are widely distributed in the body, and tissues with high expression include immune tissues, cartilage, bone, heart, and brain. Here we review the source and metabolism of adenosine and the role of adenosine in regulating immunity and cartilage biology.
Collapse
Affiliation(s)
- György Haskó
- Department of Anesthesiology, Columbia University, New York, NY 10032, USA
| | - Luca Antonioli
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | | |
Collapse
|
16
|
Infant cardiopulmonary bypass: CD73 kinetics, association with clinical outcomes, and influence on serum adenosine production capacity. Pediatr Res 2018; 83:858-865. [PMID: 29278640 PMCID: PMC5935543 DOI: 10.1038/pr.2017.325] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 12/04/2017] [Indexed: 12/20/2022]
Abstract
BackgroundExtracellular adenine nucleotides contribute to ischemia-reperfusion injury following infant cardiopulmonary bypass (CPB), whereas conversion to adenosine may be protective. Alkaline phosphatase (AP), a key enzyme responsible for this conversion, decreases after infant CPB. Indirect evidence suggests that soluble CD73 may simultaneously increase and partially offset this loss of AP. We sought to measure CD73 levels in infants undergoing CPB and determine its association with adenosine production capacity and postoperative support requirements.MethodsA prospective cohort study of infants ≤120 days of age undergoing CPB. CD73 was measured before CPB and during rewarming. Multivariable modeling evaluated the contributions of CD73/AP to adenosine production capacity and postoperative support requirements.ResultsSerum samples from 85 subjects were analyzed. The median CD73 concentration increased following CPB (95.2 vs. 179.8 ng/ml; P<0.0001). Rewarming CD73 was independently inversely associated with vasoactive inotropic support (P<0.005) and length of intensive care unit stay (P<0.005). Combined AP activity and CD73 concentration predicted adenosine production capacity (P<0.0001).ConclusionsSerum CD73 increases following infant CPB. Low rewarming CD73 is independently associated with increased postoperative support requirements. CD73 and AP together predict serum adenosine production capacity and may represent potential therapeutic targets to clear extracellular adenine nucleotides and improve outcomes following infant CPB.
Collapse
|
17
|
Yuan X, Lee JW, Bowser JL, Neudecker V, Sridhar S, Eltzschig HK. Targeting Hypoxia Signaling for Perioperative Organ Injury. Anesth Analg 2018; 126:308-321. [PMID: 28759485 PMCID: PMC5735013 DOI: 10.1213/ane.0000000000002288] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Perioperative organ injury has a significant impact on surgical outcomes and presents a leading cause of death in the United States. Recent research has pointed out an important role of hypoxia signaling in the protection from organ injury, including for example myocardial infarction, acute respiratory distress syndrome, acute kidney, or gut injury. Hypoxia induces the stabilization of hypoxia-inducible factors (HIFs), thereby leading to the induction of HIF target genes, which facilitates adaptive responses to low oxygen. In this review, we focus on current therapeutic strategies targeting hypoxia signaling in various organ injury models and emphasize potential clinical approaches to integrate these findings into the care of surgical patients. Conceptually, there are 2 options to target the HIF pathway for organ protection. First, drugs became recently available that promote the stabilization of HIFs, most prominently via inhibition of prolyl hydroxylase. These compounds are currently trialed in patients, for example, for anemia treatment or prevention of ischemia and reperfusion injury. Second, HIF target genes (such as adenosine receptors) could be activated directly. We hope that some of these approaches may lead to novel pharmacologic strategies to prevent or treat organ injury in surgical patients.
Collapse
Affiliation(s)
- Xiaoyi Yuan
- Department of Anesthesiology, the University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Jae W. Lee
- Department of Anesthesiology, the University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Jessica L. Bowser
- Department of Anesthesiology, the University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Viola Neudecker
- Department of Anesthesiology, Clinic of the University of Munich, Munich, Germany
| | - Srikanth Sridhar
- Department of Anesthesiology, the University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Holger K. Eltzschig
- Department of Anesthesiology, the University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| |
Collapse
|
18
|
Dou L, Chen YF, Cowan PJ, Chen XP. Extracellular ATP signaling and clinical relevance. Clin Immunol 2017; 188:67-73. [PMID: 29274390 DOI: 10.1016/j.clim.2017.12.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 12/12/2017] [Accepted: 12/20/2017] [Indexed: 12/20/2022]
Abstract
Since purinergic signaling was discovered in the early 1970s, it has been shown that extracellular nucleotides, and their derivative nucleosides, are released in a regulated or unregulated manner by cells in various challenging settings and then bind defined purinergic receptors to activate intricate signaling networks. Extracellular ATP plays a role based on different P2 receptor subtypes expressed on specific cell types. Sequential hydrolysis of extracellular ATP catalyzed by ectonucleotidases (e.g. CD39, CD73) is the main pathway for the generation of adenosine, which in turn activates P1 receptors. Many studies have demonstrated that extracellular ATP signaling functions as an important dynamic regulatory pathway to coordinate appropriate immune responses in various pathological processes, including intracellular infection, host-tumor interaction, pro-inflammation vascular injury, and transplant immunity. ATP receptors and CD39 also participate in related clinical settings. Here, we review the latest research in to the development of promising clinical treatment strategies.
Collapse
Affiliation(s)
- Lei Dou
- Department of Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Gerontology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi-Fa Chen
- Department of Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Peter J Cowan
- Immunology Research Centre, St Vincent's Hospital, Melbourne, Australia.
| | - Xiao-Ping Chen
- Department of Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Gerontology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
19
|
Oxidative stress inactivates ecto-5'-nucleotidase by inhibiting protein kinase C in rat hearts in vivo. Eur J Pharmacol 2017; 805:125-130. [DOI: 10.1016/j.ejphar.2017.02.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 01/30/2017] [Accepted: 02/13/2017] [Indexed: 12/25/2022]
|
20
|
Sung SSJ, Li L, Huang L, Lawler J, Ye H, Rosin DL, Vincent IS, Le TH, Yu J, Görldt N, Schrader J, Okusa MD. Proximal Tubule CD73 Is Critical in Renal Ischemia-Reperfusion Injury Protection. J Am Soc Nephrol 2017; 28:888-902. [PMID: 27628903 PMCID: PMC5328157 DOI: 10.1681/asn.2016020229] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 08/17/2016] [Indexed: 11/03/2022] Open
Abstract
CD73-derived adenosine plays an anti-inflammatory role in various organs. However, its role in renal ischemia-reperfusion injury (IRI) is controversial. We targeted CD73 mutant mice to determine the function of CD73 expressed by various renal cell types under mild IRI conditions. Mice with CD73 deletion in proximal tubules exhibited exacerbated IRI, comparable with that of CD73-/- mice compared with WT mice. Mice with CD73 deletions in other cell types, including cortical type 1 fibroblast-like cells, mesangial cells, macrophages, and dendritic cells, showed small or no increases in injury above control mice when subjected to threshold levels of ischemia. Results from adoptive transfer experiments between WT and CD73-/- mice and pharmacologic studies modulating enzymatic activity of CD73 and extracellular adenosine levels supported a critical role of adenosine generated by proximal tubule CD73 expression in abrogating IRI. Renal adenosine levels were lower before and after ischemia in CD73-deficient mice. However, reduction in total acid-extractable renal adenosine levels was inadequate to explain the marked difference in kidney injury in these CD73-deficient mice. Furthermore, CD73 inhibition and enzyme replacement studies showed no change in total kidney adenosine levels in treated mice compared with vehicle-treated controls. Protection from IRI in neutrophil-depleted WT recipients was sustained by repopulation with bone marrow neutrophils from WT mice but not by those lacking adenosine 2a receptors (from Adora2a-/- mice). These data support the thesis that local adenosine generated by cells at the injury site is critical for protection from IRI through bone marrow-derived adenosine 2a receptors.
Collapse
Affiliation(s)
- Sun-Sang J Sung
- Division of Nephrology and
- Center for Immunity, Inflammation, and Regenerative Medicine, Department of Medicine and
| | - Li Li
- Division of Nephrology and
- Center for Immunity, Inflammation, and Regenerative Medicine, Department of Medicine and
| | - Liping Huang
- Division of Nephrology and
- Center for Immunity, Inflammation, and Regenerative Medicine, Department of Medicine and
| | - Jessica Lawler
- Division of Nephrology and
- Center for Immunity, Inflammation, and Regenerative Medicine, Department of Medicine and
| | - Hong Ye
- Division of Nephrology and
- Center for Immunity, Inflammation, and Regenerative Medicine, Department of Medicine and
| | - Diane L Rosin
- Center for Immunity, Inflammation, and Regenerative Medicine, Department of Medicine and
- Departments of Pharmacology and
| | - Issah S Vincent
- Division of Nephrology and
- Center for Immunity, Inflammation, and Regenerative Medicine, Department of Medicine and
| | - Thu H Le
- Division of Nephrology and
- Center for Immunity, Inflammation, and Regenerative Medicine, Department of Medicine and
| | - Jing Yu
- Cell Biology, University of Virginia, Charlottesville, Virginia; and
| | - Nicole Görldt
- Department of Molecular Cardiology, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| | - Jürgen Schrader
- Department of Molecular Cardiology, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| | - Mark D Okusa
- Division of Nephrology and
- Center for Immunity, Inflammation, and Regenerative Medicine, Department of Medicine and
| |
Collapse
|
21
|
Tak E, Jung DH, Kim SH, Park GC, Jun DY, Lee J, Jung BH, Kirchner VA, Hwang S, Song GW, Lee SG. Protective role of hypoxia-inducible factor-1α-dependent CD39 and CD73 in fulminant acute liver failure. Toxicol Appl Pharmacol 2016; 314:72-81. [PMID: 27899277 DOI: 10.1016/j.taap.2016.11.016] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 11/16/2016] [Accepted: 11/25/2016] [Indexed: 12/21/2022]
Abstract
Acute liver failure (ALF) is a severe life-threatening disease which usually arises in patients with-irreversible liver illnesses. Although human ectonucleotide triphosphate diphosphohydrolase-1, E-NTPDase1 (CD39) and ecto-5'-nucleotidase, Ecto5'NTase (CD73) are known to protect tissues from ALF, the expression and function of CD39 and CD73 during ALF are currently not fully investigated. We tested whether CD39 and CD73 are upregulated by hypoxia inducible factor (HIF)-1α, and improve ischemic tolerance to ALF. To test our hypothesis, liver biopsies were obtained and we found that CD39 and CD73 mRNA and proteins from human specimens were dramatically elevated in ALF. We investigated that induction of CD39 and CD73 in ALF-related with wild type mice. In contrast, deletion of cd39 and cd73 mice has severe ALF. In this study, we concluded that CD39 and CD73 are molecular targets for the development of drugs for ALF patients care.
Collapse
Affiliation(s)
- Eunyoung Tak
- Asan Institute for Life Sciences and Asan-Minnesota Institute for Innovating Transplantation, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Dong-Hwan Jung
- Division of Liver Transplantation and Hepatobiliary Surgery, Asan-Minnesota Institute for Innovating Transplantation, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Seok-Hwan Kim
- Division of Liver Transplantation and Hepatobiliary Surgery, Asan-Minnesota Institute for Innovating Transplantation, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Gil-Chun Park
- Division of Liver Transplantation and Hepatobiliary Surgery, Asan-Minnesota Institute for Innovating Transplantation, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Dae Young Jun
- Asan Institute for Life Sciences and Asan-Minnesota Institute for Innovating Transplantation, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jooyoung Lee
- Asan Institute for Life Sciences and Asan-Minnesota Institute for Innovating Transplantation, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Bo-Hyun Jung
- Department of Surgery, Inje University Haeundae Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea
| | - Varvara A Kirchner
- Division of Transplantation, Department of Surgery and Asan-Minnesota Institute for Innovating Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Shin Hwang
- Division of Liver Transplantation and Hepatobiliary Surgery, Asan-Minnesota Institute for Innovating Transplantation, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Gi-Won Song
- Division of Liver Transplantation and Hepatobiliary Surgery, Asan-Minnesota Institute for Innovating Transplantation, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| | - Sung-Gyu Lee
- Division of Liver Transplantation and Hepatobiliary Surgery, Asan-Minnesota Institute for Innovating Transplantation, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
22
|
Ju C, Colgan SP, Eltzschig HK. Hypoxia-inducible factors as molecular targets for liver diseases. J Mol Med (Berl) 2016; 94:613-27. [PMID: 27094811 PMCID: PMC4879168 DOI: 10.1007/s00109-016-1408-1] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 03/04/2016] [Accepted: 03/08/2016] [Indexed: 12/11/2022]
Abstract
Liver disease is a growing global health problem, as deaths from end-stage liver cirrhosis and cancer are rising across the world. At present, pharmacologic approaches to effectively treat or prevent liver disease are extremely limited. Hypoxia-inducible factor (HIF) is a transcription factor that regulates diverse signaling pathways enabling adaptive cellular responses to perturbations of the tissue microenvironment. HIF activation through hypoxia-dependent and hypoxia-independent signals have been reported in liver disease of diverse etiologies, from ischemia-reperfusion-induced acute liver injury to chronic liver diseases caused by viral infection, excessive alcohol consumption, or metabolic disorders. This review summarizes the evidence for HIF stabilization in liver disease, discusses the mechanistic involvement of HIFs in disease development, and explores the potential of pharmacological HIF modifiers in the treatment of liver disease.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/therapeutic use
- Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors
- Basic Helix-Loop-Helix Transcription Factors/genetics
- Basic Helix-Loop-Helix Transcription Factors/metabolism
- Bevacizumab/therapeutic use
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/therapy
- Clinical Trials as Topic
- Fatty Liver/genetics
- Fatty Liver/metabolism
- Fatty Liver/pathology
- Fatty Liver/therapy
- Gene Expression Regulation
- Hepatitis, Viral, Human/genetics
- Hepatitis, Viral, Human/metabolism
- Hepatitis, Viral, Human/pathology
- Hepatitis, Viral, Human/therapy
- Humans
- Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Liver Cirrhosis/genetics
- Liver Cirrhosis/metabolism
- Liver Cirrhosis/pathology
- Liver Cirrhosis/therapy
- Liver Diseases, Alcoholic/genetics
- Liver Diseases, Alcoholic/metabolism
- Liver Diseases, Alcoholic/pathology
- Liver Diseases, Alcoholic/therapy
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Liver Neoplasms/therapy
- Molecular Targeted Therapy
- Oligonucleotides/therapeutic use
- Signal Transduction
Collapse
Affiliation(s)
- Cynthia Ju
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy, University of Colorado, Auroa, Colorado, 800045, USA.
| | - Sean P Colgan
- Department of Medicine and Mucosal Inflammation Program, School of Medicine, University of Colorado, Auroa, Colorado, 800045, USA
| | - Holger K Eltzschig
- Department of Anesthesiology and Organ Protection Program, School of Medicine, University of Colorado, Auroa, Colorado, 800045, USA
| |
Collapse
|
23
|
Inhibition of P38 MAPK Downregulates the Expression of IL-1β to Protect Lung from Acute Injury in Intestinal Ischemia Reperfusion Rats. Mediators Inflamm 2016; 2016:9348037. [PMID: 26980948 PMCID: PMC4766341 DOI: 10.1155/2016/9348037] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 12/27/2015] [Accepted: 12/29/2015] [Indexed: 02/05/2023] Open
Abstract
Acute lung injury (ALI) induced by intestinal ischemia/reperfusion (II/R) has high incidence and mortality, in which IL-1β was essential for the full development of ALI. However, the detailed regulating mechanism for this phenomenon remains to be unclear. The purpose of this study was to investigate whether inhibition of P38 MAPK could downregulate the expression of IL-1β to protect lung from acute injury in II/R rats. Here, we found that the level of pulmonary edema at 16 hours after operation (hpo) was obviously enhanced compared to that in 8hpo and sham groups. Immunofluorescent staining demonstrated that IL-1β and P38 MAPK were detected in lung tissues. And rats with II/R have the highest translation level for IL-1β and phosphorylation of P38 MAPK in lung tissues at 16hpo compared with 8hpo and sham groups. Moreover, administration of SB239063, an inhibitor of P38 α and β, could effectively downregulate the expressions of IL-1β and protects lung tissues from injury in II/R rats. Our findings indicate that the inhibition of P38 α and β may downregulate the expression of IL-1β to protect lung from acute injury in II/R, which could be used as a potential target for reducing ALI induced by II/R in the future clinical trial.
