Comprehensive Physiology Wiley Online Library

Ischemia/Reperfusion

Full Article on Wiley Online Library



ABSTRACT

Ischemic disorders, such as myocardial infarction, stroke, and peripheral vascular disease, are the most common causes of debilitating disease and death in westernized cultures. The extent of tissue injury relates directly to the extent of blood flow reduction and to the length of the ischemic period, which influence the levels to which cellular ATP and intracellular pH are reduced. By impairing ATPase‐dependent ion transport, ischemia causes intracellular and mitochondrial calcium levels to increase (calcium overload). Cell volume regulatory mechanisms are also disrupted by the lack of ATP, which can induce lysis of organelle and plasma membranes. Reperfusion, although required to salvage oxygen‐starved tissues, produces paradoxical tissue responses that fuel the production of reactive oxygen species (oxygen paradox), sequestration of proinflammatory immunocytes in ischemic tissues, endoplasmic reticulum stress, and development of postischemic capillary no‐reflow, which amplify tissue injury. These pathologic events culminate in opening of mitochondrial permeability transition pores as a common end‐effector of ischemia/reperfusion (I/R)‐induced cell lysis and death. Emerging concepts include the influence of the intestinal microbiome, fetal programming, epigenetic changes, and microparticles in the pathogenesis of I/R. The overall goal of this review is to describe these and other mechanisms that contribute to I/R injury. Because so many different deleterious events participate in I/R, it is clear that therapeutic approaches will be effective only when multiple pathologic processes are targeted. In addition, the translational significance of I/R research will be enhanced by much wider use of animal models that incorporate the complicating effects of risk factors for cardiovascular disease. © 2017 American Physiological Society. Compr Physiol 7:113‐170, 2017.

Comprehensive Physiology offers downloadable PowerPoint presentations of figures for non-profit, educational use, provided the content is not modified and full credit is given to the author and publication.

Download a PowerPoint presentation of all images


Figure 1. Figure 1. Major pathologic events contributing to ischemia/reperfusion injury. When the blood supply is markedly reduced or absent, ischemic cells switch to anaerobic metabolism to provide ATP. However, this results in cellular acidosis and insufficient ATP production to meet metabolic demand. As a consequence, ATPases are inactivated, while active Ca2+ efflux and Ca2+ reuptake by the endoplasmic reticulum are markedly reduced, with the net effect of this abherent ion transport producing Ca2+ overload in the cell. In addition, xanthine dehydrogenase is converted to XO during ischemia (see Fig. 7), coincident with accumulation of hypoxanthine, one of the substrates required to drive its enzymatic activity. On reperfusion, the delivery of oxygen and substrates required for aerobic ATP generation is restored as is extracellular pH via washout of accumulated H+ (pH paradox). The latter event promotes additional Ca2+ influx (calcium paradox), while the influx of oxygen fuels XO‐driven production of ROS (oxygen paradox) (see Fig. 7). ROS produced by this and other mechanisms can damage virtually every biomolecule found in cells, promote opening of mitochondrial PTPs, and activate inflammatory and thrombogenic cascades to exacerbate cell injury. The latter events are further amplified by release of danger signals (e.g., ATP) and other proinflammatory and thrombogenic mediators from damaged cells (see text for further explanation). The ensuing massive influx of immunocytes at previously ischemic sites contribute to cell injury via the NADPH oxidase‐driven respiratory burst, release of hydrolytic enzymes, and production of MPO‐derived hypochlorous acid and N‐chloramines. The development of the capillary no‐reflow phenomenon during reperfusion results in nutritive perfusion impairment by mechanisms outlined in Figure 11.
Figure 2. Figure 2. Total injury sustained by a tissue subjected to ischemia followed by reperfusion (I/R) (black bars) is attributable to ischemia per se (blue bars) and a component that is due to reestablishing the blood supply (red bars). At the onset of prolonged ischemia two separate general pathologic processes are initiated. The first are processes of tissue injury that are due to ischemia per se. The second are biochemical changes that occur during ischemia that contribute to the surge in generation of reactive oxygen species and infiltration of proinflammatory neutrophils and other immunocytes when molecular oxygen is reintroduced to the tissues during reperfusion. For a treatment to be effective in reducing cellular dysfunction and/or death when administered at the onset of reperfusion (therapeutic window), reestablishing the blood supply must occur before damage attributable to ischemia per se exceeds the viability threshold for irreversible damage. Concepts from Bulkley, 1987 (100).
Figure 3. Figure 3. Tissue responses to ischemia/reperfusion are bimodal (trimodal in the heart), depending on the duration and magnitude of ischemia. Prolonged and severe ischemia induces cell damage that progresses to infarction, with reperfusion paradoxically exacerbating tissue injury by invoking inflammatory responses. In the heart, shorter bouts of ischemia (5‐20 min duration) induce myocardial stunning, wherein contractile function is initially impaired on reperfusion, but slowly improves, without progression to infarction and in the absence of significant inflammation. On the other hand, prolonged exposure to subacute levels of ischemia without reperfusion may induce myocardial hibernation, wherein cardiac cells modify their metabolic phenotype to survive but with a cost of reduced mechanical function. The third mode of response is exemplified by the tissue response to short periods of ischemia (<5 min) followed by reperfusion (ischemic conditioning) that do not produce detectable injury or dysfunction. Far from being innocuous and functionally inert, the response of all organs to such conditioning ischemia is characterized by activation of cell survival programs that confer tolerance to the deleterious effects induced by subsequent exposure to prolonged I/R such that postischemic injury is dramatically reduced. Cardioprotective effects are invoked when tissues are exposed to short bouts of conditioning I/R prior to (ischemic preconditioning) or during (ischemic per‐conditioning) prolonged ischemia or at the onset of reperfusion after prolonged cessation of blood flow (ischemic postconditioning). Tolerance to prolonged I/R in one organ can also be activated by subjecting distant organs to conditioning I/R, a remote effect that can also magnify the beneficial actions of local conditioning.
Figure 4. Figure 4. Ingestion of probiotic diets modifies the composition profile of the oral and enteric microbiome to limit I/R via microflora‐dependent alterations that decrease risk for cardiovascular disease via reductions in blood pressure, oral pathogens, blood LDL and total cholesterol, preservation of endothelium‐dependent vasodilator mechanisms, activation of anti‐inflammatory and infarct‐sparing cell survival programs, and improved postischemic tissue remodeling.
Figure 5. Figure 5. The presence of coexisting risk factors including metabolic syndrome, obesity, diabetes, advancing age, smoking, and dyslipidemias not only increase the likelihood for cardiovascular disease, but also worsen the outcome for those individuals who do suffer a heart attack or stroke. Interestingly, while ischemic and pharmacologic conditioning strategies are remarkably effective in young, healthy subjects, the presence of the aforementioned comorbid factors reduces their cardioprotective effects. The mechanisms underlying the impaired efficacy of conditioning is listed below each of the italicized co‐morbid risk factors in the figure. Surprisingly little attention has been devoted to the effect of cigarette smoking to limit the efficacy of conditioning or with regard to the mechanisms by which this impairment occurs. Caffeine consumption also reduces the effectiveness preconditioning, as does the ingestion of alcoholic beverages at high levels, an effect that disappears as the absorbed ethanol is metabolized and eliminated from the blood. While use of some recreational drugs (eg, cocaine) abolishes ischemic preconditioning, morphine (or other opioids) injections or smoking marijuana may induce preconditioned phenotypes via activation opioid and cannabinoid receptors, respectively. It is also important to note that many of the drugs commonly used in the therapeutic management of patients with cardiovascular disease who are at high risk for myocardial infarction or stroke reduce or abolish the effectiveness of preconditioning stimuli by affecting their underlying signaling mechanisms. Reproduced from Ref. 451, with permission.
Figure 6. Figure 6. Cell death modalities in ischemia/reperfusion (I/R). I/R‐induced necrosis generally occurs as a result of dysfunctional ion transport mechanisms, which causes cells to swell and eventually burst, effects that are exacerbated by plasma membrane damage. Release of proinflammatory mediators and damaged biomolecules initiates the influx of inflammatory cells such as neutrophils, which disrupt the extracellular matrix and cause damage to parenchyal cells by release of cytotoxic oxidants and hydrolytic enzymes. Apopotosis is a regulated form of cell death that causes cell shrinkage and condensation of the cytosol and nucleus, which eventually form apoptotic bodies. Because they are surrounded by cell membranes, apoptotic bodies can be engulfed and digested by phagocytes without evoking an inflammatory response. Autophagy provides a mechanism to remove damaged or senescent protein aggregates and organelles by enclosing them in membrane‐lined vesicles called proteasomes which fuse with lysosomes containing enzymes that degrade the ingested material, usually without evoking an inflammatory response. While normally performing this “housekeeping” function, autophagy may also provide cells with a survival mechanism to withstand the deleterious effects of ischemia, by generating amino acids and fatty acids for cell function. However, when uncontrolled, autophagy contributes to ischemic cell death. While necrosis was once believed to occur from non‐specific trauma or injury as a result of I/R, it now appears that postischemic infarction may also be attributable to programmed events that require a dedicated molecular circuitry that has been termed programmed necrosis or necroptosis. Necroptosis is initiated by TNF‐like cytokines that activate RIP kinases to mediate necrosis via increased production of reactive oxygen species and calcium overload, which in turn modulate the mitochondrial permeability transition pore (MPTP), leading to dissipation of the proton electrochemical gradient, with subsequent ATP depletion, further ROS production, and swelling and rupture of mitochondrial membranes. Recent genetic studies have suggested that the MPTP is predominantly involved in a second form of regulated necrosis that is designated MPT‐RN that is critically dependent on cyclophilin D. Parthanotos can be distinguished from other forms of programmed cell death by its requirement for poly‐ADP‐ribose polymerase activation. Two newly described cell death modalities have been implicated in I/R, ferroptosis and oxytosis. Both involve inhibition of the cytine‐glutamate antiporter Xc, but differ in their modes of lipid peroxidation, being iron dependent and lipoxygenase dependent, respectively.
Figure 7. Figure 7. Mechanism for XO‐dependent production of ROS at the onset of reperfusion. During the period of ischemia, ATP is step‐wise catabolized to hypoxanthine, which accumulates in the tissues because the lack of blood flow does not wash out metabolites from the tissues. Coincident with these changes, xanthine dehydrogenase is converted to XO by a proteolytic mechanism. Thus, a requisite substrate (hypoxanthine) and the activated enzyme (XO) are present in excess in ischemic tissues, but the oxidation of hypoxanthine to xanthine and uric acid cannot proceed, owing to the lack of molecular oxygen that is required to fuel the reaction. On reperfusion, this requisite substrate is suddenly resupplied to the tissue, which fuels the rapid overproduction of ROS. ROS‐induced formation of chemoattractants promotes leukocyte infiltration, neutrophils in particular, which in turn exacerbate cellular injury via NADPH oxidase‐dependent respiratory burst, MPO‐mediated formation of hypochlorous acid, N‐chloramines, and 2‐chloro fatty acids (2ClFA), and release and activation of hydrolytic enzymes that target every type of biomolecule found in cells and tissues.
Figure 8. Figure 8. Generation of ROS by mitochondria (mitoROS) is a nexus for both activation of cell survival programs that mediate the effect of conditioning stimuli to enhance tolerance to I/R and serves as a focal point for overexuberant ROS‐induced ROS release that contributes to the pathogenesis of cell injury in I/R. On the one hand, ROS triggers the activation of cell survival programs in responses to a number of mildly noxious stimuli, such as short bouts of ischemia or antecedent ethanol exposure or pharmacologic agents (activators of mitochondrial ATP‐sensitive potassium (mKATP) or large conductance, calcium‐activated potassium (BKCa) channels. The enhanced tolerance to ischemia invoked by these mitoROS‐dependent conditioning stimuli, which can be delivered before (preconditioning), during (perconditioning) or at the onset of reperfusion (postconditioning), activate protective protein kinases, such as PKCϵ, the expression of prosurvival genes (e.g., heme oxygenase‐1) and mitochondrial antioxidant defenses (e.g., MnSOD, aldehyde dehydrogenase‐1, or ALDH2), as well as targeting the MPTP to maintain the channel in a closed state. On the other hand, overexuberant ROS generation at the onset of reperfusion, driven by ROS‐induced ROS release that is fueled by electron transport chain dysfunction, especially at complexes I and III, and enhanced activities of p66Shc, MAO, and NADPH oxidase‐4 (Nox4) in mitochondria, causes the MPTP to open, leading to swelling, cell disruption, and death. Not depicted is the effect of oxidants to alter the balance of mitochondrial fission and fusion in conditioning and I/R, which emerging evidence has implicated as contributory to both processes. The dual nature of ROS as protective vs damaging species relates to the type of ROS produced, their concentrations, and/or the compartmental localization for their production. MIcroRNAs (miR's) formed during conditioning and during I/R produce changes in mitochondrial energy metabolism, apoptosis, mitochondrial fission/fusion balance, and ROS production. See text for further explanation. Modified from Refs. 408 and 733.
Figure 9. Figure 9. Functional roles and target genes for miRNAs implicated in ischemia/reperfusion and preconditioning. See text for further explanation.
Figure 10. Figure 10. Neutrophil trafficking to ischemic sites occurs during reperfusion of ischemic tissues and involves 11 distinct steps. See text for further explanation. Figure modified from Refs. 923a and 816a.
Figure 11. Figure 11. Mechanisms underlying the development of the postischemic capillary no‐reflow phenomenon. See text for explanation.
Figure 12. Figure 12. Overnutrition via consumption of a Western diet rich in red meats, lipids, and fructose and lacking fermentable fiber may contribute to a shift in the intestinal microbiome to a profile that favors enhanced risk for myocardial infarction, stroke, and peripheral vascular disease. As one example, consumption of protein‐rich diets containing red meats leads to the liberation of carnitine and phosphatidyl choline as the bolus of ingested food is digested in the small bowel, which are converted to TMA by colonic microbes. Upon absorption, TMA is metabolized to the proatherogenic TMAO by the liver to promote the formation of atherosclerotic plaques. The problem is exacerbated by diet‐induced dysbiosis, which favors the translocation of bacteria into the bloodstream and promotion of plaque formation. Interestingly, a blood‐like microbiome exists in plaques that resembles the patient's oral and intestinal microbial profiles, with some of the bacterial species correlating with plasma cholesterol levels. TMAO also contributes to a prothrombogenic phenotype via effects to increase platelet IP3 levels, thereby elevating intracellular calcium levels to promote platelet aggregration and adhesion to injured endothelium.


Figure 1. Major pathologic events contributing to ischemia/reperfusion injury. When the blood supply is markedly reduced or absent, ischemic cells switch to anaerobic metabolism to provide ATP. However, this results in cellular acidosis and insufficient ATP production to meet metabolic demand. As a consequence, ATPases are inactivated, while active Ca2+ efflux and Ca2+ reuptake by the endoplasmic reticulum are markedly reduced, with the net effect of this abherent ion transport producing Ca2+ overload in the cell. In addition, xanthine dehydrogenase is converted to XO during ischemia (see Fig. 7), coincident with accumulation of hypoxanthine, one of the substrates required to drive its enzymatic activity. On reperfusion, the delivery of oxygen and substrates required for aerobic ATP generation is restored as is extracellular pH via washout of accumulated H+ (pH paradox). The latter event promotes additional Ca2+ influx (calcium paradox), while the influx of oxygen fuels XO‐driven production of ROS (oxygen paradox) (see Fig. 7). ROS produced by this and other mechanisms can damage virtually every biomolecule found in cells, promote opening of mitochondrial PTPs, and activate inflammatory and thrombogenic cascades to exacerbate cell injury. The latter events are further amplified by release of danger signals (e.g., ATP) and other proinflammatory and thrombogenic mediators from damaged cells (see text for further explanation). The ensuing massive influx of immunocytes at previously ischemic sites contribute to cell injury via the NADPH oxidase‐driven respiratory burst, release of hydrolytic enzymes, and production of MPO‐derived hypochlorous acid and N‐chloramines. The development of the capillary no‐reflow phenomenon during reperfusion results in nutritive perfusion impairment by mechanisms outlined in Figure 11.


Figure 2. Total injury sustained by a tissue subjected to ischemia followed by reperfusion (I/R) (black bars) is attributable to ischemia per se (blue bars) and a component that is due to reestablishing the blood supply (red bars). At the onset of prolonged ischemia two separate general pathologic processes are initiated. The first are processes of tissue injury that are due to ischemia per se. The second are biochemical changes that occur during ischemia that contribute to the surge in generation of reactive oxygen species and infiltration of proinflammatory neutrophils and other immunocytes when molecular oxygen is reintroduced to the tissues during reperfusion. For a treatment to be effective in reducing cellular dysfunction and/or death when administered at the onset of reperfusion (therapeutic window), reestablishing the blood supply must occur before damage attributable to ischemia per se exceeds the viability threshold for irreversible damage. Concepts from Bulkley, 1987 (100).


Figure 3. Tissue responses to ischemia/reperfusion are bimodal (trimodal in the heart), depending on the duration and magnitude of ischemia. Prolonged and severe ischemia induces cell damage that progresses to infarction, with reperfusion paradoxically exacerbating tissue injury by invoking inflammatory responses. In the heart, shorter bouts of ischemia (5‐20 min duration) induce myocardial stunning, wherein contractile function is initially impaired on reperfusion, but slowly improves, without progression to infarction and in the absence of significant inflammation. On the other hand, prolonged exposure to subacute levels of ischemia without reperfusion may induce myocardial hibernation, wherein cardiac cells modify their metabolic phenotype to survive but with a cost of reduced mechanical function. The third mode of response is exemplified by the tissue response to short periods of ischemia (<5 min) followed by reperfusion (ischemic conditioning) that do not produce detectable injury or dysfunction. Far from being innocuous and functionally inert, the response of all organs to such conditioning ischemia is characterized by activation of cell survival programs that confer tolerance to the deleterious effects induced by subsequent exposure to prolonged I/R such that postischemic injury is dramatically reduced. Cardioprotective effects are invoked when tissues are exposed to short bouts of conditioning I/R prior to (ischemic preconditioning) or during (ischemic per‐conditioning) prolonged ischemia or at the onset of reperfusion after prolonged cessation of blood flow (ischemic postconditioning). Tolerance to prolonged I/R in one organ can also be activated by subjecting distant organs to conditioning I/R, a remote effect that can also magnify the beneficial actions of local conditioning.


Figure 4. Ingestion of probiotic diets modifies the composition profile of the oral and enteric microbiome to limit I/R via microflora‐dependent alterations that decrease risk for cardiovascular disease via reductions in blood pressure, oral pathogens, blood LDL and total cholesterol, preservation of endothelium‐dependent vasodilator mechanisms, activation of anti‐inflammatory and infarct‐sparing cell survival programs, and improved postischemic tissue remodeling.


Figure 5. The presence of coexisting risk factors including metabolic syndrome, obesity, diabetes, advancing age, smoking, and dyslipidemias not only increase the likelihood for cardiovascular disease, but also worsen the outcome for those individuals who do suffer a heart attack or stroke. Interestingly, while ischemic and pharmacologic conditioning strategies are remarkably effective in young, healthy subjects, the presence of the aforementioned comorbid factors reduces their cardioprotective effects. The mechanisms underlying the impaired efficacy of conditioning is listed below each of the italicized co‐morbid risk factors in the figure. Surprisingly little attention has been devoted to the effect of cigarette smoking to limit the efficacy of conditioning or with regard to the mechanisms by which this impairment occurs. Caffeine consumption also reduces the effectiveness preconditioning, as does the ingestion of alcoholic beverages at high levels, an effect that disappears as the absorbed ethanol is metabolized and eliminated from the blood. While use of some recreational drugs (eg, cocaine) abolishes ischemic preconditioning, morphine (or other opioids) injections or smoking marijuana may induce preconditioned phenotypes via activation opioid and cannabinoid receptors, respectively. It is also important to note that many of the drugs commonly used in the therapeutic management of patients with cardiovascular disease who are at high risk for myocardial infarction or stroke reduce or abolish the effectiveness of preconditioning stimuli by affecting their underlying signaling mechanisms. Reproduced from Ref. 451, with permission.


Figure 6. Cell death modalities in ischemia/reperfusion (I/R). I/R‐induced necrosis generally occurs as a result of dysfunctional ion transport mechanisms, which causes cells to swell and eventually burst, effects that are exacerbated by plasma membrane damage. Release of proinflammatory mediators and damaged biomolecules initiates the influx of inflammatory cells such as neutrophils, which disrupt the extracellular matrix and cause damage to parenchyal cells by release of cytotoxic oxidants and hydrolytic enzymes. Apopotosis is a regulated form of cell death that causes cell shrinkage and condensation of the cytosol and nucleus, which eventually form apoptotic bodies. Because they are surrounded by cell membranes, apoptotic bodies can be engulfed and digested by phagocytes without evoking an inflammatory response. Autophagy provides a mechanism to remove damaged or senescent protein aggregates and organelles by enclosing them in membrane‐lined vesicles called proteasomes which fuse with lysosomes containing enzymes that degrade the ingested material, usually without evoking an inflammatory response. While normally performing this “housekeeping” function, autophagy may also provide cells with a survival mechanism to withstand the deleterious effects of ischemia, by generating amino acids and fatty acids for cell function. However, when uncontrolled, autophagy contributes to ischemic cell death. While necrosis was once believed to occur from non‐specific trauma or injury as a result of I/R, it now appears that postischemic infarction may also be attributable to programmed events that require a dedicated molecular circuitry that has been termed programmed necrosis or necroptosis. Necroptosis is initiated by TNF‐like cytokines that activate RIP kinases to mediate necrosis via increased production of reactive oxygen species and calcium overload, which in turn modulate the mitochondrial permeability transition pore (MPTP), leading to dissipation of the proton electrochemical gradient, with subsequent ATP depletion, further ROS production, and swelling and rupture of mitochondrial membranes. Recent genetic studies have suggested that the MPTP is predominantly involved in a second form of regulated necrosis that is designated MPT‐RN that is critically dependent on cyclophilin D. Parthanotos can be distinguished from other forms of programmed cell death by its requirement for poly‐ADP‐ribose polymerase activation. Two newly described cell death modalities have been implicated in I/R, ferroptosis and oxytosis. Both involve inhibition of the cytine‐glutamate antiporter Xc, but differ in their modes of lipid peroxidation, being iron dependent and lipoxygenase dependent, respectively.


Figure 7. Mechanism for XO‐dependent production of ROS at the onset of reperfusion. During the period of ischemia, ATP is step‐wise catabolized to hypoxanthine, which accumulates in the tissues because the lack of blood flow does not wash out metabolites from the tissues. Coincident with these changes, xanthine dehydrogenase is converted to XO by a proteolytic mechanism. Thus, a requisite substrate (hypoxanthine) and the activated enzyme (XO) are present in excess in ischemic tissues, but the oxidation of hypoxanthine to xanthine and uric acid cannot proceed, owing to the lack of molecular oxygen that is required to fuel the reaction. On reperfusion, this requisite substrate is suddenly resupplied to the tissue, which fuels the rapid overproduction of ROS. ROS‐induced formation of chemoattractants promotes leukocyte infiltration, neutrophils in particular, which in turn exacerbate cellular injury via NADPH oxidase‐dependent respiratory burst, MPO‐mediated formation of hypochlorous acid, N‐chloramines, and 2‐chloro fatty acids (2ClFA), and release and activation of hydrolytic enzymes that target every type of biomolecule found in cells and tissues.


Figure 8. Generation of ROS by mitochondria (mitoROS) is a nexus for both activation of cell survival programs that mediate the effect of conditioning stimuli to enhance tolerance to I/R and serves as a focal point for overexuberant ROS‐induced ROS release that contributes to the pathogenesis of cell injury in I/R. On the one hand, ROS triggers the activation of cell survival programs in responses to a number of mildly noxious stimuli, such as short bouts of ischemia or antecedent ethanol exposure or pharmacologic agents (activators of mitochondrial ATP‐sensitive potassium (mKATP) or large conductance, calcium‐activated potassium (BKCa) channels. The enhanced tolerance to ischemia invoked by these mitoROS‐dependent conditioning stimuli, which can be delivered before (preconditioning), during (perconditioning) or at the onset of reperfusion (postconditioning), activate protective protein kinases, such as PKCϵ, the expression of prosurvival genes (e.g., heme oxygenase‐1) and mitochondrial antioxidant defenses (e.g., MnSOD, aldehyde dehydrogenase‐1, or ALDH2), as well as targeting the MPTP to maintain the channel in a closed state. On the other hand, overexuberant ROS generation at the onset of reperfusion, driven by ROS‐induced ROS release that is fueled by electron transport chain dysfunction, especially at complexes I and III, and enhanced activities of p66Shc, MAO, and NADPH oxidase‐4 (Nox4) in mitochondria, causes the MPTP to open, leading to swelling, cell disruption, and death. Not depicted is the effect of oxidants to alter the balance of mitochondrial fission and fusion in conditioning and I/R, which emerging evidence has implicated as contributory to both processes. The dual nature of ROS as protective vs damaging species relates to the type of ROS produced, their concentrations, and/or the compartmental localization for their production. MIcroRNAs (miR's) formed during conditioning and during I/R produce changes in mitochondrial energy metabolism, apoptosis, mitochondrial fission/fusion balance, and ROS production. See text for further explanation. Modified from Refs. 408 and 733.


Figure 9. Functional roles and target genes for miRNAs implicated in ischemia/reperfusion and preconditioning. See text for further explanation.


Figure 10. Neutrophil trafficking to ischemic sites occurs during reperfusion of ischemic tissues and involves 11 distinct steps. See text for further explanation. Figure modified from Refs. 923a and 816a.


Figure 11. Mechanisms underlying the development of the postischemic capillary no‐reflow phenomenon. See text for explanation.


Figure 12. Overnutrition via consumption of a Western diet rich in red meats, lipids, and fructose and lacking fermentable fiber may contribute to a shift in the intestinal microbiome to a profile that favors enhanced risk for myocardial infarction, stroke, and peripheral vascular disease. As one example, consumption of protein‐rich diets containing red meats leads to the liberation of carnitine and phosphatidyl choline as the bolus of ingested food is digested in the small bowel, which are converted to TMA by colonic microbes. Upon absorption, TMA is metabolized to the proatherogenic TMAO by the liver to promote the formation of atherosclerotic plaques. The problem is exacerbated by diet‐induced dysbiosis, which favors the translocation of bacteria into the bloodstream and promotion of plaque formation. Interestingly, a blood‐like microbiome exists in plaques that resembles the patient's oral and intestinal microbial profiles, with some of the bacterial species correlating with plasma cholesterol levels. TMAO also contributes to a prothrombogenic phenotype via effects to increase platelet IP3 levels, thereby elevating intracellular calcium levels to promote platelet aggregration and adhesion to injured endothelium.
References
 1.Abdellatif M. Differential expression of microRNAs in different disease states. Circ Res 110: 638‐650, 2012.
 2.Abela CB, Homer‐Vanniasinkham S. Clinical implications of ischaemia‐reperfusion injury. Pathophysiology 9: 229‐240, 2003.
 3.Abonia JP, Friend DS, Austen WG, Jr., Moore FD, Jr., Carroll MC, Chan R, Afnan J, Humbles A, Gerard C, Knight P, Kanaoka Y, Yasuda S, Morokawa N, Austen KF, Stevens RL, Gurish MF. Mast cell protease 5 mediates ischemia‐reperfusion injury of mouse skeletal muscle. J Immunol 174: 7285‐7291, 2005.
 4.Abramov AY, Scorziello A, Duchon M. Three distinct mechanisms generate oxygen free radicals in neurons and contribute to cell death during anoxia and reoxygenation. J Neurosci 27: 1129‐1138, 2007.
 5.Adibhatha RM, Hatche, JF. Lipid oxidation and peroxidation in CNS health and disease: From molecular mechanisms to therapeutic opportunities. Antiox Redox Signal 12: 125‐169, 2010.
 6.Aguilar‐Nascimento JE, Salomao AB, Nochi RJ, Jr., Nascimento M, Neves S. Intraluminal injection of short chain fatty acids diminishes intestinal mucosa injury in experimental ischemia‐reperfusion. Acta Cir Bras 21: 21‐25, 2006.
 7.Ahern PP, Izcue A, Maloy KJ, Powrie F. The interleukin‐23 axis in intestinal inflammation. Immunol Rev 226: 147‐159, 2008.
 8.Ahluwalia A, De Felipe C, O'Brien J, Hunt SP, Perretti M. Impaired IL‐1beta‐induced neutrophil accumulation in tachykinin NK1 receptor knockout mice. Br J Pharmacol 124: 1013‐1015, 1998.
 9.Aikawa R, Komuro I, Yamazaki T, Zou Y, Kudoh S, Tanaka M, Shiojima I, Hiroi Y, Yazaki Y. Oxidative stress activates extracellualar signal‐regulated kinases through Src and Ras in cultured cardiac myocytes of neonatal rats. J Clin Invest 100: 1813‐1821, 1997.
 10.Aiken CE, Ozanne SE. Transgenerational developmental programming. Hum Reprod Update 20: 63‐75, 2014.
 11.Aikens J, Dix TA. Perhydroxyl radical (HOO.) initiated lipid peroxidation. The role of fatty acid hydroperoxides. J Biol Chem 266: 15091‐15098, 1991.
 12.Akhmedov A, Montecucco F, Braunersreuther V, Camici GG, Jakob P, Reiner MF, Glanzmann M, Burger F, Paneni F, Galan K, Pelli G, Vuilleumier N, Belin A, Vallee JP, Mach F, Luscher TF. Genetic deletion of the adaptor protein p66Shc increases susceptibility to short‐term ischaemic myocardial injury via intracellular salvage pathways. Eur Heart J 36: 516‐526, 2015.
 13.Alam MR, Baetz D, Ovize M. Cyclophilin D and myocardial ischemia‐reperfusion injury: A fresh perspective. J Mol Cell Cardiol 78: 80‐89, 2015.
 14.Alexander BT, Dasinger JH, Intapad S. Fetal programming and cardiovascular pathology. Compr Physiol 5: 997‐1025, 2015.
 15.Alexander JS, Elrod JW. Extracellular matrix, junctional integrity and matrix metalloproteinase interactions in endothelial permeability regulation. J Anat 200: 561‐574, 2002.
 16.Ali ZA, Callaghan CJ, Lim E, Ali AA, Nouraei SA, Akthar AM, Boyle JR, Varty K, Kharbanda RK, Dutka DP, Gaunt ME. Remote ischemic preconditioning reduces myocardial and renal injury after elective abdominal aortic aneurysm repair: A randomized controlled trial. Circulation 116: I98‐105, 2007.
 17.Allen CL, Bayraktutan U. Oxidative stress and its role in the pathogenesis of ischaemic stroke. Int J Stroke 4: 461‐470, 2009.
 18.Alverdy JC, Chang EB. The re‐emerging role of the intestinal microflora in critical illness and inflammation: Why the gut hypothesis of sepsis syndrome will not go away. J Leukoc Biol 83: 461‐466, 2008.
 19.Ames A, III, Wright RL, Kowada M, Thurston JM, Majno G. Cerebral ischemia. II. The no‐reflow phenomenon. Am J Pathol 52: 437‐453, 1968.
 20.Anand SS, Islam S, Rosengren A, Franzosi MG, Steyn K, Yusufali AH, Keltai M, Diaz R, Rangarajan S, Yusuf S. Risk factors for myocardial infarction in women and men: Insights from the INTERHEART study. Eur Heart J 29: 932‐940, 2008.
 21.Anderson CM, Lopez F, Zimmer A, Benoit JN. Placental insufficiency leads to developmental hypertension and mesenteric artery dysfunction in two generations of Sprague‐Dawley rat offspring. Biol Reprod 74: 538‐544, 2006.
 22.Andrabi SA, Dawson TM, Dawson VL. Mitochondrial and nuclear cross talk in cell death: Parthanatos. Ann N Y Acad Sci 1147: 233‐241, 2008.
 23.Andrade‐Oliveira V, Amano MT, Correa‐Costa M, Castoldi A, Felizardo RJ, de Almeida DC, Bassi EJ, Moraes‐Vieira PM, Hiyane MI, Rodas AC, Peron JP, Aguiar CF, Reis MA, Ribeiro WR, Valduga CJ, Curi R, Vinolo MA, Ferreira CM, Camara NO. Gut bacteria products prevent AKI induced by ischemia‐reperfusion. J Am Soc Nephrol 26: 1877‐1888, 2015.
 24.Andres AM, Hernandez G, Lee P, Huang C, Ratliff EP, Sin J, Thornton CA, Damasco MV, Gottlieb RA. Mitophagy is required for acute cardioprotection by simvastatin. Antioxid Redox Signal 21: 1960‐1973, 2014.
 25.Andrews RK, Berndt MC. Platelet physiology and thrombosis. Thromb Res 114: 447‐453, 2004.
 26.Anea CB, Zhang M, Chen F, Ali MI, Hart CM, Stepp DW, Kovalenkov YO, Merloiu AM, Pati P, Fulton D, Rudic RD. Circadian clock control of Nox4 and reactive oxygen species in the vasculature. PLoS One 8: e78626, 2013.
 27.Angelillo‐Scherrer A. Leukocyte‐derived microparticles in vascular homeostasis. Circ Res 110: 356‐369, 2011.
 28.Antonopoulos A, Kyriacou C, Kazianis G. Significance of endothelin‐1 in myocardial infarction. Hellenic J Cardiol 48: 161‐164, 2007.
 29.Arany I, Faisa A, Clark JS, Vera T, Baliga R, Nagamine Y. p66Shc‐mediated mitochondrial dysfunction in renal proximal tubule cells during oxidative injury. Am J Physiol 298: F1214‐F1221, 2010.
 30.Arrell DK, Elliott ST, Kane LA, Guo Y, Ko YH, Pedersen PL, Robinson J, Murata M, Murphy AM, Marban E, Van Eyk JE. Proteomic analysis of pharmacological preconditioning: Novel protein targets converge to mitochondrial metabolism pathways. Circ Res 99: 706‐714, 2006.
 31.Aspelund A, Robciuc MR, Karaman S, Makinen T, Alitalo K. Lymphatic system in cardiovascular medicine. Circ Res 118: 515‐530, 2016.
 32.Atarashi K, Hirata T, Matsumoto M, Kanemitsu N, Miyasaka M. Rolling of Th1 cells via P‐selectin glycoprotein ligand‐1 stimulates LFA‐1‐mediated cell binding to ICAM‐1. J Immunol 174: 1424‐1432, 2005.
 33.Aune SE, Herr DJ, Kutz CJ, Menick DR. Histone deacetylases exert class‐specific roles in conditioning the brain and heart against acute ischemic injury. Front Neurol 6: 145, 2015.
 34.Azad N, Iyer A, Vallyathan V, Wang L, Castranova V, Stehlik C, Rojanaskul Y. Role of oxidative/nitrosative stress‐mediated Bcl‐2 regulation in apoptosis and malignant transformation. Ann NY Acad Sci 1203: 1‐6, 2010.
 35.Baines CP. The mitochondrial permeability transition pore and ischemia‐reperfusion injury. Basic Res Cardiol 104: 181‐188, 2009.
 36.Baines CP. The molecular composition of the mitochondrial permeability transition pore. J Mol Cell Cardiol 46: 850‐857, 2009.
 37.Baines CP. The cardiac mitochondrion: Nexus of stress. Annu Rev Physiol 72: 61‐80, 2010.
 38.Baines CP. How and when do myocytes die during ischemia and reperfusion: The late phase. J Cardiovasc Pharmacol Ther 16: 239‐243, 2011.
 39.Baines CP, Kaiser RA, Purcell NH, Blair NS, Osinska H, Hambleton MA, Brunskill EW, Sayen MR, Gottlieb RA, Dorn GW, Robbins J, Molkentin JD. Loss of cyclophilin D reveals a critical role for mitochondrial permeability transition in cell death. Nature 434: 658‐662, 2005.
 40.Baines CP, Zhang J, Wang GW, Zheng YT, Xiu JX, Cardwell EM, Bolli R, Ping P. Mitochondrial PKCe and MAPK form signaling modules in the murine heart: Enhanced mitochondrial PKCe‐MAPK interactions and differential MAPK activation in PKCe‐induced cardioprotection. Circ Res 90: 390‐7, 2002.
 41.Baldwin WM, III, Su CA, Shroka TM, Fairchild RL. Experimental models of cardiac transplantation: Design determines relevance. Curr Opin Organ Transplant 19: 525‐530, 2014.
 42.Balsam LB, Wagers AJ, Christensen JL, Kofidis T, Weissman IL, Robbins RC. Haematopoietic stem cells adopt mature haematopoietic fates in ischaemic myocardium. Nature 428: 668‐673, 2004.
 43.Bang C, Fiedler J, Thum T. Cardiovascular importance of the microRNA‐23/27/24 family. Microcirculation 19: 208‐214, 2012.
 44.Barchowsky A, Munro SR, Morana SJ, Vincenti MP, Treadwell M. Oxidant‐sensitive and phosphorylation‐dependent activation of NF‐kappa B and AP‐1 in endothelial cells. Am J Physiol 269: L829‐836.
 45.Barker DJ. Fetal origins of coronary heart disease. BMJ 311: 171‐174, 1995.
 46.Barker DJ, Osmond C. Infant mortality, childhood nutrition, and ischaemic heart disease in England and Wales. Lancet 1: 1077‐1081, 1986.
 47.Barker DJ, Osmond C. Low birth weight and hypertension. BMJ 297: 134‐135, 1988.
 48.Barker DJ, Thornburg KL. The obstetric origins of health for a lifetime. Clin Obstet Gynecol 56: 511‐9, 2013.
 49.Barnabei MS, Palpant NJ, Metzger JM. Influence of genetic background on ex vivo and in vivo cardiac function in several commonly used inbred mouse strains. Physiol Genomics 42: 103‐113, 2010.
 50.Barone FC, Knudsen DJ, Nelson AH, Feuerstein GZ, Willette RN. Mouse strain differences in susceptibility to cerebral ischemia are related to cerebral vascular anatomy. J Cereb Blood Flow Metab 13: 683‐692, 1993.
 51.Barrabes JA, Inserte J, Mirabet M, Quiroga A, Hernando V, Figueras J, Garcia‐Dorado D. Antagonism of P2Y12 or GPIIb/IIIa receptors reduces platelet‐mediated myocardial injury after ischaemia and reperfusion in isolated rat hearts. Thromb Haemost 104: 128‐135, 2010.
 52.Barth AS, Tomaselli GF. Gene scanning and heart attack risk. Trends Cardiovasc Med 26: 260‐265, 2016.
 53.Bateson P, Gluckman P, Hanson M. The biology of developmental plasticity and the predictive adaptive response hypothesis. J Physiol 592: 2357‐2368, 2014.
 54.Bauer I, Wanner GA, Rensing H, Alte C, Miescher EA, Wolf B, Pannen BH, Clemens MG, Bauer M. Expression pattern of heme oxygenases 1 and 2 in normal and stress‐exposed rat liver. Hepatology 27: 829‐838, 1998.
 55.Bauer M, Bauer I. Heme oxygenase‐1: Redox regulation and role in the hepatic response to oxidative stress. Antioxid Redox Signal 4: 749‐758, 2002.
 56.Baum M. Role of the kidney in the prenatal and early postnatal programming of hypertension. Am J Physiol Renal Physiol 29: F235‐F247, 2010.
 57.Baumgartner WA, Williams GM, Fraser CD, Jr., Cameron DE, Gardner TJ, Burdick JF, Augustine S, Gaul PD, Reitz BA. Cardiopulmonary bypass with profound hypothermia. An optimal preservation method for multiorgan procurement. Transplantation 47, 123‐127, 1989.
 58.Bautista AP, Meszaros K, Bojta J, Spitzer JJ. Superoxide anion generation in the liver during the early stage of endotoxemia in rats. J Leukoc Biol 48: 123‐128, 1990.
 59.Baxter GF. The neutrophil as a mediator of myocardial ischemia‐reperfusion injury: Time to move on. Basic Res Cardiol 97: 268‐275, 2002.
 60.Beldi G, Banz Y, Kroemer A, Sun X, Wu Y, Graubardt N, Rellstab A, Nowak M, Enjyoji K, Li X, Junger WG, Candinas D, Robson SC. Deletion of CD39 on natural killer cells attenuates hepatic ischemia/reperfusion injury in mice. Hepatology 51: 1702‐1711, 2010.
 61.Belmont PJ, Chen WJ, San Pedro MN, Thuerauf DJ, Gellings Lowe N, Gude N, Hilton B, Wolkowicz R, Sussman MA, Glembotski CC. Roles for endoplasmic reticulum‐associated degradation and the novel endoplasmic reticulum stress response gene Derlin‐3 in the ischemic heart. Circ Res 106: 307‐316, 2010.
 62.Belmont PJ, Chen WJ, Thuerauf DJ, Glembotski CC. Regulation of microRNA expression in the heart by the ATF6 branch of the ER stress response. J Mol Cell Cardiol 53: 259‐267, 2012.
 63.Benakis C, Brea D, Caballero S, Faraco G, Moore J, Murphy M, Sita G, Racchumi G, Ling L, Pamer EG, Iadecola C, Anrather J. Commensal microbiota affects ischemic stroke outcome by regulating intestinal gammadelta T cells. Nat Med 22: 516‐523, 2016.
 64.Ben‐Ari Z, Pappo O, Cheporko Y, Yasovich N, Offen D, Shainberg A, Leshem D, Sulkes J, Vidne BA, Hochhauser E. Bax ablation protects against hepatic ischemia/reperfusion injury in transgenic mice. Liver Transpl 13: 1181‐1188, 2007.
 65.Benhar M, Forrester MT, Stamler JS. Protein denitrosylation: Enzymatic mechanisms and cellular functions. Nat Rev Cell Mol Biol 10: 21‐732, 2009.
 66.Bernardo BC, Gao XM, Winbanks CE, Boey EJ, Tham YK, Kiriazis H, Gregorevic P, Obad S, Kauppinen S, Du XJ, Lin RC, McMullen JR. Therapeutic inhibition of the miR‐34 family attenuates pathological cardiac remodeling and improves heart function. Proc Natl Acad Sci U S A 109: 17615‐17620, 2012.
 67.Bhatia M, Saluja AK, Hofbauer B, Frossard JL, Lee HS, Castagliuolo I, Wang CC, Gerard N, Pothoulakis C, Steer ML. Role of substance P and the neurokinin 1 receptor in acute pancreatitis and pancreatitis‐associated lung injury. Proc Natl Acad Sci U S A 95: 4760‐4765, 1998.
 68.Bhatt K, Kato M, Natarajan R. Mini‐review: Emerging roles of microRNAs in the pathophysiology of renal diseases. Am J Physiol Renal Physiol 310: F109‐118, 2016.
 69.Bhattacharya K, Farwell K, Huang M, Kempuraj D, Donelan J, Papaliodis D, Vasiadi M, Theoharides TC. Mast cell deficient W/Wv mice have lower serum IL‐6 and less cardiac tissue necrosis than their normal littermates following myocardial ischemia‐reperfusion. Int J Immunopathol Pharmacol 20: 69‐74, 2007.
 70.Bianchi P, Kunduzova O, Masini E, Cambon C, Bani D, Raimondi L, Seguelas MH, Nistri S, Colucci W, Leducq N, Parini A. Oxidative stress by monoamine oxidase mediates receptor‐independent cardiomyocyte apoptosis by serotonin and postischemic myocardial injury. Circulation 112: 3297‐3305, 2005.
 71.Bilzer M, Roggel F, Gerbes AL. Role of Kupffer cells in host defense and liver disease. Liver Int 26: 1175‐1186, 2006.
 72.Bindoli A, Rigobello MP. Principles in redox signaling: From chemistry to functional significance. Antioxid Redox Signal 18: 1557‐1593, 2013.
 73.Birdsall HH, Green DM, Trial J, Youker KA, Burns AR, MacKay CR, LaRosa GJ, Hawkins HK, Smith CW, Michael LH, Entman ML, Rossen RD. Complement C5a, TGF‐beta 1, and MCP‐1, in sequence, induce migration of monocytes into ischemic canine myocardium within the first one to five hours after reperfusion. Circulation 95: 684‐692, 1997.
 74.Bjorkegren JL, Kovacic JC, Dudley JT, Schadt EE. Genome‐wide significant loci: How important are they? Systems genetics to understand heritability of coronary artery disease and other common complex disorders. J Am Coll Cardiol 65: 830‐845, 2015.
 75.Black M, Wang W. Ischemic stroke: From next generation sequencing and GWAS to community genomics? OMICS 19: 451‐460, 2015.
 76.Bless NM, Warner RL, Padgaonkar VA, Lentsch AB, Czermak BJ, Schmal H, Friedl HP, Ward PA. Roles for C‐X‐C chemokines and C5a in lung injury after hindlimb ischemia‐reperfusion. Am J Physiol 276: L57‐63, 1999.
 77.Bliksoen M, Baysa A, Eide L, Bjoras M, Suganthan R, Vaage J, Stenslokken KO, Valen G. Mitochondrial DNA damage and repair during ischemia‐reperfusion injury of the heart. J Mol Cell Cardiol 78: 9‐22, 2015.
 78.Bliss TM, Kelly S, Shah AK, Foo WC, Kohli P, Stokes C, Sun GH, Ma M, Masel J, Kleppner SR, Schallert T, Palmer T, Steinberg GK. Transplantation of hNT neurons into the ischemic cortex: Cell survival and effect on sensorimotor behavior. J Neurosci Res 83: 1004‐1014, 2006.
 79.Bluhmki E, Chamorro A, Davalos A, Machnig T, Sauce C, Wahlgren N, Wardlaw J, Hacke W. Stroke treatment with alteplase given 3.0‐4.5 h after onset of acute ischaemic stroke (ECASS III): Additional outcomes and subgroup analysis of a randomised controlled trial. Lancet Neurol 8: 1095‐1102, 2009.
 80.Bodi V, Marrachelli VG, Husser O, Chorro FJ, Vina JR, Monleon D. Metabolomics in the diagnosis of acute myocardial ischemia. J Cardiovasc Transl Res 6: 808‐815, 2013.
 81.Boengler K, Schulz R, Heusch G. Loss of cardioprotection with ageing. Cardiovasc Res 83: 247‐261, 2009.
 82.Boersma E, Maas AC, Deckers JW, Simoons ML. Early thrombolytic treatment in acute myocardial infarction: Reappraisal of the golden hour. Lancet 348: 771‐775, 1996.
 83.Bogoyevitch MA, Gillespie‐Brown J, Ketterman AJ, Fuller SJ, Ben‐Levy R, Ashworth A, Marshall CJ, Sugden PH. Stimulation of the stress‐activated mitogen‐activated protein kinase subfamilies in perfused heart. p38/RK mitogen‐activated protein kinases and c‐Jun N‐terminal kinases are activated by ischemia/reperfusion. Circ Res 79: 162‐73, 1996.
 84.Bolli R, Marban E. Molecular and cellular mechanisms of myocardial stunning. Physiol Rev 79: 609‐663, 1999.
 85.Bonder CS, Norman MU, Macrae T, Mangan PR, Weaver CT, Bullard DC, McCafferty DM, Kubes P. P‐selectin can support both Th1 and Th2 lymphocyte rolling in the intestinal microvasculature. Am J Pathol 167: 1647‐1660, 2005.
 86.Boon RA, Dimmeler S. MicroRNAs in myocardial infarction. Nat Rev Cardiol 12: 135‐142, 2015.
 87.Boros P, Bromberg JS. New cellular and molecular immune pathways in ischemia/reperfusion injury. Am J Transplant 6: 652‐658, 2006.
 88.Bortolotto SK, Morrison WA, Han X, Messina A. Mast cells play a pivotal role in ischaemia reperfusion injury to skeletal muscles. Lab Invest 84: 1103‐1111, 2004.
 89.Botker HE, Kharbanda R, Schmidt MR, Bottcher M, Kaltoft AK, Terkelsen CJ, Munk K, Andersen NH, Hansen TM, Trautner S, Lassen JF, Christiansen EH, Krusell LR, Kristensen SD, Thuesen L, Nielsen SS, Rehling M, Sorensen HT, Redington AN, Nielsen TT. Remote ischaemic conditioning before hospital admission, as a complement to angioplasty, and effect on myocardial salvage in patients with acute myocardial infarction: A randomised trial. Lancet 375: 727‐34, 2010.
 90.Boujrad H, Gubkina O, Robert N, Krantic S, Susin SA. AIF‐mediated programmed necrosis: A highly regulated way to die. Cell Cycle 6: 2612‐9, 2007.
 91.Bozic CR, Lu B, Hopken UE, Gereard C, Berard NP. Neurogenic amplification of immune complex inflammation. Science 273: 1722‐1725, 1996.
 92.Braunersreuther V, Jaquet V. Reactive oxygen species in myocardial reperfusion injury: From physiopathology to therapeutic approaches. Curr Pharm Biotechnol 13: 97‐114, 2012.
 93.Bright R, Raval AP, Dembner JM, Pérez‐Pinzón MA, Steinberg GK, Yenari MA, Mochly‐Rosen D. Protein kinase C d mediates cerebral reperfusion injury in vivo. J Neurosci 24: 6880‐6888, 2004.
 94.Broughton BR, Reutens DC, Sobey CG. Apoptotic mechanisms after cerebral ischemia. Stroke 40: e331‐e339, 2009.
 95.Brown KE, Brunt EM, Heinecke JW. Immunohistochemical detection of myeloperoxidase and its oxidation products in Kupffer cells of human liver. Am J Pathol 159: 2081‐2088, 2001.
 96.Brown JM, Hazen SL. The gut microbial endocrine organ: Bacterially derived signals driving cardiometabolic diseases. Annu Rev Med 66: 343‐359, 2015.
 97.Bryan NS, Grisham MB. Methods to detect nitric oxide and its metabolites in biological samples. Free Radic Biol Med 43: 645‐657, 2007.
 98.Buckley CD, Ross EA, McGettrick HM, Osborne CE, Haworth O, Schmutz C, Stone PC, Salmon M, Matharu NM, Vohra RK, Nash GB, Rainger GE. Identification of a phenotypically and functionally distinct population of long‐lived neutrophils in a model of reverse endothelial migration. J Leukoc Biol 79: 303‐311, 2006.
 99.Budas GR, Churchill EN, Mochly‐Rosen D. Cardioprotective mechanisms of PKC isozyme‐selective activators and inhibitors in the treatment of ischemia‐reperfusion injury. Pharmacol Res 55: 523‐536, 2007.
 100.Bulkley GB. Free radical‐mediated reperfusion injury: A selective review. Br J Cancer Suppl 8: 66‐73, 1987.
 101.Burne MJ, Haq M, Matsuse H, Mohapatra S, Rabb H. Genetic susceptibility to renal ischemia reperfusion injury revealed in a murine model. Transplantation 69: 1023‐1025, 2000.
 102.Burne‐Taney MJ, Ascon DB, Daniels F, Racusen L, Baldwin W, Rabb H. B cell deficiency confers protection from renal ischemia reperfusion injury. J Immunol 171: 3210‐3215, 2003.
 103.Burne‐Taney MJ, Yokota‐Ikeda N, Rabb H. Effects of combined T‐ and B‐cell deficiency on murine ischemia reperfusion injury. Am J Transplant 5: 1186‐1193, 2005.
 104.Burns AR, Smith CW, Walker DC. Unique structural features that influence neutrophil emigration into the lung. Physiol Rev 83: 309‐336, 2003.
 105.Cabrera‐Fuentes HA, Ruiz‐Meana M, Simsekyilmaz S, Kostin S, Inserte J, Saffarzadeh M, Galuska SP, Vijayan V, Barba I, Barreto G, Fischer S, Lochnit G, Ilinskaya ON, Baumgart‐Vogt E, Boning A, Lecour S, Hausenloy DJ, Liehn EA, Garcia‐Dorado D, Schluter KD, Preissner KT. RNase1 prevents the damaging interplay between extracellular RNA and tumour necrosis factor‐alpha in cardiac ischaemia/reperfusion injury. Thromb Haemost 112: 1110‐1119, 2014.
 106.Caldeira MV, Salazar IL, Curcio M, Canzoniero LM, Duarte CB. Role of the ubiquitin‐proteasome system in brain ischemia: Friend or foe? Prog Neurobiol 112: 50‐69, 2014.
 107.Caldwell CC, Okaya T, Matignoni A, Husted T, Schuster R, Lentsch AB. Divergent functions of CD4+ T lymphocytes in acute liver inflammation and injury after ischemia‐reperfusion. Am J Physiol 289: G969‐G976, 2005.
 108.Caldwell‐Kenkel JC, Currin RT, Tanaka Y, Thurman PG, Lemasters JJ. Kupffer cell activation and endothelial cell damage aftrer storage of rat livers: Effect of reperfusion. Hepatology 13: 83‐95, 1991.
 109.Calvert JW, Lefer DJ, Gundewar S, Poston L, Coetzee WA. Developmental programming resulting from maternal obesity in mice: Effects on myocardial ischaemia‐reperfusion injury. Exp Physiol 94: 805‐814, 2009.
 110.Candilio L, Hausenloy DJ, Yellon DM. Remote ischemic conditioning: A clinical trial's update. J Cardiovasc Pharmacol Ther 16: 304‐312, 2011.
 111.Cao JY, Dixon SJ. Mechanisms of ferroptosis. Cell Mol Life Sci 73: 2195‐2209, 2016.
 112.Cao T, Pinter E, Al‐Rasheed S, Gerard N, Hoult JR, Brain SD. Neurokinin‐1 receptor agonists are involved in mediating neutrophil accumulation in the inflamed, but not normal, cutaneous microvasculature: An in vivo study using neurokinin‐1 receptor knockout mice. J Immunol 164: 5424‐5429, 2000.
 113.Cappola TP, Margulies KB. Functional genomics applied to cardiovascular medicine. Circulation 124: 87‐94, 2011.
 114.Carati CJ. Changes in macromolecular permeability of microvessels in rat small intestine after total occlusion ischaemia/reperfusion. Microcirc Endothelium Lymphatics 4, 69‐86, 1988.
 115.Carden DL, Granger DN. Pathophysiology of ischemia‐reperfusion injury. J Pathol 190: 255‐266, 2000.
 116.Cardinal J, Pan P, Tsung A. Protective role of cisplatin in ischemic liver injury through induction of autophagy. Autophagy 5: 1211‐2, 2009.
 117.Carloni S, Girelli S, Scopa C, Buonocore G, Longini M, Balduini W. Activation of autophagy and Akt/CREB signaling play an equivalent role in the neuroprotective effect of rapamycin in neonatal hypoxia‐ischemia. Autophagy 6: 366‐77, 2010.
 118.Carman CV, Springer TA. A transmigratory cup in leukocyte diapedesis both through individual vascular endothelial cells and between them. J Cell Biol 167: 377‐388, 2004.
 119.Carpi A, Menabò R, Kaludercic N, Pelicci P, Di Lisa F, Giorgio M. The cardioprotective effects elicited by p66(Shc) ablation demonstrate the crucial role of mitochondrial ROS formation in ischemia/reperfusion injury. Biochim Biophys Acta 1787: 774‐780, 2009.
 120.Carragher NO. Calpain inhibition: A therapeutic strategy targeting multiple disease states. Curr Pharm Des 12: 615‐638, 2006.
 121.Castello PR, David PS, McClure T, Crook Z, Poyton RO. Mitochondrial cytochrome oxidase produces nitic oxide under hypoxic conditions: Implications for oxygen sensing and hypoxic sensing in eukaryotes. Cell Metab 3: 277‐287, 2006.
 122.Cattaruzza F, Cenac N, Barocelli E, Impicciatore M, Rushbrook JI, Zhang M. Protective effect of proteinase‐activated receptor 2 activation on motility impairment and tissue damage induced by intestinal ischemia/reperfusion in rodents. Am J Pathol 169: 177‐188, 2013.
 123.Cerqueira NF, Hussni CA, Yoshida WB. Pathophysiology of mesenteric ischemia/reperfusion: A review. Acta Cir Bras 20: 336‐343, 2005.
 124.Chacko BK, Kramer PA, Ravi S, Benavides GA, Mitchell T, Dranka BP, Ferrick D, Singal AK, Ballinger SW, Bailey SM, Hard RW, Zhang J, Zhi D, Darley‐Usmar VM. The Bioenergetic Health Index: A new concept in mitochondrial translational research. Clin Sci (Lond) 127: 367‐373, 2014.
 125.Chaitanya GV, Steven AJ, Babu PP. Molecular PARP‐1 cleavage fragments: Signatures of cell‐death proteases in neurodegeneration. Cell Commun Signal 8: 31, 2010.
 126.Chamorro A, Meisel A, Planas AM, Urra X, van de Beek D, Veltkamp R. The immunology of acute stroke. Nat Rev Neurol 8: 401‐410, 2012.
 127.Chan MT, Boet R, Ng SC, Poon WS, Gin T. Effect of ischemic preconditioning on brain tissue gases and pH during temporary cerebral artery occlusion. Acta Neurochir Suppl 95: 93‐96, 2005.
 128.Charlagorla P, Liu J, Patel M, Rushbrook JI, Zhang M. Loss of plasma membrane integrity, complement response and formation of reactive oxygenspecies during early myocardial ischemia/reperfusion. Mol Immunol 56: 507‐512, 2013.
 129.Chatterjee PK, Brown PA, Cuzzocrea S, Zacharowski K, Stewart KN, Mota‐Filipe H, McDonald MC, Thiemermann C. Calpain inhibitor‐1 reduces renal ischemia/reperfusion injury in the rat. Kidney Int 59: 2073‐2083, 2001.
 130.Chatterjee PK, Todorovic Z, Sivarajah A, Mota‐Filipe H, Brown PA, Stewart KN, Mazzon E, Cuzzocrea S, Thiemermann C. Inhibitors of calpain activation (PD150606 and E‐64) and renal ischemia‐reperfusion injury. Biochem Pharmacol 69: 1121‐1131, 2005.
 131.Chavez‐Valdez R, Martin LJ, Flock DL, Northington FJ. Necrostatin‐1 attenuates mitochondrial dysfunction in neurons and astrocytes following neonatal hypoxia‐ischemia. Neuroscience 219: 192‐203, 2012.
 132.Chehal MK, Granville DJ. Cytochrome p450 2C (CYP2C) in ischemic heart injury and vascular dysfunction. Can J Physiol Pharmacol 84: 15‐20, 2006.
 133.Chen C, Feng Y, Zou L, Wang L, Chen HH, Cai JY, Xu JM, Sosnovik DE, Chao W. Role of extracellular RNA and TLR3‐Trif signaling in myocardial ischemia‐reperfusion injury. J Am Heart Assoc 3: e000683, 2014.
 134.Chen EP, Bittner HB, Davis RD, Folz RJ, Van Trigt P. Extracellular superoxide dismutase transgene overexpression preserves postischemic myocardial function in isolated murine hearts. Circulation 94: II412‐II417, 1996.
 135.Chen J, Crispín JC, Tedder TF, Dalle Lucca J, Tsokos GC. B cells contribute to ischemia/reperfusion‐mediated injury. J Autoimmunity 32: 195‐200, 2009.
 136.Chen JC, Wu ML, Huang KC, Lin WW. HMG‐CoA reductase inhibitors activate the unfolded protein response and induce cytoprotective GRP78 expression. Cardiovasc Res 80: 138‐150, 2008.
 137.Chen L, Hahn H, Wu G, Chen CH, Liron T, Schechtman D, Cavallaro G, Banci L, Guo Y, Bolli R, Dorn GW, II, Mochly‐Rosen D. Opposing cardioprotective actions and parallel hypertrophic effects of delta PKC and epsilon PKC. Proc Natl Acad Sci U S A 98: 11114‐11119, 2001.
 138.Chen L, Knowlton AA. Mitochondria and heart failure: New insights into an energetic problem. Minerva Cardioangiol 58: 213‐229, 2010.
 139.Chen M, Won DJ, Krajewski S, Gottlieb RA. Calpain and mitochondria in ischemia/reperfusion injury. J Biol Chem 277: 29181‐29186, 2002.
 140.Chen Y, Wood KJ. Interleukin‐23 and Th17 cells in transplantation immunity: Does 23+17 equal rejection? Transplant 84: 1071‐1074, 2007.
 141.Chironi GN, Boulanger CM, Simon A, Dignat‐George F, Freyssinet JM, Tedgui A. Endothelial microparticles in diseases. Cell Tissue Res 335: 143‐151, 2009.
 142.Cho YS, Challa S, Moquin D, Genga R, Ray TD, Guildford M, Chan FK. Phosphorylation‐driven assembly of the RIP1‐RIP3 complex regulates programmed necrosis and virus‐induced inflammation. Cell 137: 1112‐23, 2009.
 143.Choi DW. The role of glutamate neurotoxicity in hypoxic‐ischemic neuronal death. Annu Rev Neurosci 13: 171‐182, 1990.
 144.Chou HC, Cehn YW, Lee TR, Wu FS, Chan HT, Lyu PC, Timms JF, Chan HL. Proteomics study of oxidative stress and Src kinase inhibition in H9C2 cardiomyocytes: A cell model of heart ischemia‐reperfusion injury and treatment. Free Radic Biol Med 49: 96‐108, 2010.
 145.Chou WH, Choi DS, Zhang H, Mu D, McMahon T, Kharazia VN, Lowell CA, Ferriero DM, Messing RO. Neutrophil protein kinase Cdelta as a mediator of stroke‐reperfusion injury. J Clin Invest 114: 49‐56, 2004.
 146.Chouchani ET, Pell VR, Gaude E, Aksentijevic D, Sundier SY, Robb EL, Logan A, Nadtochiy SM, Ord EN, Smith AC, Eyassu F, Shirley R, Hu CH, Dare AJ, James AM, Rogatti S, Hartley RC, Eaton S, Costa AS, Brookes PS, Davidson SM, Duchen MR, Saeb‐Parsy K, Shattock MJ, Robinson AJ, Work LM, Frezza C, Krieg T, Murphy MP. Ischaemic accumulation of succinate controls reperfusion injury through mitochondrial ROS. Nature 515: 431‐435, 2014.
 147.Chouchani ET, Pell VR, James AM, Work LM, Saeb‐Parsy K, Frezza C, Krieg T, Murphy MP. A Unifying Mechanism for Mitochondrial Superoxide Production during Ischemia‐Reperfusion Injury. Cell Metab 23: 254‐263, 2016.
 148.Choudhuri S, Cui Y, Klaassen CD. Molecular targets of epigenetic regulation and effectors of environmental influences. Toxicol Appl Pharmacol 245: 378‐393, 2010.
 149.Churchill EN, Mochly‐Rosen D. The roles of PKCdelta and epsilon isoenzymes in the regulation of myocardial ischaemia/reperfusion injury. Biochem Soc Trans 35: 1040‐1042, 2007.
 150.Clarke SJ, McStay GP, Halestrap AP. Sanglifehrin A acts as a potent inhibitor of the mitochondrial permeability transition and reperfusion injury of the heart by binding to cyclophilin‐D at a different site from cyclosporin A. J Biol Chem 277: 34793‐34799, 2002.
 151.Colletti LM, Cortis A, Lukacs N, Kunkel SL, Green M, Strieter RM. Tumor necrosis factor up‐regulates intercellular adhesion molecule 1, which is important in the neutrophil‐dependent lung and liver injury associated with hepatic ischemia and reperfusion in the rat. Shock 10: 182‐191, 1998.
 152.Conrad M, Angeli JP, Vandenabeele P, Stockwell BR. Regulated necrosis: Disease relevance and therapeutic opportunities. Nat Rev Drug Discov 15: 348‐366, 2016.
 153.Contreras JL, Vilatoba M, Eckstein C, Bilbao G, Anthony Thompson J, Eckhoff DE. Caspase‐8 and caspase‐3 small interfering RNA decreases ischemia/reperfusion injury to the liver in mice. Surgery 136: 390‐400, 2004.
 154.Contreras L, Drago I, Zampese E, Pozzan T. Mitochondria: The calcium connection. Biochim Biophys Acta 1797: 607‐618, 2010.
 155.Coombes JS, Powers SK, Hamilton KL, Demirel HA, Shanely RA, Zergerolglu MA, Sen CK, Packer L, Ji LL. Improved cardiac performance after ischemia in aged rats supplemented with vitamin E and alpha‐lipoic acid. Am J Physiol 279: R2149‐R2155, 2000.
 156.Cooper D, Chitman KD, Williams MC, Granger DN. Time‐dependent platelet‐vessel wall interactions induced by ischemia‐reperfusion. Am J Physiol 284: G1027‐1033, 2003.
 157.Cooper D, Russell J, Chitman KD, Williams MC, Wolf RE, Granger DN. Leukocyte dependence of platelet adhesion in postcapillary venules. Am J Physiol 286: H1895‐H1900, 2004.
 158.Corbucci GG, Perrino C, Donato G, Ricchi A, Lettieri B, Troncone G, Indolfi C, Chiariello M, Avvedimento EV. Transient and reversible deoxyribonucleic acid damage in human left ventricle under controlled ischemia and reperfusion. J Am Coll Cardiol 43: 1992‐1999, 2004.
 159.Cording J, Gunther R, Vigolo E, Tscheik C, Winkler L, Schlattner I, Lorenz D, Haseloff RF, Schmidt‐Ott KM, Wolburg H, Blasig IE. Redox regulation of cell contacts by tricellulin and occludin: Redox‐sensitive cysteine sites in tricellulin regulate both tri‐ and bicellular junctions in tissue barriers as shown in hypoxia and ischemia. Antioxid Redox Signal 23: 1035‐1049, 2015.
 160.Costa FF. Non‐coding RNAs: Meet thy masters. Bioessays 32: 599‐608, 2010.
 161.Couchonnal L, Anderson ME. The role of calmodulin kinase II in myocardial physiology and disease. Physiology 23: 151‐159, 2008.
 162.Courties G, Moskowitz MA, Nahrendorf M. The innate immune system after ischemic injury: Lessons to be learned from the heart and brain. JAMA Neurol 71: 233‐236, 2014.
 163.Creemers EE, Tijsen AJ, Pinto YM. Circulating MicroRNAs: Novel biomarkers and extracellular communicators in cardiovascular disease? Circ Res 110: 483‐495, 2011.
 164.Crissinger KD, Granger DN. Characterization of intestinal collateral blood flow in the developing piglet. Pediatr Res 24: 473‐476, 1988.
 165.Croall DE, Ersfeld K. The calpains: Modular designs and functional diversity. Genome Biol 8: 218, 2007.
 166.Crosson CE, Mani SK, Husain S, Alsarraf O, Menick DR. Inhibition of histone deacetylase protects the retina from ischemic injury. Invest Ophthalmol Vis Sci 51: 3639‐3645, 2010.
 167.Cui Z, Scruggs SB, Gilda JE, Ping P, Gomes AV. Regulation of cardiac proteasomes by ubiquitination, SUMOylation, and beyond. J Mol Cell Cardiol 71: 32‐42, 2014.
 168.Czermak BJ, Sarma V, Pierson CL, Warner RL, Huber‐Lang M, Bless NM, Schmal H, Friedl HP, Ward PA. Protective effects of C5a blockade in sepsis. Nat Med 5: 788‐792, 1999.
 169.D'Autréaux B, Toledano MB. ROS as signaling molecules: Mechanisms that generate specificity in ROS homeostasis. Nat Mol Cell Biol 8: 813‐824, 2007.
 170.Daemen MA, van't Veer C, Denecker G, Heemskerk VH, Wolfs TG, Clauss M, Vandenabeele P, Buurman WA. Inhibition of apoptosis induced by ischemia‐reperfusion prevents inflammation. J Clin Invest 104: 541‐9, 1999.
 171.Dai H, Korthuis RJ. Mast cell proteases and inflammation. Drug Discov Today Dis Models 8: 47‐55, 2011.
 172.Damle SS, Moore EE, Babu AN, Meng X, Fullerton DA, Banerjee A. Hemoglobin‐based oxygen carrier induces heme oxygenase‐1 in the heart and lung but not brain. J Am Coll Surg 208: 592‐598, 2009.
 173.Dan Dunn J, Alvarez LA, Zhang X, Soldati T. Reactive oxygen species and mitochondria: A nexus of cellular homeostasis. Redox Biol 6: 472‐485, 2015.
 174.Das M, Cui J, Das DK. Generation of survival signal by differential interaction of p38MAPKalpha and p38MAPKbeta with caveolin‐1 and caveolin‐3 in the adapted heart. J Mol Cell Cardiol 42: 206‐213, 2007.
 175.De Chiara G, Marcocci ME, Torcia M, Lucibello M, Rosini P, Bonini P, Higashimoto Y, Damonte G, Armirotti A, Amodei S, Palamara AT, Russo T, Garaci E, Cozzolino F. Bcl‐2 Phosphorylation by p38 MAPK: Identification of target sites and biologic consequences. J Biol Chem 281: 21353‐21361, 2006.
 176.Degterev A, Huang Z, Boyce M, Li Y, Jagtap P, Mizushima N, Cuny GD, Mitchison TJ, Moskowitz MA, Yuan J. Chemical inhibitor of nonapoptotic cell death with therapeutic potential for ischemic brain injury. Nat Chem Biol 1: 112‐119, 2005.
 177.Degterev A, Linkermann A. Generation of small molecules to interfere with regulated necrosis. Cell Mol Life Sci 73: 2251‐2267, 2016.
 178.Deitch EA. Gut lymph and lymphatics: A source of factors leading to organ injury and dysfunction. Ann N Y Acad Sci 1207(Suppl 1): E103‐111, 2010.
 179.Deitch EA. Gut‐origin sepsis: Evolution of a concept. Surgeon 10: 350‐356, 2012.
 180.Deitch EA, Xu D, Kaise VL. Role for the gut in the development of injury‐ and shock‐induced SIRS and MODS: The gut‐lymph hypothesis, a review. Front Biosci 11: 520‐528, 2006.
 181.Deng Y, Theken KN, Lee CR. Cytochrome P450 epoxygeneases, soluble hydrolase, and the regulation of cardiovascular inflammation. J Mol Cell Cardiol 48: 331‐341, 2010.
 182.DeLeon ER, Gao Y, Huang E, Arif M, Arora N, Divietro A, Patel S, Olson KR. A case of mistaken identity: Are reactive oxygen species actually reactive sulfide species? Am J Physiol Regul Integr Comp Physiol: doi: 10.1152/ajpregu.00455.2015. [Epub ahead of print], 2016.
 183.de Moura EG, Lisboa PC, Passos MC. Neonatal programming of neuroimmunomodulation—role of adipocytokines and neuropeptides. Neuroimmunomodulation 15: 176‐188, 2008.
 184.De Pascali F, Hemann C, Samons K, Chen CA, Zweier JL. Hypoxia and reoxygenation induce endothelial nitric oxide synthase uncoupling in endothelial cells through tetrahydrobiopterin depletion and s‐glutathionlyation. Biochemistry 53: 3679‐3688, 2014.
 185.Depre C, Park JY, Shen YT, Zhao X, Qiu H, Yan L, Tian B, Vatner SF, Vatner DE. Molecular mechanisms mediating preconditioning following chronic ischemia differ from those in classical second window. Am J Physiol Heart Circ Physiol 299: H752‐H762, 2010.
 186.Depre C, Vatner SF. Cardioprotection in stunned and hibernating myocardium. Heart Fail Rev 12: 307‐317, 2007.
 187.Depre C, Vatner SF. Mechanisms of cell survival in myocardial hibernation. Trends Cardiovasc Med 15: 101‐110, 2005.
 188.Devalaraja‐Narashimha K, Diener AM, Padanilam BJ. Cyclophilin D gene ablation protects mice from ischemic renal injury. Am J Physiol Renal Physiol 297: F749‐F759, 2009.
 189.Devaux Y, Mueller M, Haaf P, Goretti E, Twerenbold R, Zangrando J, Vausort M, Reichlin T, Wildi K, Moehring B, Wagner DR, Mueller C. Diagnostic and prognostic value of circulating microRNAs in patients with acute chest pain. J Intern Med 277: 260‐271, 2015.
 190.Devaux Y, Stammet P, Friberg, H, Hassager C, Kuiper MA, Wise MP, Nielsen N. MicroRNAs: New biomarkers and therapeutic targets after cardiac arrest? Crit Care 19: 54, 2015.
 191.Devey L, Ferenbach D, Mohr E, Sangster K, Bellamy CO, Hughes J, Wigmore SJ. Tissue‐resistant macrophages protect the liver from ischemia reperfusion injury via a heme oxygenase‐1‐dependent mechanism. Mol Ther 17: 65‐72, 2009.
 192.DeWood MA, Spores J, Notske R, Mouser LT, Burroughs R, Golden MS, Lang HT. Prevalence of total coronary occlusion during the early hours of transmural myocardial infarction. N Engl J Med 303: 897‐902, 1980.
 193.Di Lisa F, Canton M, Menabò R, Kaludercic N, Bernardi P. Mitochondria and cardioprotection. Heart Fail Rev 12: 249‐60, 2007.
 194.Di Lisa F, Giorgio M, Ferdinandy P, Schulz R. New aspects of p66Shc in ischemia reperfusion injury and cardiovascular diseases. Br J Pharmacol doi: 10.1111/bph.13478. [Epub ahead of print] 2016.
 195.Di Lisa F, Kaludercic N, Carpi A, Menabo R, Giorgio M. Mitochondria and vascular pathology. Pharmacol Rep 61: 123‐130, 2009.
 196.Di Lisa F, Kaludercic N, Carpi A, Menabò R, Giorgio M. Mitochondrial pathways for ROS formation and myocardial injury: The relevance of p66(Shc) and monoamine oxidase. Basic Res Cardiol 104: 131‐139, 2009.
 197.Di Paola M, Lorusso M. Interaction of free fatty acids with mitochondria: Coupling, uncoupling and permeability transition. Biochim Biophys Acta 1757: 1330‐1337, 2006.
 198.Dinagl U, Iadecola C, Moskowitz MA. Pathobiology of ischemic stroke: An integrated view. Trends Neurosci 22: 391‐397, 1999.
 199.Divald A, Kivity S, Wang P, Hochhauser E, Roberts B, Teichberg S, Gomes AV, Powell SR. Myocardial ischemic preconditioning preserves postischemic function of the 26S proteasome through diminished oxidative damage to 19S regulatory particle subunits. Circ Res 106: 1829‐1838, 2010.
 200.Diwan A, Krenz M, Syed FM, Wansapura J, Ren X, Koesters AG, Li H, Kirshenbaum LA, Hahn HS, Robbins J, Jones WK, Dorn GW. Inhibition of ischemic cardiomyocytes apoptosis through targeted ablation of Bnip3 restrains postinfarction remodeling in mice. J Clin Invest 117: 2825‐2833, 2007.
 201.Dodd‐o JM, Hristopoulos ML, Welsh‐Servinsky LE, Tankersley CG, Pearse DB. Strain‐specific differences in sensitivity to ischemia‐reperfusion lung injury in mice. J Appl Physiol 100: 1590‐1595, 2006.
 202.Donovan N, Becker EB, Konishi Y, Bonni A. JNK phosphorylation and activation of BAD couples the stress‐activated signaling pathway to the cell death machinery. J Biol Chem 277: 40944‐40949, 2002.
 203.Drews O, Wildgruber R, Zong C, Sukop U, Nissum M, Weber G, Gomes AV, Ping P. Mammalian proteasome subpopulations with distinct molecular compositions and proteolytic activities. Mol Cell Proteomics 6: 2021‐2031, 2007.
 204.Dreyer WJ, Michael LH, Nguyen T, Smith CW, Anderson DC, Entman ML, Rossen RD. Kinetics of C5a release in cardiac lymph of dogs experiencing coronary artery ischemia‐reperfusion injury. Circ Res 71: 1518‐1524, 1992.
 205.Duan J, Kasper DL. Oxidative depolymerization of polysaccharides by reactive oxygen/nitrogen species. Glycobiology 21: 401‐409, 2011.
 206.Duffield JS, Forbes SJ, Constantinou CM, Clay S, Partolina M, Vuthoori S, Wu SJ, Wang R, Iredale JP. Selective depletion of macrophages reveals distinct, opposing roles during liver injury and repair. J Clin Invest 115: 56‐65, 2005.
 207.Duquesnes N, Lezoualc'h F, Crozatier B. PKC‐delta and PKC‐epsilon: Foes of the same family or strangers? J Mol Cell Cardiol 51: 665‐673, 2011.
 208.Dutta P, Nahrendorf M. Monocytes in myocardial infarction. Arterioscler Thromb Vasc Biol 35: 1066‐1070, 2015.
 209.Dvorak AM. Basophils and mast cells: Piecemeal degranulation in situ and ex vivo: A possible mechanism for cytokine‐induced function in disease. Immunol Ser 57: 169‐271, 1992.
 210.Dvoriantchikova G, Degterev A, Ivanov D. Retinal ganglion cell (RGC) programmed necrosis contributes to ischemia‐reperfusion‐induced retinal damage. Exp Eye Res 123: 1‐7, 2014.
 211.Dworakowski R, Alom‐Ruiz SP, Shah AM. NADPH oxidase‐derived reactive oxygen species in the regulation of endothelial phenotype. Pharmacol Rep 60: 21‐28, 2008.
 212.Edgerton C, Crispin JC, Moratz CM, Bettelli E, Oukka M, Simovic M, Zacharia A, Egan R, Chen J, Dalle Lucca JJ, Juang YT, Tsokos GC. IL‐17 producing CD4+ T cells mediate accelerated ischemia/reperfusion‐induced injury in autoimmunity‐prone mice. Clin Immunol 130: 313‐321, 2009.
 213.Edin ML, Wang Z, Bradbury JA, Graves JP, Lih FH, DeGraff LM, Foley JF, Torphy R, Ronnekleiv OK, Tomer KB, Lee CR, Zeldin DC. Endothelial expression of human cytochrome P450 epoxygenase CYP2C8 increases susceptibility to ischemia‐reperfusion injury in isolated mouse heart. FASEB J 25: 3436‐3447, 2011.
 214.Elmes MJ, Gardner DS, Langley‐Evans SC. Fetal exposure to a maternal low‐protein diet is associated with altered left ventricular pressure response to ischaemia‐reperfusion injury. Br J Nutr 98: 93‐100, 2007.
 215.Elmes MJ, McMullen S, Gardner DS, Langley‐Evans SC. Prenatal diet determines susceptibility to cardiac ischaemia‐reperfusion injury following treatment with diethylmaleic acid and N‐acetylcysteine. Life Sci 82: 149‐155, 2008.
 216.Ellett JD, Atkinson C, Evans ZP, Amani Z, Balish E, Schmid MG, von Rooijen N, Schnellmann RG, Chavin KD. Murine Kupffer cells are protective in toal hepatic ischemia/reperfusion injury with bowel congestion through IL‐10. J Immunol 184: 5849‐5858, 2010.
 217.Elrod JW, Duranski MR, Langston W, Greer JJ, Tao L, Dugas TR, Kevil CG, Champion HC, Lefer DJ. eNOS gene therapy exacerbates hepatic ischemia‐reperfusion injury in diabetes: A role for eNOS uncoupling. Circ Res 99: 78‐85, 2006.
 218.Elvington A, Atkinson C, Zhu H, Yu J, Takahashi K, Stahl GL, Kindy MS, Tomlinson S. The alternative complement pathway propagates inflammation and injury in murine ischemic stroke. J Immunol 189: 4640‐4647, 2012.
 219.Elzey BD, Tian J, Jensen RJ, Swanson AK, Lees JR, Lentz SR, Stein CS, Neiswandt B, Wang Y, Davidson BL, Ratliff TL. Platelet‐mediated modulation of adaptive immunity. A communication link between innate and adaptive immune compartments. Immunity 19: 9‐19, 2003.
 220.Endres M, Ahmadi M, Kruman I, Biniszkiewicz D, Meisel A, Gertz K. Folate deficiency increases postischemic brain injury. Stroke 36: 321‐325, 2005.
 221.Endres M, Fan G, Meisel A, Dirnagl U, Jaenisch R. Effects of cerebral ischemia in mice lacking DNA methyltransferase 1 in post‐mitotic neurons. Neuroreport 12: 3763‐6, 2001.
 222.Endres M, Meisel A, Biniszkiewicz D, Namura S, Prass K, Ruscher K, Lipski A, Jaenisch R, Moskowitz MA, Dirnagl U. DNA methyltransferase contributes to delayed ischemic brain injury. J Neurosci 20: 3175‐3181, 2000.
 223.Engler RL, Schmid‐Schonbein GW, Pavelec RS. Leukocyte capillary plugging in myocardial ischemia and reperfusion in the dog. Am J Pathol 111: 98‐111, 1983.
 224.Entman ML, Youker K, Shoji T, Kukielka G, Shappell SB, Taylor AA, Smith CW. Neutrophil induced oxidative injury of cardiac myocytes. A compartmented system requiring CD11b/CD18‐ICAM‐1 adherence. J Clin Invest 90: 1335‐1345, 1992.
 225.Erickson JR, Joiner MA, Guan X, Kurtschke W, Yang J, Oddis CV, Bartlett RK, Lowe JS, O'Donnell SE, Aykin‐Burns N, Zimmerman MC, Zimmerman K, Ham A‐JL, Weiss RM, Spitz DR, Shea MA, Colbran RJ, Mohler PJ, Anderson ME. A dynamic pathway for calcium‐independent activation of CaMKII by methionine oxidation. Cell 133: 462‐474, 2008.
 226.Erusalimsky JD, Moncada S. Nitric oxide and mitochondrial signaling: From physiology to pathophysiology. Arteroscler Thromb Vasc Biol 27: 2524‐2531, 2007.
 227.Esch JS, Jurk K, Knoefel WT, Roeder G, Voss H, Tustas RY, Schmelzle M, Krieg A, Eisenberger CF, Topp S, Rogiers X, Fischer L, Aken HV, Kehrel BE. Platelet activation and increased tissue factor expression on monocytes in reperfusion injury following orthotopic liver transplantation. Platelets 21: 348‐359, 2010.
 228.Esme H, Fidan H, Koken T, Solak O. Effect of lung ischemia‐reperfusion on oxidative stress parameters of remote tissues. Eur J CardioCardio‐Thoracic Surg 29: 294‐298, 2006.
 229.Ettinger G, MacDonald K, Reid G, Burton JP. The influence of the human microbiome and probiotics on cardiovascular Health. Gut Microbes 5: 719‐728, 2014.
 230.Fagundes CT, Amaral FA, Teixeira AL, Souza DG, Teixeira MM. Adapting to environmental stresses: The role of the microbiota in controlling innate immunity and behavioral responses. Immunol Rev 245: 250‐264, 2012.
 231.Faith JJ, Guruge JL, Charbonneau M, Subramanian S, Seedorf H, Goodman AL, Clemente JC, Knight R, Heath AC, Leibel RL, Rosenbaum M, Gordon JI. The long‐term stability of the human gut microbiota. Science 341: 1237439, 2013.
 232.Fan M, Du L, Stone AA, Gilbert KM, Chambers TC. Modulation of mitogen‐activated protein kinases and phosphorylation of Bcl‐2 by vinblastine represent persistent forms of normal fluctuations at G2‐M1. Cancer Res 60: 6403‐6407, 2000.
 233.Fang J, Song XW, Tian J, Chen HY, Li DF, Wang JF, Ren AJ, Yuan WJ, Lin L. Overexpression of microRNA‐378 attenuates ischemia‐induced apoptosis by inhibiting caspase‐3 expression in cardiac myocytes. Apoptosis 17: 410‐423, 2012.
 234.Faraco G, Pancani T, Formentini L, Mascagni P, Fossati G, Leoni F, Moroni F, Chiarugi A. Pharmacological inhibition of histone deacetylases by suberoylanilide hydroxamic acid specifically alters gene expression and reduces ischemic injury in the mouse brain. Mol Pharmacol 70: 1876‐1884, 2006.
 235.Fasanaro P, Greco S, Ivan M, Capogrossi MC, Martelli F. microRNA: Emerging therapeutic targets in acute ischemic diseases. Pharmacol Ther 125: 92‐104, 2010.
 236.Fatokun AA, Dawson VL, Dawson TM. Parthanatos: Mitochondrial‐linked mechanisms and therapeutic opportunities. Br J Pharmacol 171: 2000‐2016, 2014.
 237.Fauman EB, Saper MA. Structure and function of the protein tyrosine phosphatases. Trends Biochem Sci 21: 413‐417, 1996.
 238.Fayaz SM, Suvanish Kumar VS, Davis CK, Rajanikant GK. Novel RIPK3 inhibitors discovered through a structure‐based approach exert post‐ischemic neuroprotection. Mol Divers 20: 719‐728, 2016.
 239.Fayaz SM, Suvanish Kumar VS, Rajanikant GK. Necroptosis: Who knew there were so many interesting ways to die? CNS Neurol Disord Drug Targets 13: 42‐51, 2014.
 240.Felger JC, Abe T, Kaunzner UW, Gottfried‐Blackmore A, Gal‐Toth J, McEwen BS, Iadecola C, Bulloch K. Brain dendritic cells in ischemic stroke: Time course, activation state, and origin. Brain Behav Immun 24: 724‐737, 2010.
 241.Feng M, Wang H, Wang Q, Guan W. Matrix metalloprotease 9 promotes liver recovery from ischemia and reperfusion injury. J Surg Res 180: 156‐161, 2013.
 242.Ferdinandy P, Schulz R, Baxter GF. Interaction of cardiovascular risk factors with myocardial ischemia/reperfusion injury, preconditioning, and postconditioning. Pharmacol Rev 59: 418‐458, 2007.
 243.Fernandez‐Klett F, Offenhauser N, Dirnagl U, Priller J, Lindauer U. Pericytes in capillaries are contractile in vivo, but arterioles mediate functional hyperemia in the mouse brain. Proc Natl Acad Sci USA 107: 22290‐22295, 2010.
 244.Ferran C, Millan MT, Csizmadia V, Cooper JT, Brostjan C, Bach FH, Winkler H. Inhibition of NF‐kappa B by pyrrolidine dithiocarbamate blocks endothelial cell activation. Biochem Biophys Res Comm 214: 212‐223, 1995.
 245.Ferraro FJ, Rush BF, Jr., Simonian GT, Bruce CJ, Murphy TF, Hsieh JT, Klein K, Condon M. A comparison of survival at different degrees of hemorrhagic shock in germ‐free and germ‐bearing rats. Shock 4: 117‐120, 1995.
 246.Festjens N, Vanden Berghe T, Cornelis S, Vandenabeele P. RIP1, a kinase on the crossroads of a cell's decision to live or die. Cell Death Differ 14: 400‐410, 2007.
 247.Finsterbusch M, Voisin MB, Beyrau M, Williams TJ, Nourshargh S. Neutrophils recruited by chemoattractants in vivo induce microvascular plasma protein leakage through secretion of TNF. J Exp Med 211: 1307‐1314, 2014.
 248.Fleming I, Michaelis UR, Bredenkötter D, Fisslthaler B, Dehghani F, Brandes RP, Busse R. Endothelium‐derived hyperpolarizing factor synthase (cytochrome P450 2C9) is a functionally significant source of reactive oxygen species in coronary arteries. Circ Res 88: 44‐51, 2001.
 249.Folino A, Losano G, Rastaldo R. Balance of nitric oxide and reactive oxygen species in myocardial reperfusion injury and protection. J Cardiovasc Pharmacol 62: 567‐575, 2013.
 250.Ford DA. Lipid oxidation by hypochlorous acid: Chlorinated lipids in atherosclerosis and myocardial ischemia. Clin Lipidol 5: 835‐852, 2010.
 251.Forman HJ, Fukoto JM, Miller T, Zhang H, Rinna A, Levy S. The chemistry of cell signaling by reactive oxygen and nitrogen species and 4‐hydroxynonenal. Arch Biochem Biophys 477: 183‐195, 2008.
 252.Forman HJ, Torre M, Fukoto J. Redox signaling. Mol Cell Biochem 234: 49‐62, 2002.
 253.Foster MW, Hess DT, Stamler JS. Protein S‐nitrosylation in health and disease: A current perspective. Trends Mol Med 15: 391‐404, 2009.
 254.Frangogiannis NG. Inflammation in cardiac injury, repair and regeneration. Curr Opin Cardiol 30: 240‐245, 2015.
 255.Frangogiannis NG. The immune system and cardiac repair. Pharmacol Res 58: 88‐111, 2008.
 256.Friedmann Angeli JP, Schneider M, Proneth B, Tyurina YY, Tyurin VA, Hammond VJ, Herbach N, Aichler M, Walch A, Eggenhofer E, Basavarajappa D, Radmark O, Kobayashi S, Seibt T, Beck H, Neff F, Esposito I, Wanke R, Forster H, Yefremova O, Heinrichmeyer M, Bornkamm GW, Geissler EK, Thomas SB, Stockwell BR, O'Donnell VB, Kagan VE, Schick JA, Conrad M. Inactivation of the ferroptosis regulator Gpx4 triggers acute renal failure in mice. Nat Cell Biol 16: 1180‐1191, 2014.
 257.Fritzinger DC, Hew BE, Lee JQ, Newhouse J, Alam M, Ciallella JR, Bowers M, Gorsuch WB, Guikema BJ, Stahl GL, Vogel CW. Derivatives of human complement component C3 for therapeutic complement depletion: A novel class of therapeutic agents. Adv Exp Med Biol 632: 293‐307, 2008.
 258.Frost RJ, van Rooij E. miRNAs as therapeutic targets in ischemic heart disease. J Cardiovasc Transl Res 3: 280‐289, 2010.
 259.Fulda S. Regulation of necroptosis signaling and cell death by reactive oxygen species. Biol Chem 397: 657‐660, 2016.
 260.Gaboury JP, Johnston B, Niu X, Kubes P. Mechanisms nderlying acute mast cell‐induced leukocyte rolling and adhesion in vivo. J Immunol 154: 804‐813, 1995.
 261.Galinanes M, Hearse DJ. Species differences in susceptibility to ischemic injury and responsiveness to myocardial protection. Cardioscience 1: 127‐143, 1990.
 262.Galli SJ, Gordon JR, Wershil BK. Cytokine production by mast cells and basophils. Curr Opin Immunol 3: 865‐872, 1991.
 263.Galli SJ, Kalesnikoff J, Grimbaldeston A, Piliponsky A, Williams C, Tsai M. Mast cellsare “tunable' effector and immunoregulatory cells: Recent advances. Annu Rev Immunol 23: 749‐786, 2005.
 264.Galluzzi L, Kepp O, Krautwald S, Kroemer G, Linkermann A. Molecular mechanisms of regulated necrosis. Semin Cell Dev Biol 35: 24‐32, 2014.
 265.Gan XT, Ettinger G, Huang CX, Burton JP, Haist JV, Rajapurohitam V, Sidaway JE, Martin G, Gloor GB, Swann JR, Reid G, Karmazyn M. Probiotic administration attenuates myocardial hypertrophy and heart failure after myocardial infarction in the rat. Circ Heart Fail 7: 491‐499, 2014.
 266.Gandalfo MT, Jang HR, Bagnasco SM, Ko GJ, Agreda P, Satpute SR, Crow MT, King LS, Rabb H. Foxp3 +regulatory T cells participate in repair of ischemic acute kidney injury. Kidney Int 76: 717‐729, 2009.
 267.Gao X, Zhang H, Belmadani S, Wu J, Xu X, Elford H, Potter BJ, Zhang C. Role of TNF‐alpha‐induced reactive oxygen species in endothelial dysfunction during reperfusion injury. Am J Physiol Heart Circ Physiol 295: H2242‐2249, 2008.
 268.Gawaz M. Role of platelets in coronary thrombosis and reperfusion of ischemic myocardium. Cardiovasc Res 61: 498‐511, 2004.
 269.Ge L, Zhou X, Ji WJ, Lu RY, Zhang Y, Zhang YD, Ma YQ, Zhao JH, Li YM. Neutrophil extracellular traps in ischemia‐reperfusion injury‐induced myocardial no‐reflow: Therapeutic potential of DNase‐based reperfusion strategy. Am J Physiol Heart Circ Physiol 308: H500‐H509, 2015.
 270.Genken E, Buis CI, Visser DS, Blokzijl H, Moshage H, Nemes B, Leuvenink HG, de Jong, KP, Peeters PM, Slooff MJ, Porte RJ. Expression of heme oxygenase‐1 in human livers before transplantation correlates with graft injury and function after transplantation. Am J Transplant 5: 1875‐1885, 2005.
 271.George FD. Microparticles in vascular diseases. Thromb Res 122: 555‐559, 2008.
 272.Gerszten RE, Asnani A, Carr SA. Status and prospects for discovery and verification of new biomarkers of cardiovascular disease by proteomics. Circ Res 109: 463‐474, 2011.
 273.Giakoustidis DE, Iliadis S, Tsantilas D, Papageorgiou G, Kontos N, Kostopoulou E, Botsoglou NA, Gerasimidis T, Dimitriadou A. Blockade of Kupffer cells by gadolinium chloride reduces lipid peroxidation and protects liver from ischemia/reperfusion injury. Hepatogastroenterology 50: 1587‐1592, 2003.
 274.Giussani DA, Camm EJ, Niu Y, Richter HG, Blanco CE, Gottschalk R, Blake EZ, Horder KA, Thakor AS, Hansell JA, Kane AD, Wooding FB, Cross CM, Herrera EA. Developmental programming of cardiovascular dysfunction by prenatal hypoxia and oxidative stress. PLoS One 7: e31017, 2012.
 275.Giussani DA, Davidge ST. Developmental programming of cardiovascular disease by prenatal hypoxia. J Develop Origins Health Dis 4: 328‐337, 2013
 276.Giedt RJ, Yang C, Zweier JL, Matzavinos A, Alevriadou BR. Mitochondrial fission in endothelial cells after simulated ischemia/reperfusion: Role of nitric oxide and reactive oxygen species. Free Radic Biol Med 52: 348‐356, 2012.
 277.Glancy B, Hartnell LM, Malide D, Yu ZX, Combs CA, Connelly PS, Subramaniam S, Balaban RS. Mitochondrial reticulum for cellular energy distribution in muscle. Nature 523: 617‐620, 2015.
 278.Gnecchi M, Zhang Z, Ni A, Dzau VJ. Paracrine mechanisms in adult stem cell signaling and therapy. Circ Res 103: 1204‐1219, 2008.
 279.Go YM, Jones DP. Intracellular proatherogenic events and cell adhesion modulated by extracellular thiol/disulfide redox state. Circulation 111: 2973‐2980, 2005.
 280.Go YM, Jones DP. Redox compartmentalization in eukaryotic cells. Biochim Biophys Acta 1780: 1273‐1290, 2008.
 281.Go YM, Park H, Koval M, Orr M, Reed M, Liang Y, Smith D, Pohl J, Jones DP. A key role for mitochondria in endothelial signaling by plasma cysteine/cystine redox potential. Free Radic Biol Med 48: 275‐283, 2010.
 282.Goda N, Suzuki K, Naito M, Takeoka S, Tsuchida E, Ishimura Y, Tamatani T, Suematsu M. Distribution of heme oxygenase isoforms in rat liver. Topographic basis for carbon monoxide‐mediated microvascular relaxation. J Clin Invest 101: 604‐612, 1998.
 283.Godoy LC, Moretti AI, Jurado MC, Oxer D, Janiszewski M, Ckless K, Velasco IT, Laurindo FR, Souza HP. Loss of CD40 endogenous S‐nitrosylation during inflammatory response in endotoxemic mice and patients with sepsis. Shock 33: 626‐633, 2010.
 284.Godwin JG, Ge X, Stephan K, Jurisch A, Tullius SG, Iacomini J. Identification of a microRNA signature of renal ischemia reperfusion injury. Proc Natl Acad Sci U S A 107: 14339‐14344, 2010.
 285.Goldman G, Welbourn R, Klausne JM, Kobzik L, Valeri CR, Shepro D, Hechtman HB. Mast cells and leukotrienes mediate neutrophil sequestration and lung edema after remote ischemia in rodents. Surgery 12: 578‐586, 1992.
 286.Golwala NH, Hodenette C, Murthy SN, Nossaman BD, Kadowitz PJ. Vascular responses to nitrite are mediated by xanthine oxidoreductase and mitochondrial aldehyde dehydrogenase in the rat. Can J Physiol Pharmacol 87: 1095‐1101, 2009.
 287.Gonzalez LM, Moeser AJ, Blikslager AT. Animal models of ischemia‐reperfusion‐induced intestinal injury: Progress and promise for translational research. Am J Physiol Gastrointest Liver Physiol 308: G63‐75, 2015.
 288.Goretti E, Wagner DR, Devaux Y. miRNAs as biomarkers of myocardial infarction: A step forward towards personalized medicine? Trends Mol Med 20: 716‐725, 2014.
 289.Gorsuch WB, Chrysanthou E, Schwaeble WJ, Stahl GL. The complement system in ischemia‐reperfusion injuries. Immunobiology 217: 1026‐1033, 2012.
 290.Gottlieb RA. Cytochrome P450: Major player in reperfusion injury. Arch Biochem Biophys 420: 262‐267, 2003.
 291.Gottlieb RA, Gustafsson AB. Mitochondrial turnover in the heart. Biochim Biophys Acta 1813: 1295‐1301, 2011.
 292.Gottlieb RA, Mentzer RM. Autophagy during cardiac stress: Joys and frustrations of autophagy. Annu Rev Physiol 72: 45‐59, 2010.
 293.Gould TJ, Vu TT, Swystun LL, Dwivedi DJ, Mai SH, Weitz JI, Liaw PC. Neutrophil extracellular traps promote thrombin generation through platelet‐dependent and platelet‐independent mechanisms. Arterioscler Thromb Vasc Biol 34: 1977‐1984, 2014.
 294.Gourdin MJ, Bree B, De Kock M. The impact of ischaemia‐reperfusion on the blood vessel. Eur J Anaesthesiol 26: 537‐547, 2009.
 295.Gracanin M, Lam MA, Morgan PE, Rodgers KJ, Hawkins CL, Davies MJ. Amino acid, peptide, and protein hydroperoxides and their decomposition products modify the activity of the 26S proteasome. Free Radic Biol Med 50: 389‐399, 2011.
 296.Granger A, Abdullah I, Huebner F, Stout A, Wang T, Huebner T, Epstein JA, Gruber PJ. Histone deacetylase inhibition reduces myocardial ischemia‐reperfusion injury in mice. FASEB J 22: 3549‐3560, 2008.
 297.Granger DN. Role of xanthine oxidase and granulocytes in ischemia‐reperfusion injury. Am J Physiol 255: H1269‐H1275, 1988.
 298.Granger DN. Ischemia‐reperfusion: Mechanisms of microvascular dysfunction and the influence of risk factors for cardiovascular disease. Microcirculation 6: 167‐178, 1999.
 299.Granger DN, Korthuis RJ. Physiologic mechanisms of postischemic tissue injury. Ann Rev Physiol 57: 311‐332, 1996.
 300.Granger DN, Kvietys PR. Reperfusion injury and reactive oxygen species: The evolution of a concept. Redox Biol 6: 524‐551, 2015.
 301.Granger DN, Rutili G, McCord JM. Superoxide radicals in feline intestinal ischemia. Gastroenterology 81: 22‐29, 1981.
 302.Granger DN, Stokes KY, Shigematsu T, Cerwinka WH, Tailor A, Krieglstein CF. Splanchnic ischaemia‐reperfusion injury: Mechanistic insights provided by mutant mice. Acta Physiol Scand 173: 83‐91, 2001.
 303.Granville DJ, Tashakkor B, Takeuchi C, Gustafsson ÅB, Sayen MR, Wentworth P, Jr., Yeager M, Gottlieb RA. Reduction of ischemia and reperfusion‐induced myocardial damage by cytochrome P450 inhibitors. Proc Natl Acad Sci U S A 101: 1321‐1326, 2004.
 304.Greco S, De Simone M, Colussi C, Zaccagnini G, Fasanaro P, Pescatori M, Cardani R, Perbellini R, Isaia E, Sale P, Meola G, Capogrossi MC, Gaetano C, Martelli F. Common micro‐RNA signature in skeletal muscle damage and regeneration induced by Duchenne muscular dystrophy and acute ischemia. FASEB J 23: 3335‐3346, 2009.
 305.Gregory JC, Buffa JA, Org E, Wang Z, Levison BS, Zhu W, Wagner MA, Bennett BJ, Li L, DiDonato JA, Lusis AJ, Hazen SL. Transmission of atherosclerosis susceptibility with gut microbial transplantation. J Biol Chem 290: 5647‐5660, 2015.
 306.Grimbaldeston MA, Nakae S, Kalesnikoff J, Tsai M, Galli S. Mast cell‐derived interleukin 10 limits skin pathology in contact dermatitis and chronic irradiation with ultraviolet B. Nature Immunol 8: 1095‐1104, 2007.
 307.Grisham MB, Jourd'Heuil D, Wink DA. Nitric oxide. I. Physiological chemistry of nitric oxide and its metabolites: Implications in inflammation. Am J Physiol 276: G315‐G321, 1999.
 308.Grootjans J, Lenaerts K, Derikx JP, Matthijsen RA, de Bruine AP, van Bijnen AA, van Dam RM, Dejong CH, Buurman WA. Human intestinal ischemia‐reperfusion‐induced inflammation characterized: Experiences from a new translational model. Am J Pathol 176: 2283‐2291, 2010.
 309.Gross GJ, Falk JR, Gross ER, Isbell M, Moore J, Nithipatikom K. Cytochrome P450 and arachidonic acid metabolites: Role in myocardial ischemia/reperfusion injury revisited. Cardiovasc Res 68: 18‐25, 2005.
 310.Grover GJ, Atwal KS, Sleph PG, Wang FL, Monshizadegan H, Monticello T, Green DW. Excessive ATP hydrolysis in ischemic myocardium by mitochondrial F1F0‐ATPase: Effect of selective pharmacological inhibition of mitochondrial ATPase hydrolase activity. Am J Physiol Heart Circ Physiol 287: H1747‐H1755, 2004.
 311.Grueter CE, Abiria SA, Dzhura I, Wu Y, Ham AJ, Mohler PJ, Anderson ME, Colbran RJ. L‐type Ca2+ channel facilitation mediated by phosphorylation of the b subunit by CaMKII. Mol Cell 23: 641‐650, 2006.
 312.Guan QH, Pei DS, Zhang QG, Hao ZB, Xu TL, Zhang GY. The neuroprotective action of SP600125, a new inhibitor of JNK, on transient brain ischemia/reperfusion‐induced neuronal death in rat hippocampal CA1 via nuclear and non‐nuclear pathways. Brain Res 1035: 51‐59, 2005.
 313.Gundewar S, Calvert JW, Elrod JW, Lefer DJ. Cytoprotective effects of N,N,N‐trimethylsphingosine during ischemia‐ reperfusion injury are lost in the setting of obesity and diabetes. Am J Physiol Heart Circ Physiol 293: H2462‐H2471, 2007.
 314.Guo G, Bhat NR. p38a MAP kinase mediates hypoxia‐induced motor neuron cell death: A potential target of minocycline's neuroprotective action. Neurochem Res 32: 2160‐6, 2007.
 315.Guo Y, Flaherty MP, Wu WJ, Tan W, Zhu X, Li Q, Bolli R. Genetic background, gender, age, body temperature, and arterial blood pH have a major impact on myocardial infarct size in the mouse and need to be carefully measured and/or taken into account: Results of a comprehensive analysis of determinants of infarct size in 1,074 mice. Basic Res Cardiol 107: 288, 2012.
 316.Gurusamy N, Lekli I, Gherghiceanu M, Popescu LM, Das DK. BAG‐1 induces autophagy for cardiac cell survival. Autophagy 5: 120‐121, 2009.
 317.Gute DC, Ishida T, Yarimizu K, Korthuis RJ. Inflammatory responses to ischemia and reperfusion in skeletal muscle. Mol Cell Biochem 179: 169‐187, 1998.
 318.Gute D, Korthuis RJ. Role of leukocyte adherence in reperfusion‐induced microvascular dysfunction and tissue injury. In: Granger DN, Schmid‐Schönbein GW, editors. Leukocyte Adhesion. New York, NY: Oxford Uuniversity Press, 1995.
 319.Gysembergh A, Margonari H, Loufoua J, Ovize A, Andre‐Fouet X, Minaire Y, Ovize M. Stretch‐induced protection shares a common mechanism with ischemic preconditioning in rabbit heart. Am J Physiol 274: H955‐H964, 1998.
 320.Hacke W, Donnan G, Fieschi C, Kaste M, von Kummer R, Broderick JP, Brott T, Frankel M, Grotta JC, Haley EC, Jr., Kwiatkowski T, Levine SR, Lewandowski C, Lu M, Lyden P, Marler JR, Patel S, Tilley BC, Albers G, Bluhmki E, Wilhelm M, Hamilton S. Association of outcome with early stroke treatment: Pooled analysis of ATLANTIS, ECASS, and NINDS rt‐PA stroke trials. Lancet 363: 768‐74, 2004.
 321.Haheim LL, Olsen I, Ronningen KS. Oral infection, regular alcohol drinking pattern, and myocardial infarction. Med Hypotheses 79: 725‐730, 2012.
 322.Hales CN, Barker DJ. The thrifty phenotype hypothesis. Br Med Bull 60: 5‐20, 2001.
 323.Halestrap AP. A pore way to die: The role of mitochondria in reperfusion injury and cardioprotection. Biochem Soc Trans 38, 841‐860, 2010.
 324.Halestrap AP. What is the mitochondrial permeability transition pore? J Mol Cell Cardiol 46: 821‐31, 2009.
 325.Hall CN, Reynell C, Gesslein B, Hamilton NB, Mishra A, Sutherland BA, O'Farrell FM, Buchan AM, Lauritzen M, Attwell D. Capillary pericytes regulate cerebral blood flow in health and disease. Nature 508: 55‐60, 2014.
 326.Haller H, Kunzendorf U, Sacherer K, Lindschau C, Walz G, Distler A, Luft FC. T cell adhesion to P‐selectin induces tyrosine phosphorylation of pp125 focal adhesion kinase and other substrates. J Immunol 158: 1061‐1067, 1997.
 327.Hamacher‐Brady A, Brady NR, Logue SE, Sayen MR, Jinno M, Kirshenbaum LA, Gottlieb RA, Gustafsson AB. Response to myocardial ischemia/reperfusion injury involves Bnip3 and autophagy. Cell Death Differ 14: 146‐157, 2007.
 328.Hamilton NB, Attwell D, Hall CN. Pericyte‐mediated regulation of capillary diameter: A component of neurovascular coupling in health and disease. Front Neuroenergetics pii: 5, 2010.
 329.Hanidziar D, Koulmanda M. Inflammation and the balance of Treg and Th17 cells in transplant rejection and tolerance. Curr Opin Organ Transplant 15: 411‐415, 2010.
 330.Hanschen M, Zahler S, Krombach F, Khandoga A. Reciprocal activation between CD4+ T cells and Kupffer cells during hepatic ischemia‐reperfusion. Transplant 86: 710‐718, 2008.
 331.Hanson MA, Gluckman PD. Early developmental conditioning of later health and disease: Physiology or pathophysiology? Physiol Rev 94: 1027‐1076, 2014.
 332.Harding SJ, Browne GJ, Miller BW, Prigent SA, Dickens M. Activation of ASK1, downstream MAPKK and MAPK isoforms during cardiac ischaemia. Biochim Biophys Acta 1802: 733‐740, 2010.
 333.Hariharan N, Zhai P, Sadoshima J. Oxidative stress stimulates autophagic flux during ischemia/reperfusion. Antioxid Redox Signal 14: 2179‐2190, 2011.
 334.Hausenloy DJ, Yellon DM. Survival kinases in ischemic preconditioning and postconditioning. Cardiovasc Res 70: 240‐253, 2006.
 335.Hauton D. Hypoxia in early pregnancy induces cardiac dysfunction in adult offspring of Rattus norvegicus, a non‐hypoxia‐adapted species. Comp Biochem Physiol A Mol Integr Comp Physiol 163: 278‐285, 2012.
 336.Hauton D, Ousley V. Prenatal hypoxia induces increased cardiac contractility on a background of decreased capillary density. BMC Cardivasc Disord 9:1, 2009
 337.Hausenloy DJ, Yellon DM. Myocardial ischemia‐reperfusion injury: A neglected therapeutic target. J Clin Invest 123: 92‐100, 2013.
 338.Hausenloy DJ, Yellon DM, Mani‐Babu S, Duchen MR. Preconditioning protects by inhibiting the mitochondrial permeability transition. Am J Physiol Heart Circ Physiol 287: H841‐849, 2004.
 339.He B, Xiao J, Ren AJ, Zhang YF, Zhang H, Chen M, Xie B, Gao XG, Wang YW. Role of miR‐1 and miR‐133a in myocardial ischemic postconditioning. J Biomed Sci 18: 22, 2011.
 340.He C, Klionsky DJ. Regulation mechanisms and signaling pathways of autophagy. Annu Rev Genet 43: 67‐93, 2009.
 341.He GZ, Dong LG, Chen XF, Zhou KG, Shu H. Lymph duct ligation during ischemia/reperfusion prevents pulmonary dysfunction in a rat model with omega‐3 polyunsaturated fatty acid and glutamine. Nutrition 27: 604‐614, 2011.
 342.He S, Wang L, Miao L, Wang T, Du F, Zhao L, Wang X. Receptor interacting protein kinase‐3 determines cellular necrotic response to TNF‐alpha. Cell 137: 1100‐11, 2009.
 343.Hehner SP, Breitkreutz R, Shubinsky G, Unsoeld H, Shulze‐Osthoff K, Schmitz ML, Dröge W. Enhancement of T cell receptor signaling by a mild oxidative shift in the intracellular thiol pool. J Immunol 165: 4319‐4328, 2000.
 344.Hein TW, Zhang C, Wang W, Chang CI, Thengchaisri N, Kuo L. Ischemia‐reperfusion selectively impairs nitric oxide‐mediated dilation in coronary arterioles: Counteracting role of arginase. FASEB J 17: 2328‐2330, 2003.
 345.Hernandez LA, Grisham MB, Twohig B, Arfors KE, Harlan JM, Granger DN. Role of neutrophils in ischemia‐reperfusion‐induced microvascular injury. Am J Physiol 253: H699‐H703, 1987.
 346.Hernando V, Inserte J, Sartório CL, Parra VM, Poncelas‐Nozal M, Garcia‐Dorado D. Calpain translocation and activation as pharmacological targets during myocardial ischemia/reperfusion. J Mol Cell Cardiol 49: 271‐279, 2010.
 347.Hess DT, Stamler JS. Regulation by S‐nitrosylation of protein post‐translational modification. J Biol Chem 287: 4411‐4418, 2012.
 348.Hickey MJ, Kubes P. Role of nitric oxide in regulation of leucocyte‐endothelial cell interactions. Exp Physiol 82: 339‐348, 1997.
 349.Hill JW, Nemoto EM. Matrix‐derived inflammatory mediator N‐acetyl proline‐glycine‐proline is neurotoxic and upregulated in brain after ischemic stroke. J Neuroinflammation 12: 214, 2015.
 350.Hill JH, Ward PA. The phlogistic role of C3 leukotactic fragments in myocardial infarcts of rats. J Exp Med 133: 885‐900, 1971.
 351.Hirsch J, Niemann CU, Hansen KC, Choi SJN, Su X, Frank JA, Fang X, Hirose R, Theodore P, Sapru A, Burlingame AL, Matthay MA. Alterations in the proteome of pulmonary alveolar type II cells in the rat after hepatic ishchemia‐reperfusion. Crit Care Med 36: 1846‐1854, 2008.
 352.Hochhauser E, Cheporko Y, Yasovich N, Pinchas L, Offen D, Barhum Y, Pannet H, Tobar A, Vidne BA, Birk E. Bax deficiency reduces infarct size and improves long‐term function after myocardial infarction. Cell Biochem Biophys 47: 11‐20, 2007.
 353.Hochhauser E, Kivity S, Offen D, Maulik N, Otani H, Barhum Y, Pannet H, Shneyvays V, Shainberg A, Goldshtaub V, Tobar A, Vidne BA. Bax ablation protects against myocardial ischemia‐reperfusion injury in transgenic mice. Am J Physiol Heart Circ Physiol 284: H2351‐H2359, 2003.
 354.Hoda MN, Singh I, Singh AK, Khan M. Reduction of lipoxidative load by secretory phospholipase A2 inhibition protects against neurovascular injury following experimental stroke in rat. J Neuroinflammation 6: 21, 2009.
 355.Hodgkinson CP, Bareja A, Gomez JA, Dzau VJ. Emerging concepts in paracrine mechanisms in regenerative cardiovascular medicine and biology. Circ Res 118: 95‐107, 2016.
 356.Hofmann U, Frantz S. Role of lymphocytes in myocardial injury, healing, and remodeling after myocardial infarction. Circ Res 116: 354‐367, 2015.
 357.Hofmann U, Frantz S. Role of T‐cells in myocardial infarction. Eur Heart J 37: 873‐879, 2015.
 358.Holler N, Zaru R, Micheau O, Thome M, Attinger A, Valitutti S, Bodmer JL, Schneider P, Seed B, Tschopp J. Fas triggers an alternative, caspase‐8‐independent cell death pathway using the kinase RIP as effector molecule. Nat Immunol 1: 489‐95, 2000.
 359.Holly TA, Drincic A, Byun Y, Nakamura S, Harris K, Klocke FJ, Cryns VL. Caspase inhibition reduces myocyte cell death induced by myocardial ischemia and reperfusion in vivo. J Mol Cell Cardiol 31: 1709‐1715, 1999.
 360.Horie Y, Ishii H. Liver dysfunction elicited by gut ischemia‐reperfusion. Pathophysiology 8: 11‐20, 2001.
 361.Horie Y, Wolf R, Russell J, Shanley TP, Granger DN. Role of Kupffer cells in gut ischemia/reperfusion‐induced hepatic microvascular dysfunction in mice. Hepatology 26: 1499‐1505, 1997.
 362.Horstman LL, Jy W, Bidot CJ, Nordberg ML, Minagar A, Alexander JS, Kelley RE, Ahn YS. Potential roles of cell‐derived microparticles in ischemic brain disease. Neurol Res 31: 799‐806, 2009.
 363.Houlden A, Goldrick M, Brough D, Vizi ES, Lenart N, Martinecz B, Roberts IS, Denes A. Brain injury induces specific changes in the caecal microbiota of mice via altered autonomic activity and mucoprotein production. Brain Behav Immun 57: 10‐20, 2016.
 364.Howangyin KY, Silvestre JS. Diabetes mellitus and ischemic diseases: Molecular mechanisms of vascular repair dysfunction. Arterioscler Thromb Vasc Biol 34: 1126‐1135, 2014.
 365.Hu CJ, Chen SD, Yang DI, Lin TN, Chen CM, Huang TH, Hsu CY. Promoter region methylation and reduced expression of thrombospondin‐1 after oxygen‐glucose deprivation in murine cerebral endothelial cells. J Cereb Blood Flow Metab 26: 1519‐26, 2006.
 366.Hu Y, Deng H, Xu S, Zhang J. MicroRNAs regulate mitochondrial function in cerebral ischemia‐reperfusion injury. Int J Mol Sci 16: 24895‐24917, 2015.
 367.Huang C, Andres AM, Ratliff EP, Hernandez G, Lee P, Gottlieb RA. Preconditioning involves selective mitophagy mediated by Parkin and p62/SQSTM1. PLoS One 6: e20975, 2011.
 368.Huang C, Liu W, Perry CN, Yitzhaki S, Lee Y, Yuan H, Tsukada YT, Hamacher‐Brady A, Mentzer RM, Jr., Gottlieb RA. Autophagy and protein kinase C are required for cardioprotection by sulfaphenazole. Am J Physiol Heart Circ Physiol 298: H570‐579, 2010.
 369.Huang JQ, Radinovic S, Rezaiefar P, Black SC. In vivo myocardial infarct size reduction by a caspase inhibitor administered after the onset of ischemia. Eur J Pharmacol 402: 139‐142, 2000.
 370.Huang Y, Rabb H, Womer KL. Ischemia‐reperfusion and immediate T cell responses. Cell Immunol 248: 4‐11, 2007.
 371.Huang H, Tohme S, Al‐Khafaji AB, Tai S, Loughran P, Chen L, Wang S, Kim J, Billiar T, Wang Y, Tsung A. Damage‐associated molecular pattern‐activated neutrophil extracellular trap exacerbates sterile inflammatory liver injury. Hepatology 62: 600‐614, 2015.
 372.Hughes BG, Schulz R. Targeting MMP‐2 to treat ischemic heart injury. Basic Res Cardiol 109: 424, 2014.
 373.Hullinger TG, Montgomery RL, Seto AG, Dickinson BA, Semus HM, Lynch JM, Dalby CM, Robinson K, Stack C, Latimer PA, Hare JM, Olson EN, van Rooij E. Inhibition of miR‐15 protects against cardiac ischemic injury. Circ Res 110: 71‐81, 2011.
 374.Hulsmans M, De Keyzer D, Holvoet P. MicroRNAs regulating oxidative stress and inflammation in relation to obesity and atherosclerosis. FASEB J 25: 2515‐2527, 2011.
 375.Humphreys MR, Castle EP, Lohse CM, Sebo TJ, Leslie KO, Andrews PE. Renal ischemia time in laparoscopic surgery: An experimental study in a porcine model. Int J Urol 16: 105‐109, 2009.
 376.Ikeda H, Suzuki Y, Suzuki M, Koike M, Tamura J, Tong J, Nomura M, Itoh G. Apoptosis is a major mode of cell death caused by ischaemia and ischaemia/reperfusion injury to the rat intestinal epithelium. Gut 42: 530‐537, 1998.
 377.Inagaki T, Akiyama T, Du CK, Zhan DY, Yoshimoto M, Shirai M. Monoamine oxidase‐induced hydroxyl radical production and cardiomyocyte injury during myocardial ischemia‐reperfusion in rats. Free Radic Res 50: 645‐653, 2016.
 378.Inagaki K, Chen L, Ikeno F, Lee FH, Imahashi K, Bouley DM, Rezaee M, Yock PG, Murphy E, Mochly‐Rosen D. Inhibition of delta‐protein kinase C protects against reperfusion injury of the ischemic heart in vivo. Circulation 108: 2304‐2307, 2003.
 379.Inagaki K, Churchill E, Mochly‐Rosen D. Epsilon protein kinase C as a potential therapeutic target for the ischemic heart. Cardiovasc Res 70: 222‐230, 2006.
 380.Inderbitzen D, Beldi G, Avital I, Vinci G, Candinas D. Local and remote ischemia‐reperfusion injury is mitigated in mice overexpressing human C1 inhibitor. Eur J Surg Res 36a: 142‐147, 2004.
 381.Women's Health Research: Progress, Pitfalls, and Promise. Report Brief. Institute of Medicine. Committee on Women's Health Research, National Academies Press, Washington, DC, 2010.
 382.Ioannou A, Dalle Lucca J, Tsokos GC. Immunopathogenesis of ischemia/reperfusion‐associated tissue damage. Clin Immunol 141: 3‐14, 2011.
 383.Ivanov II, Frutos Rde L, Manel N, Yoshinaga K, Rifkin DB, Sartor RB, Finlay BB, Littman DR. Specific microbiota direct the differentiation of IL‐17‐producing T‐helper cells in the mucosa of the small intestine. Cell Host Microbe 4: 337‐349, 2008.
 384.Jaeschke H, Bautista AP, Spolarics Z, Farhood A, Spitzer JJ. Enhanced generation of superoxide by complement‐stimulated Kupffer cells and priming of neutrophils during the initial reperfusion phase after hepatic ischemia. In: Proceedings of the 5th International Congress on Oxygen Radicals. Elsevier Science Publishers, Amsterdam, 1993.
 385.Jaeschke H, Bautista AP, Spolarics Z, Spitzer JJ. Superoxide generation by neutrophils and Kupffer cells during in vivo reperfusion after hepatic ischemia in rats. J Leukoc Biol 52: 377‐382, 1992.
 386.Jaeschke H, Farhood A, Fisher MA, Smith CW. Sequestration of neutrophils in the hepatic vasculature during endotoxemia is independent of beta 2 integrins and intercellular adhesion molecule‐1. Shock 6: 351‐356, 1996.
 387.Jaeschke H, Farhood A, Kupffer. Cell activation after no‐flow ischemia versus hemorrhagic shock. Free Rad Biol Med 33: 210‐219, 2002.
 388.Jaeschke H, Farhood A, Smith CW. Neutrophils contribute to ischemia‐reperfusion injury in rat liver in vivo. FASEB J 4: 3355‐3359, 1990.
 389.Jang HR, Gandolfo MT, Ko GJ, Satpute S, Racusen L, Rabb H. Early exposure to germs modifies kidney damage and inflammation after experimental ischemia‐reperfusion injury. Am J Physiol Renal Physiol 297: F1457‐F1465, 2009.
 390.Jang HR, Rabb H. Immune cells in experimental acute kidney injury. Nat Rev Nephrol 11: 88‐101, 2015.
 391.Jennings RB. Historical perspective on the pathology of myocardial ischemia/reperfusion injury. Circ Res 113: 428‐438, 2013.
 392.Jennings RB, Reimer KA. The cell biology of acute myocardial ischemia. Annu Rev Med 42: 225‐246, 1991.
 393.Jennings RB, Sommers HM, Smyth GA, Flack HA, Linn H. Myocardial necrosis induced by temporary occlusion of a coronary artery in the dog. Arch Pathol 70: 68‐78, 1960.
 394.Jeremias I, Kupatt C, Martin‐Villalba A, Habazettl H, Schenkel J, Boekstegers P, Debatin KM. Involvement of CD95/Apo1/Fas in cell death after myocardial ischemia. Circulation 102: 915‐920, 2000.
 395.Jerome SN, Akimitsu T, Gute DC, Korthuis RJ. Ischemic preconditioning attenuates capillary no‐reflow induced by prolonged ischemia and reperfusion. Am J Physiol 266: H1316‐H1321, 1995.
 396.Jerome SN, Akimitsu T, Korthuis RJ. Leukocyte adhesion, edema, and development of postischemic capillary no‐reflow. Am J Physiol 267: H1329‐1336, 1994.
 397.Jerome SN, Dore M, Paulson JC, Smith CW, Korthuis RJ. P‐selectin and ICAM‐1‐dependent adherence reactions: Role in the genesis of postischemic no‐reflow. Am J Physiol 266: H1316‐H1321, 1994.
 398.Jerome SN, Smith CW, Korthuis RJ. CD18‐dependent adherence reactions play an important role in the development of the no‐reflow phenomenon. Am J Physiol 264: H479‐483, 1993.
 399.Jeyaseelan K, Lim KY, Armugam A. MicroRNA expression in the blood and brain of rats subjected to transient focal ischemia by middle cerebral artery occlusion. Stroke 39: 959‐966, 2008.
 400.Ji X, Luo Y, Ling F, Stetler RA, Lan J, Cao G, Chen J. Mild hypothermia diminishes oxidative DNA damage and pro‐death signaling events after cerebral ischemia: A mechanism for neuroprotection. Front Biosci 12: 1737‐1747, 2007.
 401.Jiang M, Liu K, Luo J, Dong Z. Autophagy is a renoprotective mechanism during in vitro hypoxia and in vivo ischemia‐reperfusion injury. Am J Pathol 176: 1181‐1192, 2010.
 402.Jiang X, Ma H, Li C, Cao Y, Wang Y, Zhang Y, Liu Y. Effects of neonatal dexamethasone administration on cardiac recovery ability under ischemia‐reperfu‐sion in 24‐wk‐old rats. Pediatr Res 80: 128‐135, 2016.
 403.Jiang M, Zhang Y, Dusting GJ. NADPH oxidase‐mediated redox signaling: Roles in cellular stress response, stress tolerance, and tissue repair. Pharmacol Rev 63: 218‐242, 2011.
 404.Jin G, Arai K, Murata Y, Wang S, Stins MF, Lo EH, van Leyen K. Protecting against cerebrovascular injury: Contributions of 12/15‐lipoxygenase to edema formation after transient focal ischemia. Stroke 39: 2538‐2543, 2008.
 405.Jin R, Yang G, Li G. Inflammatory mechanisms in ischemic stroke: Role of inflammatory cells. J Leukoc Biol 87(5): 779‐789, 2010.
 406.Jin Y, Silverman AJ, Vannucci SJ. Mast cell stabilization limits hypoxic‐ischemic brain damage in the immature rat. Dev Neurosci 9: 373‐384, 2007.
 407.Jin Y, Silverman AJ, Vannucci SJ. Mast cells are early responders after hypoxia‐ischemia in immature rat brain. Stroke 40: 3107‐3112, 2009.
 408.Johnston WH, Latta H. Glomerular mesangial and endothelial cell swelling following temporary renal ischemia and its role in the no‐reflow phenomenon. Am J Pathol 89: 153‐166, 1977.
 409.Jones AW, Durante W, Korthuis RJ. Heme oxygenase‐1 deficiency leads to alteration of soluble guanylate cylcase redox regulation. J Pharmacol Exp Therap 335: 85‐91, 2010.
 410.Jones DP, Sies H. The redox code. Antioxid Redox Signal 23: 734‐746, 2015.
 411.Jones SP, Tang XL, Guo Y, Steenbergen C, Lefer DJ, Kukreja RC, Kong M, Li Q, Bhushan S, Zhu X, Du J, Nong Y, Stowers HL, Kondo K, Hunt GN, Goodchild TT, Orr A, Chang CC, Ockaili R, Salloum FN, Bolli R. The NHLBI‐sponsored Consortium for preclinicAl assESsment of cARdioprotective therapies (CAESAR): A new paradigm for rigorous, accurate, and reproducible evaluation of putative infarct‐sparing interventions in mice, rabbits, and pigs. Circ Res 116: 572‐586, 2015.
 412.Jong HR, Ko GJ, Wasowska BA, Rabb H. The interaction between ischemia‐reperfusion and immune responses in the kidney. J Mol Med 87: 859‐864, 2009.
 413.Julia PL, Kofsky ER, Buckberg GD, Young HH, Bugyi HI. Studies of myocardial protection in the immature heart. I. Enhanced tolerance of immature versus adult myocardium to global ischemia with reference to metabolic differences. J Thorac Cardiovasc Surg 100: 879‐887, 1990.
 414.Kahle KT, Simard JM, Staley KJ, Nahed BV, Jones PS, Sun D. Molecular mechanisms of ischemic cerebral edema: Role of electroneutral ion transport. Physiology (Bethesda) 24: 257‐265, 2009.
 415.Kaiser RA, Bueno OF, Lips DJ, Doevendans PA, Jones F, Kimball TF, Molkentin JD. Targeted inhibition of p38 mitogen‐activated protein kinase antagonizes cardiac injury and cell death following ischemia‐reperfusion in vivo. J Biol Chem 279: 15524‐15530, 2004.
 416.Kaiser RA, Liang Q, Bueno O, Huang Y, Lackey T, Klevitsky R, Hewett TE, Molkentin JD. Genetic inhibition or activation of JNK1/2 protects the myocardium from ischemia‐reperfusion‐induced cell death in vivo. J Biol Chem 280: 32602‐32608, 2005.
 417.Kalogeris T, Baines C, Krenz M, Korthuis RJ. Cell biology of ischemia/reperfusion injury. Int Rev Cell Mol Biol 298: 229‐318, 2012.
 418.Kalogeris TJ, Bao Y, Korthuis RJ. Mitochondrial reactive oxygen species: A double‐edged sword in ischemia/reperfusion versus preconditioning. Redox Biol 2: 702‐714, 2014.
 419.Kaludercic N, Carpi A, Menabo R, Di Lisa F, Paolocci N. Monoamine oxidases (MAO) in the pathogenesis of heart failure and ischemia/reperfusion injury. Biochim Biophys Acta 1813: 1323‐1332, 2011.
 420.Kaludercic N, Mialet‐Perez J, Paolocci N, Parini A, Di Lisa F. Monoamine oxidases as sources of oxidants in the heart. J Mol Cell Cardiol 73: 34‐42, 2014.
 421.Kaludercic N, Takimoto E, Nagayama T, Lai E, Chen K, Shih JC, Pacak K, Kass DA, Di Lisa F, Paolocci N. Lack of monoamine oxidase A and B activity prevents heart failure in pressure‐overloaded mice. J Mol Cell Cardiol 48: S13, 2010.
 422.Karp CL. Links between innate and adaptive immunity. In: Serhan CN, Ward PA, Gilroy DW, editors. Fundamentals of Inflammation. New York, NY: Cambridge University Press, 2010, pp. 28‐37.
 423.Karra L, Berent‐Maoz B, Ben‐Zaimra M, Levi‐Schaffer F. Are we ready to downregulate mast cells? Curr Opin Immunol 21: 708‐714, 2009.
 424.Kassahun WT, Schulz T, Richter O, Hauss J. Unchanged high mortality rates from acute occlusive intestinal ischemia: Six year review. Langenbecks Arch Surg 393: 163‐171, 2008.
 425.Kelton JG, Warkentin TE, Hayward CP, Murphy WG, Moore JC. Calpain activity in patients with thrombotic thrombocytopenic purpura is associated with platelet microparticles. Blood 80: 2246‐2251, 1992.
 426.Kemp M, Go YM, Jones DP. Nonequilibrium thermodynamics of thiol/disulfide redox systems: A perspective on redox systems biology. Free Radic Biol Med 44: 921‐937, 2008.
 427.Khalil PN, Neuhof C, Huss R, Pollhammer M, Khalil MN, Neuhof H, Fritz H, Siebeck M. Calpain inhibition reduces infarct size and improves global hemodynamics and left ventricular contractility in a porcine myocardial ischemia/reperfusion model. Eur J Pharmacol 528: 124‐131, 2005.
 428.Khandoga A, Biberthaler P, Enders G, Axmann S, Hatter J, Messmer K, Krombach F. Platelet adhesion mediated by fibrinogen‐intercellular adhesion molecule‐1 binding induces tissue injury in the postischemic liver in vivo. Tansplantation 74: 681‐688, 2002.
 429.Khandoga A, Hanschen M, Kessler JS, Kronbach F. CD4+ T cells contribute to postischemic liver injury in mice by interacting with sinusoidal endothelium and platelets. Hepatology 43: 306‐315, 2006.
 430.Kim HJ, Rowe M, Ren M, Hong JS, Chen PS, Chuang DM. Histone deacetylase inhibitors exhibit anti‐inflammatory and neuroprotective effects in a rat permanent ischemic model of stroke: Multiple mechanisms of action. J Pharmacol Exp Ther 321: 892‐901, 2007.
 431.Kim J, Kim DS, Park MJ, Cho HJ, Zervos AS, Bonventre JV, Park KM. Omi/HtrA2 protease is associated with tubular cell apoptosis and fibrosis induced by unilateral ureteral obstruction. Am J Physiol Renal Physiol 298: F1332‐F1340, 2010.
 432.Kim JJ, Lee SB, Park JK, Yoo YD. TNF‐alpha‐induced ROS production triggering apoptosis is directly linked to Romo1 and Bcl‐X(L). Cell Death Differ 17: 1420‐1434, 2010.
 433.Kim MG, Boo CS, Ko YS, Lee HY, Cho WY, Kim HK, Jo SK. Depletion of kidney CD11c+ F4/80+ cells impairs the recovery process in ischaemia/reperfusion‐induced acute kidney injury. Nephrol Dial Transplant 25: 2908‐2921, 2010.
 434.King LA, Toledo AH, Rivera‐Chavez FA, Toledo‐Pereyra LH. Role of p38 and JNK in liver ischemia and reperfusion. J Hepatobiliary Pancreat Surg 16: 763‐770, 2009.
 435.Kinross JM, Darzi AW, Nicholson JK. Gut microbiome‐host interactions in health and disease. Genome Med 3: 14, 2011.
 436.Kinross J, Warren O, Basson S, Holmes E, Silk D, Darzi A, Nicholson JK. Intestinal ischemia/reperfusion injury: Defining the role of the gut microbiome. Biomark Med 3: 175‐192, 2009.
 437.Kinsey GR, Huang L, Vergis AL, Li L, Okusa MD. Regulatory T cells contribute to the protective effect of ischemic preconditioning in the kidney. Kidney Int 77: 771‐780, 2010.
 438.Kinsey GR, Sharma R, Huang L, Li L, Vergis AL, Ye H, Ju ST, Okusa MD. Regulatory T cells suppress innate immunity in kidney ischemia‐reperfusion injury. J Am Soc Nephrol 20: 1744‐1753, 2009.
 439.Kiray M, Bagriyanik HA, Pekcetin C, Ergur BU, Uysal N. Protective effects of deprenyl in transient cerebral ischemia in rats. Chin J Physiol 51: 275‐281, 2008.
 440.Kloner RA. Current state of clinical translation of cardioprotective agents for acute myocardial infarction. Circ Res 113: 451‐463, 2013.
 441.Kloner RA, Ganote CE, Jennings RB. The “no‐reflow” phenomenon after temporary coronary occlusion in the dog. J Clin Invest 54: 1496‐1508, 1974.
 442.Klotz L, Norman S, Vieira JM, Masters M, Rohling M, Dube KN, Bollini S, Matsuzaki F, Carr CA, Riley PR. Cardiac lymphatics are heterogeneous in origin and respond to injury. Nature 522: 62‐67, 2015.
 443.Kobayashi A, Imamura H, Isobe M, Matsuyama Y, Soeda J, Matsunaga K, Kawasaki S. Mac‐1 (CD11b/CD18) and intercellular adhesion molecule‐1 in ischemia‐reperfusion injury of rat liver. Am J Physiol Gastrointest Liver Physiol 281: G577‐585, 2001.
 444.Kobayashi M, Takeyoshi I, Yoshinari D, Matsumoto K, Morishita Y. P38 mitogen‐activated protein kinase inhibition attenuates ischemia‐reperfusion injury of the rat liver. Surgery 131: 344‐349, 2002.
 445.Kobayasi T, Hirano K, Yamamoto T, Hasegawa G, Hatakeyama K, Suematsu M, Naito M. The protective role of Kupffer cells in the ischemia‐reperfused rat liver. Arch Histol Cytol 65: 251‐261, 2002.
 446.Koboziev I, Reinoso Webb C, Furr KL, Grisham MB. Role of the enteric microbiota in intestinal homeostasis and inflammation. Free Radic Biol Med 68: 122‐133, 2014.
 447.Koeth RA, Wang Z, Levison BS, Buffa JA, Org E, Sheehy BT, Britt EB, Fu X, Wu Y, Li L, Smith JD, DiDonato JA, Chen J, Li H, Wu GD, Lewis JD, Warrier M, Brown JM, Krauss RM, Tang WH, Bushman FD, Lusis AJ, Hazen SL. Intestinal microbiota metabolism of L‐carnitine, a nutrient in red meat, promotes atherosclerosis. Nat Med 19: 576‐585, 2013.
 448.Kohda Y, Gemba M. Cephaloridine induces translocation of protein kinase C d into mitochondria and enhances mitochondrial generation of free radicals in the kidney cortex of rats causing renal dysfunction. J Pharmacol Sci 98: 49‐57, 2005.
 449.Kohli V, Madden JF, Bentley RC, Clavien PA. Calpain mediates ischemic injury of the liver through modulation of apoptosis and necrosis. Gastroenterology 116: 168‐78, 1999.
 450.Kokura S, Wolf RE, Yoshikawa T, Granger DN, Aw TY. Molecular mechanisms of neutrophil‐endothelial cell adhesion induced by redox imbalance. Circ Res 84: 516‐524, 1999.
 451.Kokura S, Wolf RE, Yoshikawa T, Ichikawa H, Granger DN, Aw TY. Endothelial cells exposed to anoxia/reoxygenation are hyperadhesive to T‐lymphocytes: Kinetics and molecular mechanisms. Microcirculation 7: 13‐23, 2000.
 452.Kolettis TM, Barton M, Langleben D, Matsumura Y. Endothelin in coronary artery disease and myocardial infarction. Cardiol Rev 21: 249‐256 2013.
 453.Koleva PT, Kim JS, Scott JA, Kozyrskyj AL. Microbial programming of health and disease starts during fetal life. Birth Defects Res C Embryo Today Rev 105: 265‐277, 2015.
 454.Kondo T, Reaume AG, Huang TT, Carlson E, Murakami K, Chen SF, Hoffman EK, Scott RW, Epstein CJ, Chan PH. Reduction of CuZn‐superoxide dismutase activity exacerbates neuronal cell injury and edema formation after transient focal cerebral ischemia. J Neurosci 17: 4180‐4189, 1997.
 455.Konstantinidis K, Whelan RS, Kitsis RN. Mechanisms of cell death in heart disease. Arterioscler Thromb Vasc Biol 32: 1552‐1562, 2012.
 456.Koponen S, Goldsteins G, Keinänen R, Koistinaho J. Induction of protein kinase Cd subspecies in neurons and microglia after transient global brain ischemia. J Cereb Blood Flow Metab 20: 93‐102, 2000.
 457.Korichneva I. Redox regulation of cardiac protein kinase C. Exp Clin Cardiol 10: 256‐261, 2005.
 458.Kornfeld OS, Hwang S, Disatnik MH, Chen CH, Qvit N, Mochly‐Rosen D. Mitochondrial reactive oxygen species at the heart of the matter: New therapeutic approaches for cardiovascular diseases. Circ Res 116: 1783‐1799, 2015.
 459.Korthuis RJ, Anderson DC, Granger DN. Role of neutrophil‐endothelial cell adhesion in inflammatory disorders. J Crit Care 9: 47‐71, 1994.
 460.Korzick DH, Kostyak JC, Hunter JC, Saupe KW. Local delivery of PKCepsilon‐activating peptide mimics ischemic preconditioning in aged hearts through GSK‐3beta but not F1‐ATPase inactivation. Am J Physiol Heart Circ Physiol 293: H2056‐2063, 2007.
 461.Kosieradzki M, Rowiński W. Ischemia/reperfusion injury in kidney transplantation: Mechanisms and prevention. Transplant Proc 40: 3279‐3288, 2008.
 462.Krenz M, Baines C, Kalogeris T, Korthuis RJ. Cell Survival Programs and Ischemia/Reperfusion: Hormesis, Preconditioning, and Cardiovascular Protection. Integrated Systems Physiology: Molecules to Function eBook series Granger DN, Granger JP, editors. Morgan‐Claypool, 2013.
 463.Krenz, M, Korthuis RJ. Moderate ethanol ingestion and cardiovascular protection: From epidemiologic associations to cellular mechanisms. J Mol Cell Cardiol 52: 93‐104, 2012.
 464.Kristián T. Metabolic stages, mitochondria and calcium in hypoxic/ischemic brain damage. Cell Calcium 36: 221‐233, 2004.
 465.Kristiansen M, Graverson JH, Jacobsen C, Sonne O, Hoffman HJ, Law SK, Moestrup SK. Identification of the haemoglobin scavenger receptor. Nature 409: 198‐201, 2001.
 466.Kroemer G, Galluzzi L, Brenner C. Mitochondrial membrane permeabilization in cell death. Physiol Rev 87: 99‐163, 2007.
 467.Kroemer G, Galluzzi L, Vandenabeele P, Abrams J, Alnemri ES, Baehrecke EH, Blagosklonny MV, El‐Deiry WS, Golstein P, Green DR, Hengartner M, Knight RA, Kumar S, Lipton SA, Malorni W, Nuñez G, Peter ME, Tschopp J, Yuan J, Piacentini M, Zhivotovsky B, Melino G. Classification of cell death: Recommendations of the Nomenclature Committee on Cell Death. Cell Death Differ 16: 3‐11, 2009.
 468.Krutzfeldt J, Rajewsky N, Braich R, Rajeev KG, Tuschl T, Manoharan M, Stoffel M. Silencing of microRNAs in vivo with ‘antagomirs'. Nature 438: 685‐689, 2005.
 469.Kuan CY, Whitmarsh AJ, Yang DD, Liao G, Schloemer AJ, Dong C, Bao J, Banasiak KJ, Haddad GG, Flavell RA, Davis RJ, Rakic P. A critical role of neural‐specific JNK3 for ischemic apoptosis. Proc Natl Acad Sci U S A 100: 15184‐15189, 2003.
 470.Kubli DA, Gustafsson AB. Mitochondria and mitophagy: The yin and yang of cell death control. Circ Res 111: 1208‐1221, 2012.
 471.Kuboki S, Sakai N, Tschöp J, Edwards MJ, Lentsch AB, Caldwell CC. Distinct contributions of CD4+ T cell subsets in hepatic ischemia/reperfusion injury. Am J Physiol 296: G1054‐G1059, 2009.
 472.Kukreja RC, Yin C, Salloum FN. MicroRNAs: New players in cardiac injury and protection. Mol Pharmacol 80: 558‐564, 2011.
 473.Kumar P, Shen Q, Pivetti CD, Lee ES, Wu MH, Yuan SY. Molecular mechanisms of endothelial hyperpermeability: Implications in inflammation. Expert Rev Mol Med 11: e19, 2009.
 474.Kume M, Yamamoto Y, Saad S, Gomi T, Kimoto S, Shimabukuro T, Yagi T, Nakagami M, Takada Y, Morimoto T, and Yamaoka Y. Ischemic preconditioning of the liver in rats: Implications of heat shock protein induction to increase tolerance of ischemia‐reperfusion injury. J Lab Clin Med 128: 251‐258, 1996.
 475.Kuppusamy P, Zweier JL. Cardiac applications of EPR imaging. NMR Biomed 17: 226‐239, 2004.
 476.Kvietys PR, Granger DN. Role of reactive oxygen and nitrogen species in the vascular responses to inflammation. Free Radic Biol Med 52: 556‐592, 2012.
 477.Lagranha CJ, Deschamps A, Aponte A, Steenbergen C, and Murphy E. Sex differences in the phosphorylation of mitochondrial proteins result in reduced production of reactive oxygen species and cardioprotection in females. Circ Res 106: 1681‐1691, 2010.
 478.Lam FW, Burns AR, Smith CW, Rumbaut RE. Platelets enhance neutrophil transendothelial migration via P‐selectin glycoprotein ligand‐1. Am J Physiol Heart Circ Physiol 300: H468‐H475, 2011.
 479.Lam V, Su J, Koprowski S, Hsu A, Tweddell JS, Rafiee P, Gross GJ, Salzman NH, Baker JE. Intestinal microbiota determine severity of myocardial infarction in rats. FASEB J 26: 1727‐1735, 2012.
 480.Lam FW, Vijayan KV, Rumbaut RE. Platelets and their interactions with other immune cells. Compr Physiol 5: 1265‐1280, 2015.
 481.Langley‐Evans SC. Hypertension induced by foetal exposure to a maternal low‐protein diet, in the rat, is prevented by pharmacological blockade of maternal glucocorticoid synthesis. J Hypertens 15: 537‐544, 1997.
 482.Langley‐Evans SC, McMullen S. Developmental origins of adult disease. Med Princ Pract 19: 87‐98, 2010.
 483.Lappas CM, Day YJ, Marshall MA, Engelhardt VH, Linden J. Adenosine A2A receptor activation reduces hepatic ischemia reperfusion injury by inhibiting CD1d‐dependent NKT activation. J Exp Med 203: 2639‐2648, 2006.
 484.Lassègue B, Clempus RE. Vascular NAD(P)H oxidases: Specific features, expression, and regulation. Am J Physiol 285: R277‐R297, 2003.
 485.LATE_Study_Group, Late Assessment of Thrombolytic Efficacy (LATE) study with alteplase 6‐24 hours after onset of acute myocardial infarction. Lancet 342: 759‐66, 1993.
 486.Lazarus B, Messina A, Barker JE, Hurley JV, Romeo R, Morrison WA, Knight KR. The role of mast cells in schaemia‐reperfusion injury in murine skeletal muscle. J Pathol 191: 443‐448, 2000.
 487.Le DA, Wu Y, Huang Z, Matsushita K, Plesnila N, Augustinack JC, Hyman BT, Yuan J, Kuida K, Flavell RA, Moskowitz MA. Caspase activation and neuroprotection in caspase‐3‐deficient mice after in vivo cerebral ischemia and in vitro oxygen glucose deprivation. Proc Natl Acad Sci U S A 99: 15188‐15193, 2002.
 488.Lecour S, Botker HE, Condorelli G, Davidson SM, Garcia‐Dorado D, Engel FB, Ferdinandy P, Heusch G, Madonna R, Ovize M, Ruiz‐Meana M, Schulz R, Sluijter JP, Van Laake LW, Yellon DM, and Hausenloy DJ. ESC working group cellular biology of the heart: Position paper: Improving the preclinical assessment of novel cardioprotective therapies. Cardiovasc Res 104: 399‐411, 2014.
 489.Lee H, Green D, Lai L, Hou YJ, Jensenius JC, Liu D, Cheong C, Park CG, Zhang M. Early complement factors in the local tissue immunocomplex generated during intestinal ischemia/reperfusion injury. Mol Immunol 47: 972‐981, 2010.
 490.Lee HA, Hong SH, Kim JW, Jang IS. Possible involvement of DNA methylation in NKCC1 gene expression during postnatal development and in response to ischemia. J Neurochem 114: 520‐9, 2010.
 491.Lee H‐L., Chen C‐L., Yeh ST, Zweier JL, Chen Y‐R. Biphasic modulation of the mitochondrial electron transport chain in myocardial ischemia and reperfusion. Am J Physiol 302: H1410‐H1422, 2012.
 492.Lee JM, Grabb MC, Zipfel GJ, Choi DW. Brain tissue responses to ischemia. J Clin Invest 106: 723‐731, 2000.
 493.Lee KH, Kim SE, Lee YS. SP600125, a selective JNK inhibitor, aggravates hepatic ischemia‐reperfusion injury. Exp Mol Med 38: 408‐16, 2006.
 494.Lee P, Sata M, Lefer DJ, Factor SM, Walsh K, Kitsis RN. Fas pathway is a critical mediator of cardiac myocyte death and MI during ischemia‐reperfusion in vivo. Am J Physiol Heart Circ Physiol 284: H456‐H463, 2003.
 495.Lee MC, Velayutham M, Komatsu T, Hille R, Zweier JL. Measurement and characterization of superoxide generation from xanthine dehydrogenase: A redox‐regulated pathway of radical generation in ischemic tissues. Biochemistry 53: 6615‐6623, 2014.
 496.Lefer DJ. Do neutrophils contribute to myocardial reperfusion injury? Basic Res Cardiol 97: 263‐267, 2002.
 497.Lei K, Davis RJ. JNK phosphorylation of Bim‐related members of the Bcl2 family induces Bax‐dependent apoptosis. Proc Natl Acad Sci U S A 100: 2432‐7, 2003.
 498.Lejay A, Fang F, John R, Van JA, Barr M, Thaveau F, Chakfe N, Geny B, Scholey JW. Ischemia reperfusion injury, ischemic conditioning and diabetes mellitus. J Mol Cell Cardiol 91: 11‐22, 2016.
 499.Lemay S, Rabb H, Postler G, Singh AK. Prominent and sustained up‐regulation of gp130‐signaling cytokines and the chemokine MIP‐2 in murine renal ischemia‐reperfusion injury. Transplant 69: 959‐963, 2000.
 500.Lerchenberger M, Uhl B, Stark K, Zuchtriegel G, Eckart A, Miller M, Puhr‐Westerheide D, Praetner M, Rehberg M, Khandoga AG, Lauber K, Massberg S, Krombach F, Reichel CA. Matrix metalloproteinases modulate ameboid‐like migration of neutrophils through inflamed interstitial tissue. Blood 122: 770‐780, 2013.
 501.Leroyer AS, Anfosso F, Lacroix R, Sabatier F, Simoncini S, Njock SM, Jourde N, Brunet P, Camoin‐Jau L, Sampol J, Dignat‐George F. Endothelial‐derived microparticles: Biological conveyors at the crossroad of inflammation, thrombosis and angiogenesis. Thromb Haemost 104: 456‐463, 2010.
 502.Lesnefsky EJ, Chen Q, Moghaddas S, Hassan MO, Tandler B, Hoppel CL. Blockade of electron transport during ischemia protects cardiac mitochondria. J Biol Chem 279: 47961‐47967, 2004.
 503.Levine B, Kroemer G. Autophagy in the pathogenesis of disease. Cell 132: 27‐42, 2008.
 504.Levonen AL, Dickinson DA, Moellering DR, Mulcahy RT, Forman HJ, Darley‐Usmar VM. Biphasic effects of 15‐deoxy‐delta (12,14)‐prostaglandin J(2) on glutathione induction and apoptosis in human endothelial cells. Arterioscler Thromb Vasc Biol 21: 1846‐1851, 2001.
 505.Lewen A, Matz P, Chan PH. Free radical pathways in CNS injury. J Neurotrauma 17: 871‐890, 2000.
 506.Li G, Xiao Y, Estrella JL, Ducsay CA, Gilbert RD, Zhang L. Effect of fetal hypoxia on heart susceptibility to ischemia and reperfusion injury in the adult rat. J Soc Gynecol Investig 10: 265‐274, 2003.
 507.Li G, Bae S, Zhang L. Effect of prenatal hypoxia on heat stress‐mediated cardioprotection in adult rat heart. Am J Physiol Heart Circ Physiol 286: H1712‐H1719, 2004.
 508.Li J, Xu J, Cheng Y, Wang F, Song Y, Xiao J. Circulating microRNAs as mirrors of acute coronary syndromes: MiRacle or quagMire? J Cell Mol Med 17: 1363‐1370, 2013.
 509.Li JY, Gu X, Zhang WH, Jia S, Zhou Y. GdCl3 abates hepatic ischemia‐reperfusion injury by inhibiting apoptosis in rats. Hepatobil Panceat Dis Int 8: 518‐523, 2009.
 510.Li R, Ding T, Liu X, Li C. Influence of SB203580 on cell apoptosis and P38MAPK in renal ischemia/reperfusion injury. J Huazhong Univ Sci Technolog Med Sci 26: 50‐52, 2006.
 511.Liao L, Harris NR, Granger DN. Oxidized low‐density lipoproteins and microvascular responses to ischemia‐reperfusion. Am J Physiol 271: H2508‐H2514, 1996.
 512.Liesz A, Dalpke A, Mracsko E, Antoine DJ, Roth S, Zhou W, Yang H, Na SY, Akhisaroglu M, Fleming T, Eigenbrod T, Nawroth PP, Tracey KJ, Veltkamp R. DAMP signaling is a key pathway inducing immune modulation after brain injury. J Neuroscience 35: 583‐598, 2015.
 513.Liesz AL, Suri‐Payer E, Veltkamp C, Doerr H, Sommer C, Rivest S, Giese T, Veltkamp R. Regulatory T cells are key cerebroprotective immunomodulators in acute experimental stroke. Nature Med 15: 192‐199, 2009.
 514.Lima B, Forrester MT, Hess DT, Stamler JS. S‐nitrosylation in cardiovascular signaling. Circ Res 106: 633‐646, 2010.
 515.Lindemann S, Klingel B, Fisch A, Meyer J, Darius H. Increased platelet sensitivity toward platelet inhibitors during physical exercise in patients with coronary artery disease. Thromb Res 93: 51‐59, 1999.
 516.Lindsay T, Romaschin A, Walker PM. Free radical mediated damage in skeletal muscle. Microcirc Endothelium Lymphatics 5: 157‐170, 1989.
 517.Lindsey ML, Mayr M, Gomes AV, Delles C, Arrell DK, Murphy AM, Lange RA, Costello CE, Jin YF, Laskowitz DT, Sam F, Terzic A, Van Eyk J, Srinivas PR. Transformative impact of proteomics on cardiovascular health and disease: A scientific statement from the American Heart Association. Circulation 132, 852‐872, 2015.
 518.Linfert D, Austen WG, Jr., Rabb H. Lymphocytes and ischemia‐reperfusion injury. Transplant Rev 23, 1‐10, 2009.
 519.Linkermann A, Brasen JH, Darding M, Jin MK, Sanz AB, Heller JO, De Zen F, Weinlich R, Ortiz A, Walczak H, Weinberg JM, Green DR, Kunzendorf U, Krautwald S. Two independent pathways of regulated necrosis mediate ischemia‐reperfusion injury. Proc Natl Acad Sci U S A 110: 12024‐12029, 2013.
 520.Linkermann A, Brasen JH, Himmerkus N, Liu S, Huber TB, Kunzendorf U, Krautwald S. Rip1 (receptor‐interacting protein kinase 1) mediates necroptosis and contributes to renal ischemia/reperfusion injury. Kidney Int 81: 751‐761, 2012.
 521.Linkermann A, De Zen F, Weinberg J, Kunzendorf U, Krautwald S. Programmed necrosis in acute kidney injury. Nephrol Dial Transplant 27: 3412‐3419, 2012.
 522.Linkermann A, Hackl MJ, Kunzendorf U, Walczak H, Krautwald S, Jevnikar AM. Necroptosis in immunity and ischemia‐reperfusion injury. Am J Transplant 13: 2797‐2804, 2013.
 523.Linkermann A, Heller JO, Prokai A, Weinberg JM, De Zen F, Himmerkus N, Szabo AJ, Brasen JH, Kunzendorf U, Krautwald S. The RIP1‐kinase inhibitor necrostatin‐1 prevents osmotic nephrosis and contrast‐induced AKI in mice. J Am Soc Nephrol 24: 1545‐1557, 2013.
 524.Linkermann A, Skouta R, Himmerkus N, Mulay SR, Dewitz C, De Zen F, Prokai A, Zuchtriegel G, Krombach F, Welz PS, Green DR. Necroptosis. N Engl J Med 370: 455‐465
 525.Lira VA, Brown DL, Lira AK, Kavazis AN, Soltow QA, Zeanah EH, Criswell DS. Nitric oxide and AMPK cooperatively regulate PGC‐1 in skeletal muscle cells. J Physiol 588: 3551‐3566, 2010.
 526.Liu D, Gan X, Huang P, Chen X, Ge M, Hei Z. Inhibiting tryptase after ischemia limits small intestinal ischemia‐reperfusion injury through protease‐activated receptor 2 in rats. J Trauma Acute Care Surg 73: 1138‐1144, 2012.
 527.Liu L, Kubes P. Molecular mechanisms of leukocyte recruitment: Organ‐specific mechanisms of action. Thromb Haemost 89: 213‐220, 2003.
 528.Liu M, Chien CC, Grigoryev DN, Gandalfo MT, Colvin RB, Rabb H. Effect of T cells on vascular permeability in early ischemic acute kidney injury in mice. Microvasc Res 77: 340‐347, 2009.
 529.Liu P, McGuire PM, Fisher MA, Farhood A, Smith CW, Jaeschke H. Activation of Kupffer cells and neutrophils for reactive oxygen formation is responsible for endotoxin‐enhanced liver injury after hepatic ischemia. Shock 3: 56‐62, 1995.
 530.Liu XH, Zhang ZY, Sun S, Wu XD. Ischemic postconditioning protects myocardium from ischemia/reperfusion injury through attenuating endoplasmic reticulum stress. Shock 30: 422‐427, 2008.
 531.Liu Y, Kato H, Nakata N, Kogure K. Protection of rat hippocampus against ischemic neuronal damage by pretreatment with sublethal ischemia. Brain Res 586: 121‐124, 1992.
 532.Loi P, Paular F, Pajak B, Nagy N, Salmon I, Moser M, Goldman M, Flamand V. The fate of dendritic cells in a mouse model of liver ischemia/reperfusion injury. Transplant Proc 36: 1275‐1279, 2004.
 533.Lorenzen JM. Vascular and circulating microRNAs in renal ischaemia‐reperfusion injury. J Physiol 593: 1777‐1784, 2015.
 534.Lu X, Kwong JQ, Molkentin JD, Bers DM. Individual Cardiac Mitochondria Undergo Rare Transient Permeability Transition Pore Openings. Circ Res 118: 834‐841, 2016.
 535.Lu YY, Li ZZ, Jiang DS, Wang L, Zhang Y, Chen K, Zhang XF, Liu Y, Fan GC, Chen Y, Yang Q, Zhou Y, Zhang XD, Liu DP, Li H. TRAF1 is a critical regulator of cerebral ischaemia‐reperfusion injury and neuronal death. Nat Commun 4: 2852, 2013.
 536.Lu X, Li N, Shushakova N, Schmitt R, Menne J, Susnik N, Meier M, Leitges M, Haller H, Gueler F, Rong S. C57BL/6 and 129/Sv mice: Genetic difference to renal ischemia‐reperfusion. J Nephrol 25: 738‐743, 2012.
 537.Lu X, Li N, Shushakova N, Schmitt R, Menne J, Susnik N, Meier M, Leitges M, Haller H, Gueler F, Rong S, LueddeLuedde M, Lutz M, Carter N, Sosna J, Jacoby C, Vucur M, Gautheron J, Roderburg C, Borg N, Reisinger F, Hippe HJ, Linkermann A, Wolf MJ, Rose‐John S, Lullmann‐Rauch R, Adam D, Flogel U, Heikenwalder M, Luedde T, Frey N. RIP3, a kinase promoting necroptotic cell death, mediates adverse remodelling after myocardial infarction. Cardiovasc Res 103: 206‐216, 2014.
 538.Lucchesi BR, Kilgore KS. Complement inhibitors in myocardial ischemia/reperfusion injury. Immunopharmacology 38: 27‐42, 1997.
 539.Ma M. Role of calpains in the injury‐induced dysfunction and degeneration of the mammalian axon. Neurobiol Dis 60: 61‐79, 2013.
 540.Ma X, Becker Buscaglia LE, Barker JR, Li Y. MicroRNAs in NF‐kappaB signaling. J Mol Cell Biol 3: 159‐166, 2011.
 541.Ma Y, Wang J, Wang Y, Yang GY. The biphasic function of microglia in ischemic stroke. Prog Neurobiol 2016. pii: S0301‐0082(15)30070‐8. doi: 10.1016/j.pneurobio.2016.01.005. [Epub ahead of print].
 542.Maclean D, Fishbein MC, Braunwald E, Maroko PR. Long‐term preservation of ischemic myocardium after experimental coronary artery occlusion. J Clin Invest 61: 541‐551, 1978.
 543.Maekawa A, Lee JK, Nagaya T, Kamiya K, Yasui K, Horiba M, Miwa K, Uzzaman M, Maki M, Ueda Y, Kodama I. Overexpression of calpastatin by gene transfer prevents troponin I degradation and ameliorates contractile dysfunction in rat hearts subjected to ischemia/reperfusion. J Mol Cell Cardiol. 35: 1277‐84, 2003.
 544.Maggi CA. The effects of tachykinin on inflammatory and immune cells. Regul Petides 70: 75‐90, 1997.
 545.Mao XR, Crowder CM. Protein misfolding induces hypoxic preconditioning via a subset of the unfolded protein response machinery. Mol Cell Biol 30: 5033‐5042, 2010.
 546.Mariappan N, Soorappan RN, Haque M, Sriramula, S, Francis J. TNF‐α‐induced mitochondrial stress and cardiac dysfunction: Restoration by superoxide dismutase mimetic Tempol. Am J Physiol 293: H2776‐H2737, 2007.
 547.Maroko PR, Carpenter CB, Chiariello M, Fishbein MC, Radvany P, Knostman JD, Hale SL. Reduction by cobra venom factor of myocardial necrosis after coronary artery occlusion. J Clin Invest 61: 661‐670, 1978.
 548.Marshall HE, Merchant K, Stamler JS. Nitrosation and oxidation in the regulation of gene expression. FASEB J. 14: 1889‐1900, 2000.
 549.Martin M, Mory C, Piescher A, Wittekind C, Fiedler M, Uhlmann D. Protective effects of early CD4 +T cell reduction in hepatic ischemia/reperfusion injury. J Gastrointest Surg 14: 511‐519, 2010.
 550.Martindale JJ, Fernandez R, Thuerauf D, Whittaker R, Gude N, Sussman MA, Glembotski CC. Endoplasmic reticulum stress gene induction and protection from ischemia/reperfusion injury in the hearts of transgenic mice with a tamoxifen‐regulated form of ATF6. Circ Res 98: 1186‐1193, 2006.
 551.Martindale JJ, Metzger JM. Uncoupling of increased cellular oxidative stress and myocardial ischemia reperfusion injury by directed sarcolemma stabilization. J Mol Cell Cardiol 67: 26‐37, 2014.
 552.Martinez D, Pentinat T, Ribo S, Daviaud C, Bloks VW, Cebria J, Villalmanzo N, Kalko SG, Ramon‐Krauel M, Diaz R, Plosch T, Tost J, Jimenez‐Chillaron JC. In utero undernutrition in male mice programs liver lipid metabolism in the second‐generation offspring involving altered Lxra DNA methylation. Cell Metab 19: 941‐951, 2014.
 553.Martyn CN, Hales CN, Barker DJ, Jespersen S. Fetal growth and hyperinsulinaemia in adult life. Diabet Med 15: 688‐694, 1998.
 554.Marzocco S, Di Paola R, Autore G, Mazzon E, Pinto A, Caputi AP, Thiemermann C, Cuzzocrea S. Calpain inhibitor I reduces intestinal ischemia‐reperfusion injury in the rat. Shock 21: 38‐44, 2004.
 555.Massberg S, Enders G, Matos FC, Tomic LI, Leiderer R, Eisenmenger S, Messmer K, Krombach F. Fibrinogen deposition at the postischemic vessel wall promotes platelet adhesion during ischemia‐reperfusion in vivo. Blood 94: 3829‐3838, 1999.
 556.Matsui Y, Kyoi S, Takagi H, Hsu CP, Hariharan N, Ago T, Vatner SF, Sadoshima J. Molecular mechanisms and physiological significance of autophagy during myocardial ischemia and reperfusion. Autophagy 4: 409‐415, 2008.
 557.Matsui Y, Takagi H, Qu X, Abdellatif M, Sakoda H, Asano T, Levine B, Sadoshima J. Distinct roles of autophagy in the heart during ischemia and reperfusion: Roles of AMP‐activated protein kinase and Beclin 1 in mediating autophagy. Circ Res 100: 914‐922, 2007.
 558.Matsumoto S, Sakata Y, Nakatani D, Suna S, Mizuno H, Shimizu M, Usami M, Sasaki T, Sato H, Kawahara Y, Hamasaki T, Nanto S, Hori M, Komuro I. A subset of circulating microRNAs are predictive for cardiac death after discharge for acute myocardial infarction. Biochem Biophys Res Commun 427: 280‐284, 2012.
 559.Matsumoto S, Sakata Y, Suna S, Nakatani D, Usami M, Hara M, Kitamura T, Hamasaki T, Nanto S, Kawahara Y, Komuro I. Circulating p53‐responsive microRNAs are predictive indicators of heart failure after acute myocardial infarction. Circ Res 113: 322‐326, 2013.
 560.Matzinger P. The danger model: A renewed sense of self. Science 296: 301‐305, 2002.
 561.Maurer M, Wedemeyer J, Metz M, Piliponsky AM, Welle,r K., Chatterjea D, Clouthier DE, Yanagisawa MM. Mast cells promote homeostasis by limiting endothelin‐1‐induced toxicity. Nature 432: 512‐516, 2004.
 562.Mause SF, Weber C. Microparticles: Protagonists of a novel communication network for intercellular information exchange. Circ Res 107: 1047‐1057, 2010.
 563.Mazzoni MC, Borgstrom P, Warnke KC, Skalak TC, Intaglietta M, Arfors KE. Mechanisms and implications of capillary endothelial swelling and luminal narrowing in low‐flow ischemias. Int J Microcirc Clin Exp 15: 265‐270, 1995.
 564.McCall CE, El Gazzar M, Liu T, Vachharajani V, Yoza B. Epigenetics, bioenergetics, and microRNA coordinate gene‐specific reprogramming during acute systemic inflammation. J Leukoc Biol 90: 439‐446, 2011.
 565.McDonald B, Pittman K, Menezes GB, Hirota SA, Slaba I, Waterhouse CCM, Beck PL, Muruve DA, Kubes P. Intravascular danger signals guide neutrophils to sites of sterile inflammation. Science 330: 362‐366, 2010.
 566.McDougal WS. Renal perfusion/reperfusion injuries. J Urol 140: 1325‐30, 1988.
 567.McManus DD, Freedman JE. MicroRNAs in platelet function and cardiovascular disease. Nat Rev Cardiol 12: 711‐717, 2015.
 568.Mehta D, Malik AB. Signaling mechanisms regulating endothelial permeability. Physiol. Rev 86: 279‐367, 2006.
 569.Meldrum DR, Cleveland JC, Jr., Meng X, Sheridan BC, Gamboni F, Cain BS, Harken AH, Banerjee A. Protein kinase C isoform diversity in preconditioning. J Surg Res 69: 183‐187, 1997.
 570.Meller R, Pearson A, Simon RP. Dynamic changes in DNA methylation in ischemic tolerance. Front Neurol 6: 102, 2015.
 571.Mendez I, Vazquez‐Martinez O, Hernandez‐Munoz R, Valente‐Godinez H, Diaz‐Munoz M. Redox regulation and pro‐oxidant reactions in the physiology of circadian systems. Biochimie 124: 178‐186, 2016.
 572.Menger MD, Thierjung C, Hammersen F, Messmer K. Dextran vs. hydroxyethylstarch in ischemic tolerance. Frontiersinhibitioninhibition of postischemic leukocyte adherence in neurology 6, 102striatedstriated muscle. Circ Shock 41: 248‐255, 1993.
 573.Menger MD, Pelikan S, Steiner D, Messmer K. Microvascular ischemia‐reperfusion injury in striated muscle: Significance of “reflow paradox”. Am J Physiol 263: H1901‐1906, 1992.
 574.Menger MD, Steiner D, Messmer K. Microvascular ischemia‐reperfusion injury in striated muscle: Significance of “no reflow”. Am J Physiol 263: H1892‐1900, 1992.
 575.Mengesdorf T, Jensen PH, Mies G, Aufenberg C, Paschen W. Down‐regulation of parkin protein in transient focal cerebral ischemia: A link between stroke and degenerative disease? Proc Natl Acad Sci U S A 99: 15042‐15047, 2002.
 576.Menini S, Amadio L, Oddi G, Ricci C, Pesci C, Pugliese F, Giorgio M, Migliaccio E, Pelicci P, Iacobini C, Pugliese G. Deletion of p66Shc longevity gene protects against experimental diabetic glomerulopathy by preventing diabetes‐induced oxidative stress. Diabetes 55: 1642‐1650, 2006.
 577.Metukuri MR, Beer‐Stolz D, Namas RA, Dhupar R, Torres A, Loughran PA, Jefferson BS, Tsung A, Billiar TR, Vodovotz Y, Zamora R. Expression and subcellular localization of BNIP3 in hypoxic hepatocytes and liver stress. Am J Physiol Gastrointest Liver Physiol 296: G499‐509, 2009.
 578.Metz M, Maurer M. Mast cells‐key effector cells in immune responses. Trends Immunol 28: 234‐241, 2007.
 579.Meyer K, Zhang H, Zhang L. Direct effect of cocaine on epigenetic regulation of PKCe gene repression in the fetal rat heart. J Mol Cell Cardiol 47, 504‐511, 2009.
 580.Miao W, Qu Z, Shi K, Zhang D, Zong Y, Zhang G, Hu S. RIP3 S‐nitrosylation contributes to cerebral ischemic neuronal injury. Brain Res 1627: 165‐176, 2015.
 581.Milerova M, Charvatova Z, Skarka L, Ostadalova I, Drahota Z, Fialova M, Ostadal B. Neonatal cardiac mitochondria and ischemia/reperfusion injury. Mol Cell Biochem 335: 147‐153, 2010.
 582.Minamino T, Komuro I, Kitakaze M. Endoplasmic reticulum stress as a therapeutic target in cardiovascular disease. Circ Res 107: 1071‐1082, 2010.
 583.Miura T, Miki T. Limitation of myocardial infarct size in the clinical setting: Current status and challenges in translating animal experiments into clinical therapy. Basic Res Cardiol 103: 501‐513, 2008.
 584.Moquin D, Chan FK. The molecular regulation of programmed necrotic cell injury. Trends Biochem Sci 35: 434‐441, 2010.
 585.Morgan MJ, Kim YS, Liu ZG. TNFa and reactive oxygen species in necrotic cell death. Cell Res 18: 343‐349, 2008.
 586.Moritz KM, De Matteo R, Dodic M, Jefferies AJ, Arena D, Wintour EM, Probyn ME, Bertram JF, Singh RR, Zanini S, Evans RG. Prenatal glucocorticoid exposure in the sheep alters renal development in utero: Implications for adult renal function and blood pressure control. Am J Physiol Regul Integr Comp Physiol 301: R500‐R509, 2011.
 587.Muller FL, Song W, Liu Y, Chaudhuri A, Pieke‐Dahl S, Strong R, Huang TT, Epstein CJ, Roberts LJ, Jr., Csete M, Faulkner JA, Van Remmen H. Absence of CUZn superoxide dismutase leads to elevated oxidative stress and acceleration of age‐dependent skeletal muscle atrophy. Free Radic Biol Med 40: 1993‐2004, 2006.
 588.Muramatsu Y, Furuichi Y, Tojo N, Moriguchi A, Maemoto T, Nakada H, Hino M, Matsuoka N. Neuroprotective efficacy of FR901459, a novel derivative of cyclosporin A, in in vitro mitochondrial damage and in vivo transient cerebral ischemia models. Brain Res 1149: 181‐190, 2007.
 589.Murayama T, Tanabe M, Matsuda S, Shimazu M, Kamei S, Wakabayashi G, Kawachi S, Matsumoto K, Yamazaki K, Matsumoto K, Koyasu S, Kitajima M. JNK (c‐Jun NH2 terminal kinase) and p38 during ischemia reperfusion injury in the small intestine. Transplantation 81: 1325‐1330, 2006.
 590.Murphy E, Steenbergen C. Ion transport and energetics during cell death and protection. Physiology (Bethesda) 23: 115‐123, 2008.
 591.Murphy SP, Porrett PM, Turka LA. Innate immunity in transplant tolerance and rejection. Immunol Rev 241: 39‐48, 2011.
 592.Murriel CL, Churchill E, Inagaki K, Szweda LI, Mochly‐Rosen D. Protein kinase Cd activation induces apoptosis in response to cardiac ischemia and reperfusion damage: A mechanism involving BAD and the mitochondria. J Biol Chem 279: 47985‐47991, 2004.
 593.Murriel CL, Mochly‐Rosen D. Opposing roles of delta and epsilonPKC in cardiac ischemia and reperfusion: Targeting the apoptotic machinery. Arch Biochem Biophys 420: 246‐254, 2003.
 594.Murry CE, Jennings RB, Reimer KA. Preconditioning with ischemia: A delay of lethal cell injury in ischemic myocardium. Circulation 74: 1124‐1136, 1986.
 595.Murry CE, Soonpaa MH, Reinecke H, Nakajima H, Nakajima HO, Rubart M, Pasumarthi KB, Virag JI, Bartelmez SH, Poppa V, Bradford G, Dowell JD, Williams DA, Field LJ. Haematopoietic stem cells do not transdifferentiate into cardiac myocytes in myocardial infarcts. Nature 428: 664‐668, 2004.
 596.Muschen M, Warskulat U, Peters‐Regehr T, Bode JG, Kubitz R, Haussinger D. Involvement of CD95 (Apo‐1/Fas) ligand expressed by rat Kupffer cells in hepatic immunoregulation. Gastroenterol 116: 666‐677, 1999.
 597.Muthusamy V, Bosenberg M, Wajapeyee N. Redefining regulation of DNA methylation by RNA interference. Genomics 96: 191‐198, 2010.
 598.Nakahira K, Hisata S, Choi AM. The roles of mitochondrial damage‐associated molecular patterns in diseases. Antioxid Redox Signal 23: 1329‐1350, 2015.
 599.Nakano Y, Kondo T, Matsuo R, Hashimoto I, Kawasaki T, Kohno K, Myronovych A, Tadano S, Hisakura K, Ikeda O, Watganabe M, Murata S, Fukunaga K, Ohkohchi N. Platelet dynamics in the early phase of postischemic liver in vivo. J Surg Res 149: 192‐198, 2008.
 600.Nardone G, Compare D, Liguori E, Di Mauro V, Rocco A, Barone M, Napoli A, Lapi D, Iovene MR, Colantuoni A. Protective effects of Lactobacillus paracasei F19 in a rat model of oxidative and metabolic hepatic injury. Am J Physiol Gastrointest Liver Physiol 299: G669‐G676, 2010.
 601.Netticadan T, Temsah R, Osada M, Dhalla NS. Status of Ca2+/calmodulin protein kinase phosphorylation of cardiac SR proteins in ischemia‐reperfusion. Am J Physiol 277: C384‐C391, 1999.
 602.Neuhof C, Neuhof H. Calpain system and its involvement in myocardial ischemia and reperfusion injury. World J Cardiol 6: 638‐652, 2014.
 603.Ngoh GA, Facundo HT, Hamid T, Dillmann W, Zachara NE, Jones SP. Unique hexosaminidase reduces metabolic survival signal and sensitizes cardiac myocytes to hypoxia/reoxygenation injury. Circ Res 104: 41‐49, 2009.
 604.Niccoli G, Burzotta F, Galiuto L, Crea F. Myocardial no‐reflow in humans. J Am Coll Cardiol 54: 281‐292, 2009.
 605.Niccoli G, Cosentino N, Spaziani C, Minelli S, Fracassi F, Crea F. New strategies for the management of no‐reflow after primary percutaneous coronary intervention. Expert Rev Cardiovasc Ther 9: 615‐630, 2011.
 606.Nielsen M, Zimmer J, Diemer NH. Endonuclease G expression in thalamic reticular nucleus after global cerebral ischemia. Exp Brain Res 190: 81‐89, 2008.
 607.Nieswandt B, Pleines I, Bender M. Platelet adhesion and activation mechanisms in arterial thrombosis and ischaemic stroke. J Thromb Haemost 9(Suppl 1): 92‐104, 2011.
 608.Nilakantan V, Liang HL, Rajesh S, Mortensen J, Chandran K. Time‐dependant protective effects of mangenese(III) tetrakis (1‐methyl‐4‐pyridyl) porphyrin on mitochondrial function following renal ischemia‐reperfusion injury. Free Radic Res 44: 773‐782, 2010.
 609.Nordente A, Martorana GE, Miggianno GA, Petitti T, Giardina B, Littaru GP, Santini SA. Free radical production by activated haem proteins: Protective effect of coenzyme Q. Mol Aspects Med 15(Suppl): S109‐S115, 1994.
 610.Norman MU, Zbytnuik L, Kubes P. Interferon‐γ limits lymphocyte adhesion to inflamed endothelium: A nitric oxide regulatory feedback mechanism. Eur J Immunol 38: 1368‐1380, 2008.
 611.Northington FJ, Chavez‐Valdez R, Graham EM, Razdan S, Gauda EB, Martin LJ. Necrostatin decreases oxidative damage, inflammation, and injury after neonatal HI. J Cereb Blood Flow Metab 31: 178‐189, 2011.
 612.Nourshargh S, Alon R. Leukocyte migration into inflamed tissues. Immunity 41: 694‐707, 2014.
 613.Nourshargh S, Hordijk PL, Sixt M. Breaching multiple barriers: Leukocyte motility through venular walls and the interstitium. Nat Rev Mol Cell Biol 11: 366‐378, 2010.
 614.Nourshargh S, Renshaw SA, Imhof BA. Reverse migration of neutrophils: Where, when, how, and why? Trends Immunol 37: 273‐286, 2016.
 615.O'Donnell CJ, Nabel EG. Genomics of cardiovascular disease. N Engl J Med 365: 2098‐2109, 2011.
 616.O'Farrell FM, Attwell D. A role for pericytes in coronary no‐reflow. Nat Rev Cardiol 11: 427‐432, 2014.
 617.Oerlemans MI, Koudstaal S, Chamuleau SA, de Kleijn DP, Doevendans PA, Sluijter JP. Targeting cell death in the reperfused heart: Pharmacological approaches for cardioprotection. Int J Cardiol 165: 410‐422, 2013.
 618.Ojha S, Fainberg HP, Sebert S, Budge H, Symonds ME. Maternal health and eating habits: Metabolic consequences and impact on child health. Trends Mol Med 21: 126‐133, 2015.
 619.Okuno S, Saito A, Hayashi T, Chan PH. The c‐Jun N‐terminal protein kinase signaling pathway mediates Bax activation and subsequent neuronal apoptosis through interaction with Bim after transient focal cerebral ischemia. J Neurosci 24: 7879‐7887, 2004.
 620.Oliver MG, Specian RD, Perry MA, Granger DN. Morphologic assessment of leukocyte‐endothelial cell interactions in mesenteric venules subjected to ischemia and reperfusion. Inflammation 15: 331‐346, 1991.
 621.Ong SB, Gustafsson AB. New roles for mitochondria in cell death in the reperfused myocardium. Cardiovasc Res 94: 190‐193, 2012.
 622.Ong SB, Samangouei P, Kalkhoran SB, Hausenloy DJ. The mitochondrial permeability transition pore and its role in myocardial ischemia reperfusion injury. J Mol Cell Cardiol 78: 23‐34, 2015.
 623.Ong SB, Subrayan S, Lim SY, Yellon DM, Davidson SM, Hausenloy DJ. Inhibiting mitochondrial fission protects the heart against ischemia/reperfusion injury. Circulation 121: 2012‐2022, 2010.
 624.Ordy JM, Wengenack TM, Bialobok P, Coleman PD, Rodier P, Baggs RB, Dunlap WP, Kates B. Selective vulnerability and early progression of hippocampal CA1 pyramidal cell degeneration and GFAP‐positive astrocyte reactivity in the rat four‐vessel occlusion model of transient global ischemia. Exp Neurol 119: 128‐139, 1993.
 625.Orlic D, Kajstura J, Chimenti S, Jakoniuk I, Anderson SM, Li B, Pickel J, McKay R, Nadal‐Ginard B, Bodine DM, Leri A, Anversa P. Bone marrow cells regenerate infarcted myocardium. Nature 410: 701‐705, 2001.
 626.Orsini F, Moroni M, Contursi C, Yano M, Pelicci P, Giorgio M, Migliaccio E. Regulatory effects of the mitochondrial energetic status on mitochondrial p66Shc. Biol Chem 387: 1405‐1410, 2006.
 627.Osada M, Netticadan T, Kawabata K, Tamura K, Dhalla NS. Ischemic preconditioning prevents I/R‐induced alterations in SR calcium‐calmodulin protein kinase II. Am J Physiol 278: H1791‐H1798, 2000.
 628.Osman M, Russell J, Granger DN. Lymphocyte‐derived interferon‐γ mediates ischemia‐reperfusion‐induced leukocyte and platelet adhesion in intestinal microcirculation. Am J Physiol 296: G659‐G663, 2009.
 629.Ostadal B, Ostadalova I, Kolar F, Sedmera D. Developmental determinants of cardiac sensitivity to hypoxia. Can J Physiol Pharmacol 92: 566‐574, 2014.
 630.Ostadal B, Ostadalova I, Kolar F, Sedmera D. The use of microRNAs to modulate redox and immune response to stroke. Antioxid Redox Signal 22: 187‐202, 2015.
 631.OuyangOuyang YB, Stary CM, White RE, Giffard RG. Developmental determinants of cardiac sensitivity to hypoxia. Can J Physiol Pharmacol 92: 566‐574, 2015.
 632.Ovize M, Thibault H, Przyklenk K. Myocardial conditioning: Opportunities for clinical translation. Circ Res 113: 439‐450, 2013.
 633.Pacher P, Beckman JS, Liaudet L. Nitric oxide and peroxynitrite in health and disease. Physiol Rev 87: 315‐424, 2007.
 634.Palladini G, Ferrigno A, Richelmi P, Perlini S, Vairetti M. Role of matrix metalloproteinases in cholestasis and hepatic ischemia/reperfusion injury: A review. World J Gastroenterol 21: 12114‐12124, 2015.
 635.Pallast S, Arai K, Pekcec A, Yigitkanli K, Yu Z, Wang X, Lo EH, van Leyen K. Increased nuclear apoptosis‐inducing factor after transient focal ischemia: A 12/15‐lipoxygenase‐dependent organelle damage pathway. J Cereb Blood Flow Metab 30: 1157‐1167, 2010.
 636.Pallast S, Arai K, Wang X, Lo EH, van Leyen K. 12/15‐Lipoxygenase targets neuronal mitochondria under oxidative stress. J Neurochem 111: 882‐889, 2009.
 637.Paradies G, Petrosillo G, Paradies V, Ruggiero FM. Role of cardiolipin peroxidation and Ca2+ in mitochondrial dysfunction and disease. Cell Calcium 45: 643‐650, 2009.
 638.Parks DA, Bulkley GB, Granger DN, Hamilton SR, McCord JM. Ischemic injury in the cat small intestine: Role of superoxide radicals. Gastroenterology 82: 9‐15, 1982.
 639.Park P, Haas M, Cunningham PN, Bao L, Alexander JJ, Quigg RJ. Injury in renal ischemia‐reperfusion is independent from immunoglobulins and T‐lymphocytes. Am J Physiol 282: F352‐F357, 2002.
 640.Park PO, Haglund U. Regeneration of small bowel mucosa after intestinal ischemia. Crit Care Med 20: 135‐139, 1992.
 641.Park S, Kondo T, Nakano Y, Murata S, Fukunaga K, Oda T, Sasakik R, Ohkohchi N. Platelet adhesion in the sinusoid caused hepatic injury by neutrophils after hepatic ischemia reperfusion. Platelets 21: 282‐288, 2010.
 642.Parks DA, Granger DN. Xanthine oxidase: Biochemistry, distribution and physiology. Acta Physiol Scand 548: 87‐99, 1986.
 643.Patel B, Fisher M. Therapeutic advances in myocardial microvascular resistance: Unravelling the enigma. Pharmacol Ther 127: 131‐147, 2010.
 644.Patterson AJ, Chen M, Xue Q, Xiao D, Zhang L. Chronic prenatal hypoxia induces epigenetic programming of PKC{epsilon} gene repression in rat hearts. Circ Res 107: 365‐373, 2010.
 645.Patterson AJ, Zhang L. Hypoxia and fetal heart development. Curr Mol Med 10: 653‐666, 2010.
 646.Patterson AJ, Xiao D, Xiong F, Dixon B, Zhang L. Hypoxia‐derived oxidative stress mediates epigenetic repression of PKCϵ gene in foetal rat hearts. Cardiovasc Res 93: 302‐310, 2012.
 647.Pavlov VI, Tan YS, McClure EE, La Bonte LR, Zou C, Gorsuch WB, Stahl GL. Human mannose‐binding lectin inhibitor prevents myocardial injury and arterial thrombogenesis in a novel animal model. Am J Pathol 185: 347‐355, 2015.
 648.Pell VR, Chouchani ET, Murphy MP, Brookes PS, Krieg T. Moving forwards by blocking back‐flow: The Yin and Yang of MI Therapy. Circ Res 118: 898‐906, 2016.
 649.Peng S, Kuang Z, Zhang Y, Xu H, Cheng Q. The protective effects and potential mechanism of Calpain inhibitor Calpeptin against focal cerebral ischemia‐reperfusion injury in rats. Mol Biol Rep 38: 905‐912, 2011.
 650.Pepe S. Mitochondrial function in ischaemia and reperfusion of the ageing heart. Clin Exp Pharmacol Physiol 27: 745‐750, 2000.
 651.Peppiatt CM, Howarth C, Mobbs P, Attwell D. Bidirectional control of CNS capillary diameter by pericytes. Nature 443: 700‐704, 2006.
 652.Perez‐Chanona E, Muhlbauer M, Jobin C. The microbiota protects against ischemia/reperfusion‐induced intestinal injury through nucleotide‐binding oligomerization domain‐containing protein 2 (NOD2) signaling. Am J Pathol 184: 2965‐2975, 2014.
 653.Perrelli, M‐G, Pagliaro P, Penna C. Ischemia/reperfusion injury and cardioprotective mechanisms: Role of mitochondria and reactive oxygen species. World J Cardiol 3: 186‐200, 2011.
 654.Peters MJ, Dixon G, Kotowicz KT, Hatch DJ, Heyderman RS, Klein NJ. Circulating platelet‐neutrophil complexes represent a subpopulation of activated neutrophils primed for adhesion, phagocytosis and intracellular killing. Br J Haematol 106: 391‐399, 1999.
 655.Petrovic‐Djergovic D, Goonewardena SN, Pinsky DJ. Inflammatory disequilibrium in stroke. Circ Res 119: 142‐158, 2016.
 656.Phillips MJ, Voeltz GK. Structure and function of ER membrane contact sites with other organelles. Nat Rev Mol Cell Biol 17: 69‐82, 2016.
 657.Phillis JW, O'Regan MH. The role of phospholipases, cyclooxygenases, and lipoxygenases in cerebral ischemic/traumatic injuries. Crit Rev Neurobiol 15: 61‐90, 2003.
 658.Piao CS, Kim JB, Han PL, Lee JK. Administration of the p38 MAPK inhibitor SB203580 affords brain protection with a wide therapeutic window against focal ischemic insult. J Neurosci Res 73: 537‐544, 2003.
 659.Pigazzi A, Heydrick S, Folli F, Benoit S, Michelson A, Loscalzo J. Nitric oxide inhibits thrombin receptor‐activating peptide‐induced phosphoinositide 3‐kinase activity in human platelets. J Biol Chem 274: 14368‐14375, 1999.
 660.Pinto AR, Ilinykh A, Ivey MJ, Kuwabara JT, D'Antoni ML, Debuque R, Chandran A, Wang L, Arora K, Rosenthal NA, Tallquist ML. Revisiting cardiac cellular composition. Circ Res 118: 400‐409, 2016.
 661.Podestà MA, Cucchiari D, Ponticelli C. The diverging roles of dendritic cells in kidney allotransplantation. Transplant Rev 29: 114‐120, 2015.
 662.Prabhu SD, Frangogiannis NG. The biological basis for cardiac repair after myocardial infarction: From inflammation to fibrosis. Circ Res 119: 91‐112, 2016.
 663.Prakash A, Sundar SV, Zhu YG, Tran A, Lee JW, Lowell C, Hellman J. Lung ischemia‐reperfusion is a sterile inflammatory process influenced by commensal microbiota in mice. Shock 44: 272‐279, 2015.
 664.Premen AJ, Banchs V, Womack WA, Kvietys PR, Granger DN. Importance of collateral circulation in the vascularly occluded feline intestine. Gastroenterology 92: 1215‐1219, 1987.
 665.Pridjian AK, Levitsky S, Krukenkamp I, Silverman NA, Feinberg H. Developmental changes in reperfusion injury. A comparison of intracellular cation accumulation in the newborn, neonatal, and adult heart. J Thorac Cardiovasc Surg 93: 428‐433, 1987.
 666.Prime TA, Blaikie FH, Evans C, Nadtochiy SM, James AM, Dahm CC, Vitturi DA, Patel RP, Hiley CR, Abakumova I, Requejo R, Chouchani ET, Hurd TR, Garvey JF, Taylor CT, Brookes PS, Smith RA, Murphy MP. A mitochondria‐targeted S‐nitrosothiol modulates respiration, nitrosates thiols, and protects against ischemia‐reperfusion injury. Proc Natl Acad Sci 106: 10764‐10769, 2009.
 667.Proebstl D, Voisin MB, Woodfin A, Whiteford J, D'Acquisto F, Jones GE, Rowe D, Nourshargh S. Pericytes support neutrophil subendothelial cell crawling and breaching of venular walls in vivo. J Exp Med 209: 1219‐1234, 2012.
 668.Przyklenk K. Ischaemic conditioning: Pitfalls on the path to clinical translation. Br J Pharmacol 172: 1961‐1973, 2015.
 669.Puglisi RN, Strande L, Santos M, Schulte G, Hewitt CW, Whalen TV. Beneficial effects of cyclosporine and rapamycin in small bowel ischemic injury. J Surg Res 65: 115‐118, 1996.
 670.Qin B, Yang H, Xiao B. Role of microRNAs in endothelial inflammation and senescence. Mol Biol Rep 39: 4509‐4518, 2012.
 671.Quartara L, Maggi CA. The tachykinin NK1 receptor. Part II: Distribution and pathophysiological roles. Neuropeptides 32: 1‐49, 1998.
 672.Quesnelle KM, Bystrom PV, Toledo‐Pereyra LH. Molecular responses to ischemia and reperfusion in the liver. Arch Toxicol 89: 651‐657, 2015.
 673.Raedschelders K, Ansley DM, Chen DDY. The cellular and molecular origin of reactive oxygen species generation during myocardial ischemia and reperfusion. Pharmacol Therap 133: 230‐255, 2012.
 674.Rahimtoola SH. The hibernating myocardium. Am Heart J 117: 211‐221, 1989.
 675.Ramos G, Hofmann U, Frantz S. Myocardial fibrosis through the lenses of T‐cell biology. J Mol Cell Cardiol 92: 41‐45, 2016.
 676.Rautou PE, Vion AC, Amabile N, Chironi G, Simon A, Tedgui A, Boulanger CM. Microparticles, vascular function, and atherothrombosis. Circ Res 109: 593‐606, 2011.
 677.Ren M, Leng Y, Jeong M, Leeds PR, Chuang DM. Valproic acid reduces brain damage induced by transient focal cerebral ischemia in rats: Potential roles of histone deacetylase inhibition and heat shock protein induction. J Neurochem 89: 1358‐1367, 2004.
 678.Ren XP, Wu J, Wang X, Sartor MA, Qian J, Jones K, Nicolaou P, Pritchard TJ, Fan GC. MicroRNA‐320 is involved in the regulation of cardiac ischemia/reperfusion injury by targeting heat‐shock protein 20. Circulation 119: 2357‐2366, 2009.
 679.Ren Z, Jiang J, Lu H, Chen X, He Y, Zhang H, Xie H, Wang W, Zheng S, Zhou L. Intestinal microbial variation may predict early acute rejection after liver transplantation in rats. Transplantation 98: 844‐852, 2014.
 680.Reynaert NL, Guala AS, Wouters EF, van der Vliet A, Janssen‐Heininger YM. Nitric oxide represses inhibitory κB kinase through S‐nitrosylation. Proc Natl Acad Sci U S A 101: 8945‐8950, 2004.
 681.Reynolds RM. Corticosteroid‐mediated programming and the pathogenesis of obesity and diabetes. J Steroid Biochem Mol Biol 122: 3‐9, 2010.
 682.Riaz AA, Wan MX, Schaefer T, Schramm R, Ekberg H, Menger MD, Jeppsson B, Thorlacius H. Fundamental and distinct roles of P‐selectin and LFA‐1 in ischemia/reperfusion‐induced leukocyte‐endothelium interactions in the mouse colon. Ann Surg 236: 777‐784; discussion 784, 2002.
 683.Ribatti D. The crucial role of mast cells in blood‐brain barrier alterations. Exp Cell Res 338: 119‐125, 2015.
 684.Roberts VH, Frias AE, Grove KL. Impact of maternal obesity on fetal programming of cardiovascular disease. Physiology (Bethesda) 30(3): 224‐231, 2015.
 685.Rocha‐Ferreira E, Phillips E, Francesch‐Domenech E, Thei L, Peebles DM, Raivich G, Hristova M. The role of different strain backgrounds in bacterial endotoxin‐mediated sensitization to neonatal hypoxic‐ischemic brain damage. Neuroscience 311: 292‐307, 2015.
 686.Rodriguez SF, Granger DN. Role of blood cells in ischemia‐reperfusion‐induced endothelial barrier failure. Cardiovasc Res 87: 291‐299, 2010.
 687.Roe ND, Ren J. Nitric oxide synthase uncoupling: A therapeutic target in cardiovascular diseases. Vascul Pharmacol 57: 168‐172, 2012.
 688.Roerecke M, Rehm J. Alcohol consumption, drinking patterns, and ischemic heart disease: A narrative review of meta‐analyses and a systematic review and meta‐analysis of the impact of heavy drinking occasions on risk for moderate drinkers. BMC Med 12: 182, 2014.
 689.Rossaint J, Zarbock A. Platelets in leucocyte recruitment and function. Cardiovasc Res 107: 386‐395, 2015.
 690.Roy S, Khanna S, Hussain SR, Biswas S, Azad A, Rink C, Gnyawali S, Shilo, S, Nuovo GJ, Sen CK. MicroRNA expression in response to murine myocardial infarction: miR‐21 regulates fibroblast metalloprotease‐2 via phosphatase and tensin homologue. Cardiovasc Res 82: 21‐29, 2009.
 691.Romagnani S. Regulation of the T cell response. Clin Exptl Allergy 36: 1357‐1366, 2006.
 692.Romson JL, Hook BG, Kunkel SL, Abrams GD, Schork MA, Lucchesi BR. Reduction of the extent of ischemic myocardial injury by neutrophil depletion in the dog. Circulation 67: 1016‐1023, 1983.
 693.Rork TH, Hadzimichalis NM, Kappil MA, Merrill GF. Acetaminophen attenuates peroxynitrite‐activated matrix metalloprotease‐2‐mediated troponin I cleavage in the isolated guinea pig myocardium. J Mol Cell Cardiol 40: 553‐561, 2006.
 694.Rosen SE, Henry S, