Number of Circulating CD 73-Expressing Lymphocytes Correlates With Survival After Cardiac Arrest

Sergey Ryzhov, Teresa May, John Dziodzio, Ivette F Emery, F L Lucas, Angela Leclerc, Barbara McCrum, Christine Lord, Ashley Eldridge, Michel P Robich, Fumito Ichinose, Douglas B Sawyer, Richard Riker, David B Seder, Sergey Ryzhov, Teresa May, John Dziodzio, Ivette F Emery, F L Lucas, Angela Leclerc, Barbara McCrum, Christine Lord, Ashley Eldridge, Michel P Robich, Fumito Ichinose, Douglas B Sawyer, Richard Riker, David B Seder

Abstract

Background Patients resuscitated from cardiac arrest ( CA ) have highly variable neurological, circulatory, and systemic ischemia-reperfusion injuries. After the initial hypoxic-ischemic insult, a cascade of immune and inflammatory responses develops and is often fatal. The role of the immune response in pathophysiological characteristics and recovery is not well understood. We studied immune cell activity and its association with outcomes in a cohort of CA survivors. Methods and Results After informed consent, we collected blood samples at intervals over a week after resuscitation from CA . We examined the expression of CD 39 and CD 73 (alias 5'-nucleotidase), production of tumor necrosis factor-α, generation of reactive oxygen species, and secretion of vascular endothelial growth factor by circulating myeloid and lymphoid cells, in comparison to cells obtained from control subjects before coronary artery bypass grafting surgery. The number of circulating total and CD 73-expressing lymphocytes correlated with survival after CA . Incubation of immune cells, obtained from post- CA subjects, with AMP , a substrate for CD 73, resulted in inhibition of tumor necrosis factor-α production and generation of reactive oxygen species. This effect was blocked by adenosine 5'-(α, β-methylene) diphosphate, a specific inhibitor of CD 73 and ZM 241385, an A2 adenosine receptor antagonist. We also found that AMP -dependent activation of CD 73 induces production of vascular endothelial growth factor. Conclusions CD 73-expressing lymphocytes mediate cellular protection from inflammation after CA through inhibition of proinflammatory activation of myeloid cells and promotion of vascular endothelial growth factor secretion. The contribution of CD 73 lymphocytes in the regulation of acute inflammation and tissue injury after CA warrants further study.

Keywords: CD73; cardiac arrest; inflammation; lymphocytes.

Figures

Figure 1
Figure 1
Time‐dependent changes in subpopulations of white blood cells (WBCs) after cardiac arrest (CA). A, Representative flow cytometric contour plots demonstrating percentages of neutrophils (upper gate), monocytes (middle gate), and lymphocytes (lower gate) in peripheral blood of subjects who underwent preoperative coronary artery bypass grafting (control) and subjects with CA on different time points after return of spontaneous circulation (ROSC). B through E, Graphical representation of flow cytometry data showing total number of WBCs (B) and cells in neutrophil (C), monocyte (D), and lymphocyte (E) gates in groups of control subjects (n=30) and subjects with CA (n=48). Data are presented in standard percentile format (minimum value, 25th percentile; median, 75th percentile; and maximum value). Statistical significance was calculated using Kruskal‐Wallis test with Dunn's multiple‐comparisons posttest, and P values are indicated. F and G, The number of cells in neutrophil (F) and lymphocyte (G) gates in survivors (n=19) and nonsurvivors (n=29) after CA. Mann‐Whitney test was used, and P values are indicated. SSC indicates side scatter.
Figure 2
Figure 2
Number of CD3 T cells is significantly decreased after cardiac arrest (CA) and associated with survival. A and B, Representative flow cytometric plots demonstrating percentage of CD3‐positive (CD3pos; upper left quadrant), CD19‐positive (CD19pos; lower right quadrant), and CD3‐negative (CD3neg)/CD19‐negative (CD19neg; where negative indicates no expression; lower left quadrant) cells within the side scatter (SSC) low CD45high lymphocyte gate (contour plots) in control subjects (A) and subjects with CA (B). C through E, Graphical representation of flow cytometry data showing total number of CD3pos T cells (C), CD19pos B cells (D), and CD3neg/CD19neg cells (E) in groups of control subjects (n=30) and subjects with CA (n=48). The number of cells was calculated using total number of white blood cells, percentage of cells in the lymphocyte gate, and percentage of cells corresponding to specific cell subpopulation. Statistical significance was calculated using Kruskal‐Wallis test with Dunn's multiple‐comparisons posttest, and P values are indicated. F and G, The number of CD3pos T cells (F) and CD19pos B cells (G) in survivors (n=19) and nonsurvivors (n=29) after CA. Mann‐Whitney test was used. ROSC indicates return of spontaneous circulation.
Figure 3
Figure 3
Higher number of CD73‐positive (CD73pos) lymphocytes but not CD39‐positive (CD39pos) cells is associated with survival after cardiac arrest (CA). A, Representative flow cytometric plots showing total percentage of CD39pos cells (left and middle contour plots) and subsets of CD39‐expressing CD3‐positive (CD3pos), CD19‐positive (CD19pos), and CD3‐negative (CD3neg)/CD19‐negative (CD19neg) cells (where negative indicates no expression; right dot plots) in control subjects (top) and subjects with CA (bottom; 6 hours after return of spontaneous circulation [ROSC]). B, Graphical representation of flow cytometry data showing total number of CD39‐expressing cells in control subjects (n=30) and subjects with CA (n=48) at different times after ROSC. Kruskal‐Wallis test and Dunn's multiple‐comparisons posttest were used. C, The number of CD39pos cells in survivors (n=19) and nonsurvivors (n=29) after CA. Mann‐Whitney test was used. D, Flow cytometric plots showing total percentage of CD73pos cells in peripheral blood and subsets of CD73‐expressing CD3pos and CD19pos cells. E, Graphical representation of flow cytometry data showing total number of CD73‐expressing cells in control subjects (n=30) and subjects with CA (n=48) at different times after ROSC. Kruskal‐Wallis test and Dunn's multiple‐comparisons posttest were used. F, The number of CD73pos cells in survivors (n=19) and nonsurvivors (n=29) after CA. Mann‐Whitney test was used. FITC indicates fluorescein isothiocyanate; SSC, side scatter.
Figure 4
Figure 4
CD73 mediates inhibition of lipopolysaccharide‐induced production of tumor necrosis factor‐α (TNF‐α) and generation of reactive oxygen species (ROS) by myeloid cells. A through C, E, and F, White blood cells (WBCs) were isolated 24 hours after return of spontaneous circulation (ROSC) from 5 subjects with cardiac arrest (CA) with a median value of 177 (interquartile range [IQR], 162–300) CD73‐expressing lymphocytes/μL of blood, which closely resembles the median value and IQR found in a group of survivors (median, 182; IQR, 150–275 CD73‐expressing lymphocytes/μL of blood) at 24 hours after ROSC. A, Representative flow cytometric plots showing cells with high levels of TNF‐α protein production in subpopulations of neutrophils (upper gate), monocytes (intermediate gate), and lymphocytes (lower gate) in the absence (basal) or presence of 10 ng/mL lipopolysaccharide alone or in combinations with 100 μmol/L AMP (lipopolysaccharide+AMP), 100 μmol/L of CD73 inhibitor, adenosine 5′‐(α, β‐methylene) diphosphate (APCP; lipopolysaccharide+AMP+APCP), and adenosine receptor inhibitor, 300 nmol/L ZM 241385 (lipopolysaccharide+AMP+ZM). Cells were incubated for 6 hours in the presence of brefeldin A to prevent secretion of TNF‐α from cells and 10 μmol/L erythro‐9‐(2‐hydroxy‐3‐nonyl)−adenine hydrochloride to prevent degradation of adenosine in cell culture. B, Graphical representation of flow cytometric data demonstrating percentage of cells with high expression of TNF‐α (n=5); 1‐way ANOVA was used, and P values from Tukey's multiple‐comparisons test are shown. C, Levels of TNF‐α protein in supernatant of WBCs measured by ELISA (n=5); 1‐way ANOVA was used with Tukey's multiple‐comparisons test. D, Levels of TNF‐α protein in the peripheral circulation of control subjects (n=30) and subjects with CA (n=48). Kruskal‐Wallis test and Dunn's multiple‐comparisons posttest were used. E, Representative flow cytometric histograms demonstrating basal (gray‐shaded) and 10 ng/mL lipopolysaccharide‐induced (open histogram) levels of ROS generation in major subpopulations of WBCs obtained from subjects with CA. F, Graphical representation of ROS generation in the absence (basal) or presence of lipopolysaccharide alone or in combinations with 100 μmol/L AMP (lipopolysaccharide+AMP) and 100 μmol/L of CD73 inhibitor, APCP (lipopolysaccharide+AMP+APCP) (n=5); 1‐way ANOVA with Tukey's multiple‐comparisons test was used. MFI indicates mean fluorescence intensity.
Figure 5
Figure 5
CD73 mediates upregulation of vascular endothelial growth factor (VEGF) protein secretion from white blood cells (WBCs) of subjects with cardiac arrest (CA). A, Level of VEGF protein in supernatant of WBCs obtained from subjects with CA and incubated for 6 hours in the absence (basal) or presence of 100 μmol/L AMP alone or in combination with 100 μmol/L adenosine 5′‐(α, β‐methylene) diphosphate (APCP; AMP+APCP). WBCs were isolated 24 hours after return of spontaneous circulation (ROSC) from 5 subjects with CA (n=5), with a median value of 177 (interquartile range [IQR], 162–300) CD73‐expressing lymphocytes/μL of blood, which closely resembles the median value and IQR found in a group of survivors (median, 182; IQR, 150–275 CD73‐expressing lymphocytes/μL of blood) at 24 hours after ROSC. Incubation medium contained 10 μmol/L erythro‐9‐(2‐hydroxy‐3‐nonyl)−adenine hydrochloride, an adenosine deaminase inhibitor, to decelerate adenosine catabolism in cell culture. Data presented as mean±SEM (n=5); 1‐way ANOVA with Tukey's multiple‐comparisons test was used. B, Levels of VEGF protein in peripheral circulation of control subjects (n=30) and subjects with CA (n=48); Kruskal‐Wallis test was used, and P values are indicated (Dunn's multiple‐comparisons test). C, The correlation between levels of VEGF protein and number of CD73‐positive (CD73pos) lymphocytes in blood of subjects with CA at 6 and 12 hours after ROSC (n=48). Spearman's correlation coefficient and P values are as indicated.
Figure 6
Figure 6
Proposed role of CD73/5′‐nucleotidase–expressing lymphocytes in the control of immune response after cardiac arrest. Myeloid cells (neutrophils and monocytes) express CD39 ectonucleotidases that mediate hydrolysis of ATP, released from dead and apoptotic cells, to ADP and AMP. Then, AMP is hydrolyzed by CD73/5′‐nucleotidase, expressed on lymphocytes, to adenosine, which suppresses proinflammatory activation of myeloid cells, secretion of tumor necrosis factor‐α (TNF‐α), and production of reactive oxygen species (ROS) and promotes production of prosurvival vascular endothelial growth factor (VEGF).

References

    1. Bobrow BJ, Spaite DW, Berg RA, Stolz U, Sanders AB, Kern KB, Vadeboncoeur TF, Clark LL, Gallagher JV, Stapczynski JS, LoVecchio F, Mullins TJ, Humble WO, Ewy GA. Chest compression‐only CPR by lay rescuers and survival from out‐of‐hospital cardiac arrest. JAMA. 2010;304:1447–1454.
    1. Bobrow BJ, Clark LL, Ewy GA, Chikani V, Sanders AB, Berg RA, Richman PB, Kern KB. Minimally interrupted cardiac resuscitation by emergency medical services for out‐of‐hospital cardiac arrest. JAMA. 2008;299:1158–1165.
    1. Peberdy MA, Callaway CW, Neumar RW, Geocadin RG, Zimmerman JL, Donnino M, Gabrielli A, Silvers SM, Zaritsky AL, Merchant R, Vanden Hoek TL, Kronick SL; American Heart Association . Part 9: post‐cardiac arrest care: 2010 American Heart Association guidelines for cardiopulmonary resuscitation and emergency cardiovascular care. Circulation. 2010;122:S768–S786.
    1. Writing Group Members , Mozaffarian D, Benjamin EJ, Go AS, Arnett DK, Blaha MJ, Cushman M, Das SR, de Ferranti S, Despres JP, Fullerton HJ, Howard VJ, Huffman MD, Isasi CR, Jimenez MC, Judd SE, Kissela BM, Lichtman JH, Lisabeth LD, Liu S, Mackey RH, Magid DJ, McGuire DK, Mohler ER III, Moy CS, Muntner P, Mussolino ME, Nasir K, Neumar RW, Nichol G, Palaniappan L, Pandey DK, Reeves MJ, Rodriguez CJ, Rosamond W, Sorlie PD, Stein J, Towfighi A, Turan TN, Virani SS, Woo D, Yeh RW, Turner MB; American Heart Association Statistics Committee, Stroke Statistics Subcommittee . Heart disease and stroke statistics—2016 update: a report from the American Heart Association. Circulation. 2016;133:e38–e60.
    1. Adrie C, Adib‐Conquy M, Laurent I, Monchi M, Vinsonneau C, Fitting C, Fraisse F, Dinh‐Xuan AT, Carli P, Spaulding C, Dhainaut JF, Cavaillon JM. Successful cardiopulmonary resuscitation after cardiac arrest as a “sepsis‐like” syndrome. Circulation. 2002;106:562–568.
    1. Bro‐Jeppesen J, Kjaergaard J, Wanscher M, Nielsen N, Friberg H, Bjerre M, Hassager C. Systemic inflammatory response and potential prognostic implications after out‐of‐hospital cardiac arrest: a substudy of the target temperature management trial. Crit Care Med. 2015;43:1223–1232.
    1. Neumar RW, Nolan JP, Adrie C, Aibiki M, Berg RA, Bottiger BW, Callaway C, Clark RS, Geocadin RG, Jauch EC, Kern KB, Laurent I, Longstreth WT Jr, Merchant RM, Morley P, Morrison LJ, Nadkarni V, Peberdy MA, Rivers EP, Rodriguez‐Nunez A, Sellke FW, Spaulding C, Sunde K, Vanden Hoek T. Post‐cardiac arrest syndrome: epidemiology, pathophysiology, treatment, and prognostication: a consensus statement from the international liaison committee on resuscitation (American Heart Association, Australian and New Zealand Council on Resuscitation, European Resuscitation Council, Heart and Stroke Foundation of Canada, Interamerican Heart Foundation, Resuscitation Council of Asia, and the Resuscitation Council of Southern Africa); the American Heart Association Emergency Cardiovascular Care Committee; the Council on Cardiovascular Surgery and Anesthesia; the Council on Cardiopulmonary, Perioperative, and Critical Care; the Council on Clinical Cardiology; and the Stroke Council. Circulation. 2008;118:2452–2483.
    1. Adrie C, Laurent I, Monchi M, Cariou A, Dhainaou JF, Spaulding C. Postresuscitation disease after cardiac arrest: a sepsis‐like syndrome? Curr Opin Crit Care. 2004;10:208–212.
    1. Bro‐Jeppesen J, Kjaergaard J, Stammet P, Wise MP, Hovdenes J, Aneman A, Horn J, Devaux Y, Erlinge D, Gasche Y, Wanscher M, Cronberg T, Friberg H, Wetterslev J, Pellis T, Kuiper M, Nielsen N, Hassager C; TTM‐Trial Investigators . Predictive value of interleukin‐6 in post‐cardiac arrest patients treated with targeted temperature management at 33 degrees C or 36 degrees C. Resuscitation. 2016;98:1–8.
    1. Vaahersalo J, Skrifvars MB, Pulkki K, Stridsberg M, Rosjo H, Hovilehto S, Tiainen M, Varpula T, Pettila V, Ruokonen E; FINNRESUSCI Laboratory Study Group . Admission interleukin‐6 is associated with post resuscitation organ dysfunction and predicts long‐term neurological outcome after out‐of‐hospital ventricular fibrillation. Resuscitation. 2014;85:1573–1579.
    1. Ridker PM, Luscher TF. Anti‐inflammatory therapies for cardiovascular disease. Eur Heart J. 2014;35:1782–1791.
    1. Jin R, Yang G, Li G. Inflammatory mechanisms in ischemic stroke: role of inflammatory cells. J Leukoc Biol. 2010;87:779–789.
    1. Benakis C, Garcia‐Bonilla L, Iadecola C, Anrather J. The role of microglia and myeloid immune cells in acute cerebral ischemia. Front Cell Neurosci. 2014;8:461.
    1. Elliott MR, Ravichandran KS. Clearance of apoptotic cells: implications in health and disease. J Cell Biol. 2010;189:1059–1070.
    1. Herz J, Filiano AJ, Smith A, Yogev N, Kipnis J. Myeloid cells in the central nervous system. Immunity. 2017;46:943–956.
    1. Xiang Y, Zhao H, Wang J, Zhang L, Liu A, Chen Y. Inflammatory mechanisms involved in brain injury following cardiac arrest and cardiopulmonary resuscitation. Biomed Rep. 2016;5:11–17.
    1. Drifte G, Dunn‐Siegrist I, Tissieres P, Pugin J. Innate immune functions of immature neutrophils in patients with sepsis and severe systemic inflammatory response syndrome. Crit Care Med. 2013;41:820–832.
    1. Yune HY, Chung SP, Park YS, Chung HS, Lee HS, Lee JW, Park JW, You JS, Park I, Lee HS. Delta neutrophil index as a promising prognostic marker in out of hospital cardiac arrest. PLoS One. 2015;10:e0120677.
    1. Rock KL, Lai JJ, Kono H. Innate and adaptive immune responses to cell death. Immunol Rev. 2011;243:191–205.
    1. Liebetrau C, Hoffmann J, Dorr O, Gaede L, Blumenstein J, Biermann H, Pyttel L, Thiele P, Troidl C, Berkowitsch A, Rolf A, Voss S, Hamm CW, Nef H, Mollmann H. Release kinetics of inflammatory biomarkers in a clinical model of acute myocardial infarction. Circ Res. 2015;116:867–875.
    1. Sawant AC, Adhikari P, Narra SR, Srivatsa SS, Mills PK, Srivatsa SS. Neutrophil to lymphocyte ratio predicts short‐ and long‐term mortality following revascularization therapy for ST elevation myocardial infarction. Cardiol J. 2014;21:500–508.
    1. He J, Li J, Wang Y, Hao P, Hua Q. Neutrophil‐to‐lymphocyte ratio (NLR) predicts mortality and adverse‐outcomes after ST‐segment elevation myocardial infarction in Chinese people. Int J Clin Exp Pathol. 2014;7:4045–4056.
    1. Mayadas TN, Cullere X, Lowell CA. The multifaceted functions of neutrophils. Annu Rev Pathol. 2014;9:181–218.
    1. Weiser C, Schwameis M, Sterz F, Herkner H, Lang IM, Schwarzinger I, Spiel AO. Mortality in patients resuscitated from out‐of‐hospital cardiac arrest based on automated blood cell count and neutrophil lymphocyte ratio at admission. Resuscitation. 2017;116:49–55.
    1. Baser K, Bas HD, Attaluri P, Rodrigues T, Nichols J, Nugen K. Changes in neutrophil‐to‐lymphocyte ratios in postcardiac arrest patients treated with targeted temperature management. Anatol J Cardiol. 2017;18:215–222.
    1. Villois P, Grimaldi D, Spadaro S, Shinotsuka CR, Fontana V, Scolletta S, Franchi F, Vincent JL, Creteur J, Taccone FS. Lymphopaenia in cardiac arrest patients. Ann Intensive Care. 2017;7:85.
    1. Nakai A, Hayano Y, Furuta F, Noda M, Suzuki K. Control of lymphocyte egress from lymph nodes through beta2‐adrenergic receptors. J Exp Med. 2014;211:2583–2598.
    1. Young AJ. The physiology of lymphocyte migration through the single lymph node in vivo. Semin Immunol. 1999;11:73–83.
    1. Westermann J, Pabst R. Lymphocyte subsets in the blood: a diagnostic window on the lymphoid system? Immunol Today. 1990;11:406–410.
    1. Asmussen A, Fink K, Busch HJ, Helbing T, Bourgeois N, Bode C, Grundmann S. Inflammasome and toll‐like receptor signaling in human monocytes after successful cardiopulmonary resuscitation. Crit Care. 2016;20:170.
    1. Bergt S, Guter A, Grub A, Wagner NM, Beltschany C, Langner S, Wree A, Hildebrandt S, Noldge‐Schomburg G, Vollmar B, Roesner JP. Impact of toll‐like receptor 2 deficiency on survival and neurological function after cardiac arrest: a murine model of cardiopulmonary resuscitation. PLoS One. 2013;8:e74944.
    1. Sun P, Xu L, Zhang Q, Li Q. Impact of toll‐like receptor 4 deficiency on cerebrocardiac syndrome. J Huazhong Univ Sci Technolog Med Sci. 2014;34:161–164.
    1. Zhang Q, Li G, Xu L, Li Q, Wang Q, Zhang Y, Zhang Q, Sun P. Tolllike receptor 4 contributes to acute kidney injury after cardiopulmonary resuscitation in mice. Mol Med Rep. 2016;14:2983–2990.
    1. Khan MM, Gandhi C, Chauhan N, Stevens JW, Motto DG, Lentz SR, Chauhan AK. Alternatively‐spliced extra domain A of fibronectin promotes acute inflammation and brain injury after cerebral ischemia in mice. Stroke. 2012;43:1376–1382.
    1. Cronstein BN. Adenosine, an endogenous anti‐inflammatory agent. J Appl Physiol. 1994;76:5–13.
    1. Hasko G, Kuhel DG, Chen JF, Schwarzschild MA, Deitch EA, Mabley JG, Marton A, Szabo C. Adenosine inhibits IL‐12 and TNF‐[alpha] production via adenosine A2a receptor‐dependent and independent mechanisms. FASEB J. 2000;14:2065–2074.
    1. Ryzhov S, Zaynagetdinov R, Goldstein AE, Novitskiy SV, Blackburn MR, Biaggioni I, Feoktistov I. Effect of A2B adenosine receptor gene ablation on adenosine‐dependent regulation of proinflammatory cytokines. J Pharmacol Exp Ther. 2008;324:694–700.
    1. Lappas CM, Rieger JM, Linden J. A2A adenosine receptor induction inhibits IFN‐gamma production in murine CD4+ T cells. J Immunol. 2005;174:1073–1080.
    1. Linden J, Cekic C. Regulation of lymphocyte function by adenosine. Arterioscler Thromb Vasc Biol. 2012;32:2097–2103.
    1. Deaglio S, Dwyer KM, Gao W, Friedman D, Usheva A, Erat A, Chen JF, Enjyoji K, Linden J, Oukka M, Kuchroo VK, Strom TB, Robson SC. Adenosine generation catalyzed by CD39 and CD73 expressed on regulatory T cells mediates immune suppression. J Exp Med. 2007;204:1257–1265.
    1. Mandapathil M, Hilldorfer B, Szczepanski MJ, Czystowska M, Szajnik M, Ren J, Lang S, Jackson EK, Gorelik E, Whiteside TL. Generation and accumulation of immunosuppressive adenosine by human CD4+CD25highFOXP3+ regulatory T cells. J Biol Chem. 2010;285:7176–7186.
    1. Zhang JG, Hepburn L, Cruz G, Borman RA, Clark KL. The role of adenosine A2A and A2B receptors in the regulation of TNF‐alpha production by human monocytes. Biochem Pharmacol. 2005;69:883–889.
    1. Csoka B, Himer L, Selmeczy Z, Vizi ES, Pacher P, Ledent C, Deitch EA, Spolarics Z, Nemeth ZH, Hasko G. Adenosine A2A receptor activation inhibits T helper 1 and T helper 2 cell development and effector function. FASEB J. 2008;22:3491–3499.
    1. Sun WC, Moore JN, Hurley DJ, Vandenplas ML, Murray TF. Effects of stimulation of adenosine A2A receptors on lipopolysaccharide‐induced production of reactive oxygen species by equine neutrophils. Am J Vet Res. 2007;68:649–656.
    1. di Penta A, Moreno B, Reix S, Fernandez‐Diez B, Villanueva M, Errea O, Escala N, Vandenbroeck K, Comella JX, Villoslada P. Oxidative stress and proinflammatory cytokines contribute to demyelination and axonal damage in a cerebellar culture model of neuroinflammation. PLoS One. 2013;8:e54722.
    1. Ryzhov S, Biktasova A, Goldstein AE, Zhang Q, Biaggioni I, Dikov MM, Feoktistov I. Role of JunB in adenosine A2B receptor‐mediated vascular endothelial growth factor production. Mol Pharmacol. 2014;85:62–73.
    1. Adair TH, Cotten R, Gu JW, Pryor JS, Bennett KR, McMullan MR, McDonnell P, Montani JP. Adenosine infusion increases plasma levels of VEGF in humans. BMC Physiol. 2005;5:10.
    1. Pichiule P, Chavez JC, Xu K, LaManna JC. Vascular endothelial growth factor upregulation in transient global ischemia induced by cardiac arrest and resuscitation in rat brain. Brain Res Mol Brain Res. 1999;74:83–90.
    1. Zhou L, Lin Q, Wang P, Yao L, Leong K, Tan Z, Huang Z. Enhanced neuroprotective efficacy of bone marrow mesenchymal stem cells co‐overexpressing BDNF and VEGF in a rat model of cardiac arrest‐induced global cerebral ischemia. Cell Death Dis. 2017;8:e2774.
    1. Seder DB, Van der Kloot TE. Methods of cooling: practical aspects of therapeutic temperature management. Crit Care Med. 2009;37:S211–S222.
    1. Koyasu S, Moro K. Role of innate lymphocytes in infection and inflammation. Front Immunol. 2012;3:101.
    1. Corthay A. How do regulatory T cells work? Scand J Immunol. 2009;70:326–336.
    1. Belge KU, Dayyani F, Horelt A, Siedlar M, Frankenberger M, Frankenberger B, Espevik T, Ziegler‐Heitbrock L. The proinflammatory CD14+CD16+DR++ monocytes are a major source of TNF. J Immunol. 2002;168:3536–3542.
    1. Khoa ND, Montesinos MC, Reiss AB, Delano D, Awadallah N, Cronstein BN. Inflammatory cytokines regulate function and expression of adenosine A(2A) receptors in human monocytic THP‐1 cells. J Immunol. 2001;167:4026–4032.
    1. Venet F, Cour M, Demaret J, Monneret G, Argaud L. Decreased monocyte HLA‐DR expression in patients after non‐shockable out‐of‐hospital cardiac arrest. Shock. 2016;46:33–36.
    1. Resta V, Novelli E, Di Virgilio F, Galli‐Resta L. Neuronal death induced by endogenous extracellular ATP in retinal cholinergic neuron density control. Development. 2005;132:2873–2882.
    1. Antonioli L, Pacher P, Vizi ES, Hasko G. CD39 and CD73 in immunity and inflammation. Trends Mol Med. 2013;19:355–367.
    1. Vreugdenhil AC, Snoek AM, van ‘t Veer C, Greve JW, Buurman WA. LPS‐binding protein circulates in association with apoB‐containing lipoproteins and enhances endotoxin‐LDL/VLDL interaction. J Clin Invest. 2001;107:225–234.
    1. Deng G, Carter J, Traystman RJ, Wagner DH, Herson PS. Pro‐inflammatory T‐lymphocytes rapidly infiltrate into the brain and contribute to neuronal injury following cardiac arrest and cardiopulmonary resuscitation. J Neuroimmunol. 2014;274:132–140.
    1. Qi Z, Liu Q, Zhang Q, Liu B, Li C. Overexpression of programmed cell death‐1 and human leucocyte antigen‐DR on circulatory regulatory T cells in out‐of‐hospital cardiac arrest patients in the early period after return of spontaneous circulation. Resuscitation. 2018;130:13–20.
    1. Paes‐de‐Carvalho R, Maia GA, Ferreira JM. Adenosine regulates the survival of avian retinal neurons and photoreceptors in culture. Neurochem Res. 2003;28:1583–1590.
    1. Serchov T, Atas HC, Normann C, van Calker D, Biber K. Genetically controlled upregulation of adenosine A(1) receptor expression enhances the survival of primary cortical neurons. Mol Neurobiol. 2012;46:535–544.
    1. Wiese S, Jablonka S, Holtmann B, Orel N, Rajagopal R, Chao MV, Sendtner M. Adenosine receptor A2A‐R contributes to motoneuron survival by transactivating the tyrosine kinase receptor TrkB. Proc Natl Acad Sci USA. 2007;104:17210–17215.

Source: PubMed

3
Předplatit