Collapse
|
24
|
Wang B, Wan J, Gong X, Kuang G, Cheng X, Min S. Mangiferin attenuates renal ischemia-reperfusion injury by inhibiting inflammation and inducing adenosine production. Int Immunopharmacol 2015; 25:148-54. [DOI: 10.1016/j.intimp.2014.11.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 11/03/2014] [Accepted: 11/10/2014] [Indexed: 12/19/2022]
|
25
|
Harnoss JM, Strowitzki MJ, Radhakrishnan P, Platzer LK, Harnoss JC, Hank T, Cai J, Ulrich A, Schneider M. Therapeutic inhibition of prolyl hydroxylase domain-containing enzymes in surgery: putative applications and challenges. HYPOXIA 2015; 3:1-14. [PMID: 27774478 PMCID: PMC5045068 DOI: 10.2147/hp.s60872] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Oxygen is essential for metazoans to generate energy. Upon oxygen deprivation adaptive and protective pathways are induced, mediated by hypoxia-inducible factors (HIFs) and prolyl hydroxylase domain-containing enzymes (PHDs). Both play a pivotal role in various conditions associated with prolonged ischemia and inflammation, and are promising targets for therapeutic intervention. This review focuses on aspects of therapeutic PHD modulation in surgically relevant disease conditions such as hepatic and intestinal disorders, wound healing, innate immune responses, and tumorigenesis, and discusses the therapeutic potential and challenges of PHD inhibition in surgical patients.
Collapse
Affiliation(s)
- Jonathan Michael Harnoss
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Moritz Johannes Strowitzki
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Praveen Radhakrishnan
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Lisa Katharina Platzer
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Julian Camill Harnoss
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Thomas Hank
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Jun Cai
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Alexis Ulrich
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Martin Schneider
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
26
|
Bartels K, Grenz A, Eltzschig HK. Sphingosine-1-phosphate receptor signaling during acute kidney injury: the tissue is the issue. Kidney Int 2014; 85:733-5. [PMID: 24682118 PMCID: PMC4007344 DOI: 10.1038/ki.2013.435] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Sphingosine-1-phospate is a lipid mediator that has been implicated in
protection from acute kidney injury (AKI) by activation of the
sphingosine-1-phosphate receptor S1P1R. A recent study demonstrates
that mice with induced deletion of S1P1R on endothelial cells experience
increased ischemia-induced AKI. These findings have important translational
implications. Indeed, S1P1R agonists have been used for the treatment of
patients suffering from autoimmune encephalitis. As such, endothelial
S1P1R-signaling could be targeted for AKI prevention in surgical
patients.
Collapse
Affiliation(s)
- Karsten Bartels
- Organ Protection Program, Department of Anesthesiology, University of Colorado, Aurora, Colorado, USA
| | - Almut Grenz
- Organ Protection Program, Department of Anesthesiology, University of Colorado, Aurora, Colorado, USA
| | - Holger K Eltzschig
- Organ Protection Program, Department of Anesthesiology, University of Colorado, Aurora, Colorado, USA
| |
Collapse
|
27
|
Chiu GS, Freund GG. Modulation of neuroimmunity by adenosine and its receptors: metabolism to mental illness. Metabolism 2014; 63:1491-8. [PMID: 25308443 PMCID: PMC4252699 DOI: 10.1016/j.metabol.2014.09.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2014] [Revised: 08/14/2014] [Accepted: 09/19/2014] [Indexed: 12/20/2022]
Abstract
Adenosine is a pleiotropic bioactive with potent neuromodulatory properties. Due to its ability to easily cross the blood-brain barrier, it can act as a signaling molecule between the periphery and the brain. It functions through four (A1, A2A, A2B, and A3) cell surface G protein-coupled adenosine receptors (ARs) that are expressed in some combination on nearly all cells types within the CNS. By regulating the activity of adenylyl cyclase and changing the intracellular concentration of cAMP, adenosine can alter neuronal function and neurotransmission. A variety of illnesses related to metabolic dysregulation, such as type 1 diabetes and Alzheimer's disease, are associated with an elevated serum concentration of adenosine and a pathogenesis rooted in inflammation. This review describes the accepted physiologic function of adenosine in neurological disease and explores its new potential as a peripheral to central danger signal that can activate the neuroimmune system and contribute to symptoms of sickness and psychopathologies.
Collapse
Affiliation(s)
- Gabriel S Chiu
- Division of Nutritional Sciences, University of Illinois, Urbana IL, USA; Department of Pathology, Program in Integrative Immunology and Behavior, University of Illinois, Urbana IL, USA
| | - Gregory G Freund
- Division of Nutritional Sciences, University of Illinois, Urbana IL, USA; Department of Pathology, Program in Integrative Immunology and Behavior, University of Illinois, Urbana IL, USA; Department of Animal Sciences, University of Illinois, Urbana IL, USA.
| |
Collapse
|
28
|
Csóka B, Németh ZH, Törő G, Koscsó B, Kókai E, Robson SC, Enjyoji K, Rolandelli RH, Erdélyi K, Pacher P, Haskó G. CD39 improves survival in microbial sepsis by attenuating systemic inflammation. FASEB J 2014; 29:25-36. [PMID: 25318479 DOI: 10.1096/fj.14-253567] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Sepsis remains the leading cause of morbidity and mortality in critically ill patients. Excessive inflammation is a major cause of organ failure and mortality in sepsis. Ectonucleoside triphosphate diphosphohydrolase 1, ENTPDase1 (CD39) is a cell surface nucleotide-metabolizing enzyme, which degrades the extracellular purines ATP and ADP, thereby regulating purinergic receptor signaling. Although the role of purinergic receptor signaling in regulating inflammation and sepsis has been addressed previously, the role of CD39 in regulating the host's response to sepsis is unknown. We found that the CD39 mimic apyrase (250 U/kg) decreased and knockout or pharmacologic blockade with sodium polyoxotungstate (5 mg/kg; IC50 ≈ 10 μM) of CD39 increased mortality of mice with polymicrobial sepsis induced by cecal ligation and puncture. CD39 decreased inflammation, organ damage, immune cell apoptosis, and bacterial load. Use of bone marrow chimeric mice revealed that CD39 expression on myeloid cells decreases inflammation in septic mice. CD39 expression is upregulated during sepsis in mice, as well as in both murine and human macrophages stimulated with Escherichia coli. Moreover, E. coli increases CD39 promoter activity in macrophages. Altogether, these data indicate CD39 as an evolutionarily conserved inducible protective pathway during sepsis. We propose CD39 as a novel therapeutic target in the management of sepsis.
Collapse
Affiliation(s)
- Balázs Csóka
- Department of Surgery and Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | | | | | - Balázs Koscsó
- Department of Surgery and Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | | | - Simon C Robson
- Department of Medicine, Gastroenterology and Transplant Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA; and
| | - Keiichi Enjyoji
- Department of Medicine, Gastroenterology and Transplant Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA; and
| | | | - Katalin Erdélyi
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA
| | - Pál Pacher
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA
| | - György Haskó
- Department of Surgery and Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, New Jersey, USA;
| |
Collapse
|
29
|
Idzko M, Ferrari D, Riegel AK, Eltzschig HK. Extracellular nucleotide and nucleoside signaling in vascular and blood disease. Blood 2014; 124:1029-37. [PMID: 25001468 PMCID: PMC4133480 DOI: 10.1182/blood-2013-09-402560] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2013] [Accepted: 06/16/2014] [Indexed: 12/17/2022] Open
Abstract
Nucleotides and nucleosides-such as adenosine triphosphate (ATP) and adenosine-are famous for their intracellular roles as building blocks for the genetic code or cellular energy currencies. In contrast, their function in the extracellular space is different. Here, they are primarily known as signaling molecules via activation of purinergic receptors, classified as P1 receptors for adenosine or P2 receptors for ATP. Because extracellular ATP is rapidly converted to adenosine by ectonucleotidase, nucleotide-phosphohydrolysis is important for controlling the balance between P2 and P1 signaling. Gene-targeted mice for P1, P2 receptors, or ectonucleotidase exhibit only very mild phenotypic manifestations at baseline. However, they demonstrate alterations in disease susceptibilities when exposed to a variety of vascular or blood diseases. Examples of phenotypic manifestations include vascular barrier dysfunction, graft-vs-host disease, platelet activation, ischemia, and reperfusion injury or sickle cell disease. Many of these studies highlight that purinergic signaling events can be targeted therapeutically.
Collapse
Affiliation(s)
- Marco Idzko
- Department of Pneumology, Freiburg University Medical Center, Albert-Ludwigs-University, Freiburg, Germany
| | - Davide Ferrari
- Department of Morphology, Surgery and Experimental Medicine, Section of General Pathology, Oncology and Experimental Biology, University of Ferrara, Ferrara, Italy; and
| | - Ann-Kathrin Riegel
- Organ Protection Program, Department of Anesthesiology, University of Colorado School of Medicine, Aurora, CO
| | - Holger K Eltzschig
- Organ Protection Program, Department of Anesthesiology, University of Colorado School of Medicine, Aurora, CO
| |
Collapse
|
30
|
Akhtar MZ, Sutherland AI, Huang H, Ploeg RJ, Pugh CW. The role of hypoxia-inducible factors in organ donation and transplantation: the current perspective and future opportunities. Am J Transplant 2014; 14:1481-7. [PMID: 24909061 DOI: 10.1111/ajt.12737] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 02/11/2014] [Accepted: 03/07/2014] [Indexed: 01/25/2023]
Abstract
Hypoxia-inducible factors are the universal cellular oxygen-sensitive transcription factors that activate a number of hypoxia responsive genes, some of which are responsible for protective cellular functions. During organ donation, allografts are exposed to significant periods of hypoxia and ischemia. Exploiting this pathway during donor management and organ preservation could prevent and reduce allograft injury and improve the outcomes of organ transplantation. We review the evidence on this pathway in organ preservation, drawing on experimental studies on donor management and ischemia reperfusion injury focusing on kidney, liver, cardiac and lung transplantation. We review the major technical and experimental challenges in exploring this pathway and suggest potential future avenues for research.
Collapse
Affiliation(s)
- M Z Akhtar
- Nuffield Department of Surgical Sciences, Oxford Transplant Centre, University of Oxford, Oxford, UK; Centre for Cellular and Molecular Physiology, Old Road Campus, University of Oxford, Oxford, UK
| | | | | | | | | |
Collapse
|
31
|
Kim M, Ham A, Kim KYM, Brown KM, Lee HT. The volatile anesthetic isoflurane increases endothelial adenosine generation via microparticle ecto-5'-nucleotidase (CD73) release. PLoS One 2014; 9:e99950. [PMID: 24945528 PMCID: PMC4063779 DOI: 10.1371/journal.pone.0099950] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 02/18/2014] [Indexed: 11/26/2022] Open
Abstract
Endothelial dysfunction is common in acute and chronic organ injury. Isoflurane is a widely used halogenated volatile anesthetic during the perioperative period and protects against endothelial cell death and inflammation. In this study, we tested whether isoflurane induces endothelial ecto-5′-nucleotidase (CD73) and cytoprotective adenosine generation to protect against endothelial cell injury. Clinically relevant concentrations of isoflurane induced CD73 activity and increased adenosine generation in cultured human umbilical vein or mouse glomerular endothelial cells. Surprisingly, isoflurane-mediated induction of endothelial CD73 activity occurred within 1 hr and without synthesizing new CD73. We determined that isoflurane rapidly increased CD73 containing endothelial microparticles into the cell culture media. Indeed, microparticles isolated from isoflurane-treated endothelial cells had significantly higher CD73 activity as well as increased CD73 protein. In vivo, plasma from mice anesthetized with isoflurane had significantly higher endothelial cell-derived CD144+ CD73+ microparticles and had increased microparticle CD73 activity compared to plasma from pentobarbital-anesthetized mice. Supporting a critical role of CD73 in isoflurane-mediated endothelial protection, a selective CD73 inhibitor (APCP) prevented isoflurane-induced protection against human endothelial cell inflammation and apoptosis. In addition, isoflurane activated endothelial cells Rho kinase evidenced by myosin phosphatase target subunit-1 and myosin light chain phosphorylation. Furthermore, isoflurane-induced release of CD73 containing microparticles was significantly attenuated by a selective Rho kinase inhibitor (Y27632). Taken together, we conclude that the volatile anesthetic isoflurane causes Rho kinase-mediated release of endothelial microparticles containing preformed CD73 and increase adenosine generation to protect against endothelial apoptosis and inflammation.
Collapse
Affiliation(s)
- Mihwa Kim
- Department of Anesthesiology, College of Physicians and Surgeons of Columbia University, New York, New York, United States of America
| | - Ahrom Ham
- Department of Anesthesiology, College of Physicians and Surgeons of Columbia University, New York, New York, United States of America
| | - Katelyn Yu-Mi Kim
- Department of Anesthesiology, College of Physicians and Surgeons of Columbia University, New York, New York, United States of America
| | - Kevin M. Brown
- Department of Anesthesiology, College of Physicians and Surgeons of Columbia University, New York, New York, United States of America
| | - H. Thomas Lee
- Department of Anesthesiology, College of Physicians and Surgeons of Columbia University, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
32
|
Yang Y, Qiu Y, Wang W, Xiao W, Liang H, Zhang C, Yang H, Teitelbaum DH, Sun LH, Yang H. Adenosine A2B receptor modulates intestinal barrier function under hypoxic and ischemia/reperfusion conditions. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2014; 7:2006-2018. [PMID: 24966910 PMCID: PMC4069946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 04/02/2014] [Indexed: 06/03/2023]
Abstract
BACKGROUND Intestinal barrier function failure from ischemia/reperfusion (I/R) and acute hypoxia has been implicated as a critical determinant in the predisposition to intestinal inflammation and a number of inflammatory disorders. Here, we identified the role of Adenosine A2B receptor (A2BAR) in the regulation of intestinal barrier function under I/R and acute hypoxic conditions. METHODS C57BL/6J mice were used, and were randomized into three groups: Sham, I/R, IR+PSB1115 (a specific A2BAR antagonist) groups. After surgery, the small bowel was harvested for immunohistochemical staining, RNA and protein content, and intestinal permeability analyses. Using an epithelial cell culture model, we investigated the influence of hypoxia on the epithelial function, and the role of A2BAR in the expressions of tight junction and epithelial permeability. The expressions of Claudin-1, occludin and ZO-1 were detected by RT-PCR and Western-Blot. Epithelial barrier function was assessed with transepithelial resistance (TER). RESULTS AND CONCLUSIONS The A2BAR antagonist, PSB1115, significantly increased tight junction protein expression after intestinal I/R or acute hypoxia conditions. PSB1115 also attenuated the disrupted distribution of TJ proteins. Furthermore, inhibition of A2BAR attenuated the decrease in TER induced by I/R or acute hypoxic conditions, and maintained intestinal barrier function. Antagonism of A2BAR activity improves intestinal epithelial structure and barrier function in a mouse model of intestinal I/R and a cell model of acute hypoxia. These findings support a potentially destructive role for A2BAR under intestinal I/R and acute hypoxic conditions.
Collapse
Affiliation(s)
- Yang Yang
- Department of General Surgery, Xinqiao Hospital, Third Military Medical UniversityChongqing, China
| | - Yuan Qiu
- Department of General Surgery, Xinqiao Hospital, Third Military Medical UniversityChongqing, China
| | - Wensheng Wang
- Department of General Surgery, Xinqiao Hospital, Third Military Medical UniversityChongqing, China
| | - Weidong Xiao
- Department of General Surgery, Xinqiao Hospital, Third Military Medical UniversityChongqing, China
| | - Hongyin Liang
- Department of General Surgery, Xinqiao Hospital, Third Military Medical UniversityChongqing, China
| | - Chaojun Zhang
- Department of General Surgery, Xinqiao Hospital, Third Military Medical UniversityChongqing, China
| | | | | | - Li-Hua Sun
- Department of General Surgery, Xinqiao Hospital, Third Military Medical UniversityChongqing, China
| | - Hua Yang
- Department of General Surgery, Xinqiao Hospital, Third Military Medical UniversityChongqing, China
| |
Collapse
|
33
|
Bartels K, Karhausen J, Clambey ET, Grenz A, Eltzschig HK. Perioperative organ injury. Anesthesiology 2014; 119:1474-89. [PMID: 24126264 DOI: 10.1097/aln.0000000000000022] [Citation(s) in RCA: 125] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Despite the fact that a surgical procedure may have been performed for the appropriate indication and in a technically perfect manner, patients are threatened by perioperative organ injury. For example, stroke, myocardial infarction, acute respiratory distress syndrome, acute kidney injury, or acute gut injury are among the most common causes for morbidity and mortality in surgical patients. In the current review, the authors discuss the pathogenesis of perioperative organ injury, and provide select examples for novel treatment concepts that have emerged over the past decade. Indeed, the authors are of the opinion that research to provide mechanistic insight into acute organ injury and identification of novel therapeutic approaches for the prevention or treatment of perioperative organ injury represent the most important opportunity to improve outcomes of anesthesia and surgery.
Collapse
Affiliation(s)
- Karsten Bartels
- * Fellow in Critical Care Medicine and Cardiothoracic Anesthesiology, † Assistant Professor of Anesthesiology, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina. ‡ Assistant Professor of Anesthesiology, § Associate Professor of Anesthesiology, ‖ Professor of Anesthesiology, Department of Anesthesiology, University of Colorado Denver, Aurora, Colorado
| | | | | | | | | |
Collapse
|
34
|
Burnstock G, Ralevic V. Purinergic signaling and blood vessels in health and disease. Pharmacol Rev 2013; 66:102-92. [PMID: 24335194 DOI: 10.1124/pr.113.008029] [Citation(s) in RCA: 227] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Purinergic signaling plays important roles in control of vascular tone and remodeling. There is dual control of vascular tone by ATP released as a cotransmitter with noradrenaline from perivascular sympathetic nerves to cause vasoconstriction via P2X1 receptors, whereas ATP released from endothelial cells in response to changes in blood flow (producing shear stress) or hypoxia acts on P2X and P2Y receptors on endothelial cells to produce nitric oxide and endothelium-derived hyperpolarizing factor, which dilates vessels. ATP is also released from sensory-motor nerves during antidromic reflex activity to produce relaxation of some blood vessels. In this review, we stress the differences in neural and endothelial factors in purinergic control of different blood vessels. The long-term (trophic) actions of purine and pyrimidine nucleosides and nucleotides in promoting migration and proliferation of both vascular smooth muscle and endothelial cells via P1 and P2Y receptors during angiogenesis and vessel remodeling during restenosis after angioplasty are described. The pathophysiology of blood vessels and therapeutic potential of purinergic agents in diseases, including hypertension, atherosclerosis, ischemia, thrombosis and stroke, diabetes, and migraine, is discussed.
Collapse
Affiliation(s)
- Geoffrey Burnstock
- Autonomic Neuroscience Centre, University College Medical School, Rowland Hill Street, London NW3 2PF, UK; and Department of Pharmacology, The University of Melbourne, Australia.
| | | |
Collapse
|
35
|
Tak E, Ridyard D, Kim JH, Zimmerman M, Werner T, Wang XX, Shabeka U, Seo SW, Christians U, Klawitter J, Moldovan R, Garcia G, Levi M, Haase V, Ravid K, Eltzschig HK, Grenz A. CD73-dependent generation of adenosine and endothelial Adora2b signaling attenuate diabetic nephropathy. J Am Soc Nephrol 2013; 25:547-63. [PMID: 24262796 DOI: 10.1681/asn.2012101014] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Nucleotide phosphohydrolysis by the ecto-5'-nucleotidase (CD73) is the main source for extracellular generation of adenosine. Extracellular adenosine subsequently signals through four distinct adenosine A receptors (Adora1, Adora2a, Adora2b, or Adora3). Here, we hypothesized a functional role for CD73-dependent generation and concomitant signaling of extracellular adenosine during diabetic nephropathy. CD73 transcript and protein levels were elevated in the kidneys of diabetic mice. Genetic deletion of CD73 was associated with more severe diabetic nephropathy, whereas treatment with soluble nucleotidase was therapeutic. Transcript levels of renal adenosine receptors showed a selective induction of Adora2b during diabetic nephropathy. In a transgenic reporter mouse, Adora2b expression localized to the vasculature and increased after treatment with streptozotocin. Adora2b(-/-) mice experienced more severe diabetic nephropathy, and studies in mice with tissue-specific deletion of Adora2b in tubular epithelia or vascular endothelia implicated endothelial Adora2b signaling in protection from diabetic nephropathy. Finally, treatment with a selective Adora2b agonist (BAY 60-6583) conveyed potent protection from diabetes-associated kidney disease. Taken together, these findings implicate CD73-dependent production of extracellular adenosine and endothelial Adora2b signaling in kidney protection during diabetic nephropathy.
Collapse
|
36
|
Zimmerman MA, Kam I, Eltzschig H, Grenz A. Biological implications of extracellular adenosine in hepatic ischemia and reperfusion injury. Am J Transplant 2013; 13:2524-9. [PMID: 23924168 PMCID: PMC3805691 DOI: 10.1111/ajt.12398] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Revised: 06/17/2013] [Accepted: 06/18/2013] [Indexed: 01/25/2023]
Abstract
The purine nucleoside adenosine is clinically employed in the treatment of supraventricular tachycardia. In addition, it has direct coronary vasodilatory effects, and may influence platelet aggregation. Experimental observations mechanistically link extracellular adenosine to cellular adaptation to hypoxia. Adenosine generation has been implicated in several pathophysiologic processes including angiogenesis, tumor defenses and neurodegeneration. In solid organ transplantation, prolonged tissue ischemia and subsequent reperfusion injury may lead to profound graft dysfunction. Importantly, conditions of limited oxygen availability are associated with increased production of extracellular adenosine and subsequent tissue protection. Within the rapidly expanding field of adenosine biology, several enzymatic steps in adenosine production have been characterized and multiple receptor subtypes have been identified. In this review, we briefly examine the biologic steps involved in adenosine generation and chronicle the current state of adenosine signaling in hepatic ischemia and reperfusion injury.
Collapse
Affiliation(s)
- M A Zimmerman
- Division of Transplant Surgery, and the Mucosal Inflammation Program, University of Colorado, Denver, CO
| | | | | | | |
Collapse
|
37
|
Eckle T, Brodsky K, Bonney M, Packard T, Han J, Borchers CH, Mariani TJ, Kominsky DJ, Mittelbronn M, Eltzschig HK. HIF1A reduces acute lung injury by optimizing carbohydrate metabolism in the alveolar epithelium. PLoS Biol 2013; 11:e1001665. [PMID: 24086109 DOI: 10.1371/journal.pbio.1001665] [Citation(s) in RCA: 127] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Accepted: 08/12/2013] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND While acute lung injury (ALI) contributes significantly to critical illness, it resolves spontaneously in many instances. The majority of patients experiencing ALI require mechanical ventilation. Therefore, we hypothesized that mechanical ventilation and concomitant stretch-exposure of pulmonary epithelia could activate endogenous pathways important in lung protection. METHODS AND FINDINGS To examine transcriptional responses during ALI, we exposed pulmonary epithelia to cyclic mechanical stretch conditions--an in vitro model resembling mechanical ventilation. A genome-wide screen revealed a transcriptional response similar to hypoxia signaling. Surprisingly, we found that stabilization of hypoxia-inducible factor 1A (HIF1A) during stretch conditions in vitro or during ventilator-induced ALI in vivo occurs under normoxic conditions. Extension of these findings identified a functional role for stretch-induced inhibition of succinate dehydrogenase (SDH) in mediating normoxic HIF1A stabilization, concomitant increases in glycolytic capacity, and improved tricarboxylic acid (TCA) cycle function. Pharmacologic studies with HIF activator or inhibitor treatment implicated HIF1A-stabilization in attenuating pulmonary edema and lung inflammation during ALI in vivo. Systematic deletion of HIF1A in the lungs, endothelia, myeloid cells, or pulmonary epithelia linked these findings to alveolar-epithelial HIF1A. In vivo analysis of ¹³C-glucose metabolites utilizing liquid-chromatography tandem mass-spectrometry demonstrated that increases in glycolytic capacity, improvement of mitochondrial respiration, and concomitant attenuation of lung inflammation during ALI were specific for alveolar-epithelial expressed HIF1A. CONCLUSIONS These studies reveal a surprising role for HIF1A in lung protection during ALI, where normoxic HIF1A stabilization and HIF-dependent control of alveolar-epithelial glucose metabolism function as an endogenous feedback loop to dampen lung inflammation.
Collapse
Affiliation(s)
- Tobias Eckle
- Organ Protection Program, Department of Anesthesiology, University of Colorado School of Medicine, Denver, Colorado, United States of America
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
|
39
|
Signaling through hepatocellular A2B adenosine receptors dampens ischemia and reperfusion injury of the liver. Proc Natl Acad Sci U S A 2013; 110:12012-7. [PMID: 23812746 DOI: 10.1073/pnas.1221733110] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Ischemia and reperfusion significantly contributes to the morbidity and mortality of liver surgery and transplantation. Based on studies showing a critical role for adenosine signaling in mediating tissue adaptation during hypoxia, we hypothesized that signaling events through adenosine receptors (ADORA1, ADORA2A, ADORA2B, or ADORA3) attenuates hepatic ischemia and reperfusion injury. Initial screening studies of human liver biopsies obtained during hepatic transplantation demonstrated a selective and robust induction of ADORA2B transcript and protein following ischemia and reperfusion. Subsequent exposure of gene-targeted mice for each individual adenosine receptor to liver ischemia and reperfusion revealed a selective role for the Adora2b in liver protection. Moreover, treatment of wild-type mice with an Adora2b-selective antagonist resulted in enhanced liver injury, whereas Adora2b-agonist treatment was associated with attenuated hepatic injury in wild-type, but not in Adora2b(-/-) mice. Subsequent studies in mice with Adora2b deletion in different tissues--including vascular endothelia, myeloid cells, and hepatocytes--revealed a surprising role for hepatocellular-specific Adora2b signaling in attenuating nuclear factor NF-κB activation and thereby mediating liver protection from ischemia and reperfusion injury. These studies provide a unique role for hepatocellular-specific Adora2b signaling in liver protection during ischemia and reperfusion injury.
Collapse
|
40
|
Ehrentraut H, Clambey ET, McNamee EN, Brodsky KS, Ehrentraut SF, Poth JM, Riegel AK, Westrich JA, Colgan SP, Eltzschig HK. CD73+ regulatory T cells contribute to adenosine-mediated resolution of acute lung injury. FASEB J 2013; 27:2207-19. [PMID: 23413361 PMCID: PMC3659359 DOI: 10.1096/fj.12-225201] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Accepted: 02/04/2013] [Indexed: 12/22/2022]
Abstract
Acute lung injury (ALI) is characterized by alveolar injury and uncontrolled inflammation. Since most cases of ALI resolve spontaneously, understanding the endogenous mechanisms that promote ALI resolution is important to developing effective therapies. Previous studies have implicated extracellular adenosine signaling in tissue adaptation and wound healing. Therefore, we hypothesized a functional contribution for the endogenous production of adenosine during ALI resolution. As a model, we administered intratracheal LPS and observed peak lung injury at 3 d, with resolution by d 14. Treatment with pegylated adenosine-deaminase to enhance extracellular adenosine breakdown revealed impaired ALI resolution. Similarly, genetic deletion of cd73, the pacemaker for extracellular adenosine generation, was associated with increased mortality (0% wild-type and 40% in cd73(-/-) mice; P<0.05) and failure to resolve ALI adequately. Studies of inflammatory cell trafficking into the lungs during ALI resolution revealed that regulatory T cells (Tregs) express the highest levels of CD73. While Treg numbers in cd73(-/-) mice were similar to controls, cd73-deficient Tregs had attenuated immunosuppressive functions. Moreover, adoptive transfer of cd73-deficient Tregs into Rag(-/-) mice emulated the observed phenotype in cd73(-/-) mice, while transfer of wild-type Tregs was associated with normal ALI resolution. Together, these studies implicate CD73-dependent adenosine generation in Tregs in promoting ALI resolution.
Collapse
Affiliation(s)
- Heidi Ehrentraut
- Department of Anesthesiology and
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| | | | | | | | - Stefan F. Ehrentraut
- Department of Medicine, Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, Colorado, USA; and
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Jens M. Poth
- Department of Anesthesiology and
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| | | | | | - Sean P. Colgan
- Department of Medicine, Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, Colorado, USA; and
| | | |
Collapse
|
41
|
Rowland KJ, Yao J, Wang L, Erwin CR, Maslov KI, Wang LV, Warner BW. Up-regulation of hypoxia-inducible factor 1 alpha and hemodynamic responses following massive small bowel resection. J Pediatr Surg 2013; 48:1330-9. [PMID: 23845627 PMCID: PMC3755458 DOI: 10.1016/j.jpedsurg.2013.03.031] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Accepted: 03/08/2013] [Indexed: 02/07/2023]
Abstract
PURPOSE Massive small bowel resection (SBR) results in an adaptive response within the remnant bowel. We have previously shown an immediate reduction in intestinal blood flow and oxygen saturation (sO2) after SBR. We therefore sought to determine the duration of resection-induced intestinal hypoxia and expression of hypoxia-inducible factors (HIFs) following SBR. METHODS C57B6 mice were subjected to 50% proximal SBR or a sham procedure. Photoacoustic microscopy (PAM) was used to measure blood flow and sO2 on postoperative days (PODs) 1, 3, and 7. Ileal tissue was harvested 6h postoperatively and on PODs 1 and 2, and HIF1α, HIF2α, and VEGF mRNA expression were assessed via RT-PCR. A p value of less than 0.05 was considered significant. RESULTS Following SBR, reduction in intestinal blood flow persists for 24h and is followed with hyperemia by POD 3. The immediate reduction in venous sO2 and increased tissue oxygen utilization continued through POD 7. Enhanced expression of HIF1α was demonstrated 6h following SBR. CONCLUSION Massive SBR results in an immediate relative hypoxic state within the remnant bowel with early enhanced expression of HIF1α. On POD 7, increased tissue oxygen extraction and elevated blood flow persist in the adapting intestine.
Collapse
Affiliation(s)
- Kathryn J. Rowland
- Division of Pediatric Surgery, St Louis Children's Hospital, Department of Surgery, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Junjie Yao
- Optical Imaging Laboratory, Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Lidai Wang
- Optical Imaging Laboratory, Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Christopher R. Erwin
- Division of Pediatric Surgery, St Louis Children's Hospital, Department of Surgery, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Konstantin I. Maslov
- Optical Imaging Laboratory, Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Lihong V. Wang
- Optical Imaging Laboratory, Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA,Corresponding author. Tel.: +1 314 454 6022; fax: +1 314 454 2442. (L.V. Wang), (B.W. Warner)
| | - Brad W. Warner
- Division of Pediatric Surgery, St Louis Children's Hospital, Department of Surgery, Washington University School of Medicine, St Louis, MO 63110, USA,Corresponding author. Tel.: +1 314 454 6022; fax: +1 314 454 2442. (L.V. Wang), (B.W. Warner)
| |
Collapse
|
42
|
Mononuclear cell adenosine deaminase and CD26/dipeptidylpeptidase-IV activities are sensitive markers of reperfusion during percutaneous transluminal angioplasty. Int J Cardiol 2013; 166:225-9. [DOI: 10.1016/j.ijcard.2011.10.090] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2011] [Revised: 09/21/2011] [Accepted: 10/18/2011] [Indexed: 11/21/2022]
|
43
|
Dong W, Li F, Pan Z, Liu S, Yu H, Wang X, Bi S, Zhang W. Resveratrol ameliorates subacute intestinal ischemia-reperfusion injury. J Surg Res 2013; 185:182-9. [PMID: 23735732 DOI: 10.1016/j.jss.2013.05.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Revised: 04/26/2013] [Accepted: 05/03/2013] [Indexed: 01/11/2023]
Abstract
BACKGROUND Resveratrol has been shown to attenuate reactive oxygen species formation and protect against ischemia-reperfusion (I/R) injury. However, the effects of resveratrol against subacute intestinal I/R injury are not clearly elucidated. Therefore, this study was designed to investigate the effects and possible protective mechanisms of resveratrol on subacute intestinal I/R injury in mice. METHODS BALB/c mice were subjected to 1 h ischemia by occluding the superior mesenteric artery and 24 h reperfusion. Histologic injury; myeloperoxidase, superoxide dismutase, and glutathione peroxidase activity; malondialdehyde level; inducible nitric oxide synthase (iNOS), Ac-NF-κBp65, and sirtuin 1 (SIRT1) expression; NF-κB translocation; and nitric oxide (NO) production were examined in treated with or without resveratrol in the absence or presence of pharmacologic inhibitors. RESULTS Resveratrol significantly ameliorated subacute intestinal I/R injury accompanied with the decrease of NO production as well as iNOS expression. In addition, resveratrol obviously upregulated the expression of SIRT1 and inhibited the activity of NF-κB. After application of iNOS inhibitor S-methylisothiourea and NF-κB inhibitor pyrrolidine dithiocarbamate, the protective effect of resveratrol was significantly augmented by attenuating iNOS and NO production, indicating that resveratrol exerted its protective effect on intestinal I/R injury via NF-κB-mediated iNOS pathway. Furthermore, the protective effect of resveratrol was correlated with SIRT1, because application of SIRT1 inhibitor nicotinamide strikingly weakened the protective effect of resveratrol. CONCLUSIONS Taken together, our findings showed that resveratrol protects intestinal subacute I/R injury via the SIRT1-NF-κB pathway in an iNOS-NO-dependent manner. Therefore, resveratrol has a potential clinical prospect for further development of anti-injury therapy.
Collapse
Affiliation(s)
- WenPeng Dong
- Department of Cardiovascular Surgery, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, China
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Protective role for netrin-1 during diabetic nephropathy. J Mol Med (Berl) 2013; 91:1071-80. [PMID: 23636509 DOI: 10.1007/s00109-013-1041-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Revised: 04/03/2013] [Accepted: 04/11/2013] [Indexed: 02/08/2023]
Abstract
Recent studies implicate neuronal guidance molecules in the orchestration of inflammatory events. For example, previous studies demonstrate a functional role for netrin-1 in attenuating acute kidney injury. Here, we hypothesized a kidney-protective role for netrin-1 during chronic kidney disease, such as occurs during diabetic nephropathy. To study the role of netrin-1 during diabetic nephropathy, we induced diabetes in mice at the age of 8 weeks by streptocotozin (STZ) treatment. Sixteen weeks after STZ treatment, we examined the kidneys. Initial studies in wild-type mice demonstrated robust induction of renal, urinary, and plasma netrin-1 protein levels during diabetic nephropathy. Subsequent genetic studies in mice with partial netrin-1 deficiency (Ntrn1(+/-) mice) revealed a more severe degree of diabetic nephropathy, including more severe loss of kidney function (albuminuria, glomerular filtration rate, histology). We subsequently performed pharmacologic studies with recombinant netrin-1 treatment given continuously via osmotic pump. Indeed, netrin-1 treatment was associated with attenuated albuminuria and improved histologic scores for diabetic nephropathy compared to controls. Consistent with previous studies implicating purinergic signaling in netrin-1-elicited tissue protection, mice deficient in the Adora2b adenosine receptor were not protected. Taken together, these studies demonstrate a functional role for endogenous netrin-1 in attenuating diabetic kidney disease.
Collapse
|
45
|
Eckle T, Hughes K, Ehrentraut H, Brodsky KS, Rosenberger P, Choi DS, Ravid K, Weng T, Xia Y, Blackburn MR, Eltzschig HK. Crosstalk between the equilibrative nucleoside transporter ENT2 and alveolar Adora2b adenosine receptors dampens acute lung injury. FASEB J 2013; 27:3078-89. [PMID: 23603835 DOI: 10.1096/fj.13-228551] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The signaling molecule adenosine has been implicated in attenuating acute lung injury (ALI). Adenosine signaling is terminated by its uptake through equilibrative nucleoside transporters (ENTs). We hypothesized that ENT-dependent adenosine uptake could be targeted to enhance adenosine-mediated lung protection. To address this hypothesis, we exposed mice to high-pressure mechanical ventilation to induce ALI. Initial studies demonstrated time-dependent repression of ENT1 and ENT2 transcript and protein levels during ALI. To examine the contention that ENT repression represents an endogenous adaptive response, we performed functional studies with the ENT inhibitor dipyridamole. Dipyridamole treatment (1 mg/kg; EC50=10 μM) was associated with significant increases in ALI survival time (277 vs. 395 min; P<0.05). Subsequent studies in gene-targeted mice for Ent1 or Ent2 revealed a selective phenotype in Ent2(-/-) mice, including attenuated pulmonary edema and improved gas exchange during ALI in conjunction with elevated adenosine levels in the bronchoalveolar fluid. Furthermore, studies in genetic models for adenosine receptors implicated the A2B adenosine receptor (Adora2b) in mediating ENT-dependent lung protection. Notably, dipyridamole-dependent attenuation of lung inflammation was abolished in mice with alveolar epithelial Adora2b gene deletion. Our newly identified crosstalk pathway between ENT2 and alveolar epithelial Adora2b in lung protection during ALI opens possibilities for combined therapies targeted to this protein set.
Collapse
Affiliation(s)
- Tobias Eckle
- Mucosal Inflammation Program, Department of Anesthesiology, University of Colorado School of Medicine, 12700 E. 19th Ave., Aurora, CO 80045, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Antonioli L, Pacher P, Vizi ES, Haskó G. CD39 and CD73 in immunity and inflammation. Trends Mol Med 2013; 19:355-67. [PMID: 23601906 DOI: 10.1016/j.molmed.2013.03.005] [Citation(s) in RCA: 845] [Impact Index Per Article: 76.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Revised: 03/14/2013] [Accepted: 03/15/2013] [Indexed: 12/11/2022]
Abstract
The enzymatic activities of CD39 and CD73 play strategic roles in calibrating the duration, magnitude, and chemical nature of purinergic signals delivered to immune cells through the conversion of ADP/ATP to AMP and AMP to adenosine, respectively. This drives a shift from an ATP-driven proinflammatory environment to an anti-inflammatory milieu induced by adenosine. The CD39/CD73 pathway changes dynamically with the pathophysiological context in which it is embedded. It is becoming increasingly appreciated that altering this catabolic machinery can change the course or dictate the outcome of several pathophysiological events, such as AIDS, autoimmune diseases, infections, atherosclerosis, ischemia-reperfusion injury, and cancer, suggesting these ectoenzymes are novel therapeutic targets for managing a variety of disorders.
Collapse
Affiliation(s)
- Luca Antonioli
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | | | | | | |
Collapse
|
47
|
Antonioli L, Colucci R, Pellegrini C, Giustarini G, Tuccori M, Blandizzi C, Fornai M. The role of purinergic pathways in the pathophysiology of gut diseases: pharmacological modulation and potential therapeutic applications. Pharmacol Ther 2013; 139:157-88. [PMID: 23588157 DOI: 10.1016/j.pharmthera.2013.04.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Accepted: 03/15/2013] [Indexed: 02/08/2023]
Abstract
Gut homeostasis results from complex neuro-immune interactions aimed at triggering stereotypical and specific programs of coordinated mucosal secretion and powerful motor propulsion. A prominent role in the regulation of this highly integrated network, comprising a variety of immune/inflammatory cells and the enteric nervous system, is played by purinergic mediators. The cells of the digestive tract are literally plunged into a "biological sea" of functionally active nucleotides and nucleosides, which carry out the critical task of driving regulatory interventions on cellular functions through the activation of P1 and P2 receptors. Intensive research efforts are being made to achieve an integrated view of the purinergic system, since it is emerging that the various components of purinergic pathways (i.e., enzymes, transporters, mediators and receptors) are mutually linked entities, deputed to finely modulating the magnitude and the duration of purinergic signaling, and that alterations occurring in this balanced network could be intimately involved in the pathophysiology of several gut disorders. This review article intends to provide a critical appraisal of current knowledge on the purinergic system role in the regulation of gastrointestinal functions, considering these pathways as a whole integrated network, which is capable of finely controlling the levels of bioactive nucleotides and nucleosides in the biophase of their respective receptors. Special attention is paid to the mechanisms through which alterations in the various compartments of the purinergic system could contribute to the pathophysiology of gut disorders, and to the possibility of counteracting such dysfunctions by means of pharmacological interventions on purinergic molecular targets.
Collapse
Affiliation(s)
- Luca Antonioli
- Department of Clinical and Experimental Medicine, University of Pisa, Italy.
| | | | | | | | | | | | | |
Collapse
|
48
|
Eltzschig HK, Bonney SK, Eckle T. Attenuating myocardial ischemia by targeting A2B adenosine receptors. Trends Mol Med 2013; 19:345-54. [PMID: 23540714 DOI: 10.1016/j.molmed.2013.02.005] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Revised: 02/12/2013] [Accepted: 02/20/2013] [Indexed: 02/06/2023]
Abstract
Myocardial ischemia is associated with profound tissue hypoxia due to an imbalance in oxygen supply and demand, and studies of hypoxia-elicited adaptive responses during myocardial ischemia revealed a cardioprotective role for the signaling molecule adenosine. In ischemic human hearts, the A2B adenosine receptor (ADORA2B) is selectively induced. Functional studies in genetic models show that ADORA2B signaling attenuates myocardial infarction by adapting metabolism towards more oxygen efficient utilization of carbohydrates. This adenosine-mediated cardio-adaptive response involves the transcription factor hypoxia-inducible factor HIF1α and the circadian rhythm protein PER2. In this article, we discuss advances in the understanding of adenosine-elicited cardioprotection with particular emphasis on ADORA2B, its downstream targets, and the implications for novel strategies to prevent or treat myocardial ischemia.
Collapse
Affiliation(s)
- Holger K Eltzschig
- Mucosal Inflammation Program, Department of Anesthesiology, University of Colorado School of Medicine, Aurora, CO, USA.
| | | | | |
Collapse
|
49
|
Kim M, Ham A, Kim JY, Brown KM, D'Agati VD, Lee HT. The volatile anesthetic isoflurane induces ecto-5'-nucleotidase (CD73) to protect against renal ischemia and reperfusion injury. Kidney Int 2013; 84:90-103. [PMID: 23423261 PMCID: PMC3676468 DOI: 10.1038/ki.2013.43] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Revised: 11/21/2012] [Accepted: 12/17/2012] [Indexed: 12/17/2022]
Abstract
The volatile anesthetic isoflurane protects against renal ischemia and reperfusion injury by releasing renal tubular TGF-β1. Since adenosine is a powerful cytoprotective molecule, we tested whether TGF-β1 generated by isoflurane induces renal tubular ecto-5′-nucleotidase (CD73) and adenosine to protect against renal ischemia and reperfusion injury. Isoflurane induced new CD73 synthesis and increased adenosine generation in cultured kidney proximal tubule cells and in mouse kidney. Moreover, a TGF-β1 neutralizing antibody prevented isoflurane-mediated induction of CD73 activity. Mice anesthetized with isoflurane after renal ischemia and reperfusion had significantly reduced plasma creatinine and decreased renal tubular necrosis, neutrophil infiltration and apoptosis compared to pentobarbital-anesthetized mice. Isoflurane failed to protect against renal ischemia and reperfusion injury in CD73 deficient mice, in mice pretreated with a selective CD73 inhibitor or mice treated with an adenosine receptor antagonist. The TGF-β1 neutralizing antibody or the CD73 inhibitor attenuated isoflurane-mediated protection against HK-2 cell apoptosis. Thus, isoflurane causes TGF-β1-dependent induction of renal tubular CD73 and adenosine generation to protect against renal ischemia and reperfusion injury. Modulation of this pathway may have important therapeutic implications to reduce morbidity and mortality arising from ischemic acute kidney injury.
Collapse
Affiliation(s)
- Mihwa Kim
- Department of Anesthesiology, College of Physicians and Surgeons of Columbia University, Anesthesiology Research Laboratories, Columbia University, New York, New York, USA
| | | | | | | | | | | |
Collapse
|
50
|
Poth JM, Brodsky K, Ehrentraut H, Grenz A, Eltzschig HK. Transcriptional control of adenosine signaling by hypoxia-inducible transcription factors during ischemic or inflammatory disease. J Mol Med (Berl) 2013; 91:183-93. [PMID: 23263788 PMCID: PMC3560301 DOI: 10.1007/s00109-012-0988-7] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2012] [Revised: 11/30/2012] [Accepted: 12/04/2012] [Indexed: 02/08/2023]
Abstract
Inflammatory lesions, ischemic tissues, or solid tumors are characterized by the occurrence of severe tissue hypoxia within the diseased tissue. Subsequent stabilization of hypoxia-inducible transcription factors-particularly of hypoxia-inducible factor 1α (HIF1A)--results in significant alterations of gene expression of resident cells or inflammatory cells that have been recruited into such lesions. Interestingly, studies of hypoxia-induced changes of gene expression identified a transcriptional program that promotes extracellular adenosine signaling. Adenosine is a signaling molecule that functions through the activation of four distinct adenosine receptors--the ADORA1, ADORA2A, ADORA2B, and ADORA3 receptors. Extracellular adenosine is predominantly derived from the phosphohydrolysis of precursor nucleotides, such as adenosine triphosphate or adenosine monophosphate. HIF1A-elicited alterations in gene expression enhance the enzymatic capacity within inflamed tissues to produce extracellular adenosine. Moreover, hypoxia-elicited induction of adenosine receptors--particularly of ADORA2B--results in increased signal transduction. Functional studies in genetic models for HIF1A or adenosine receptors implicate this pathway in an endogenous feedback loop that dampens excessive inflammation and promotes injury resolution, while at the same time enhancing ischemia tolerance. Therefore, pharmacological strategies to enhance HIF-elicited adenosine production or to promote adenosine signaling through adenosine receptors are being investigated for the treatment of acute inflammatory or ischemic diseases characterized by tissue hypoxia.
Collapse
Affiliation(s)
- Jens M. Poth
- Mucosal Inflammation Program, Department of Anesthesiology, University of Colorado School of Medicine, Aurora, USA
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Bonn, Germany
| | - Kelley Brodsky
- Mucosal Inflammation Program, Department of Anesthesiology, University of Colorado School of Medicine, Aurora, USA
| | - Heidi Ehrentraut
- Mucosal Inflammation Program, Department of Anesthesiology, University of Colorado School of Medicine, Aurora, USA
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Bonn, Germany
| | - Almut Grenz
- Mucosal Inflammation Program, Department of Anesthesiology, University of Colorado School of Medicine, Aurora, USA
| | - Holger K. Eltzschig
- Mucosal Inflammation Program, Department of Anesthesiology, University of Colorado School of Medicine, Aurora, USA
| |
Collapse
|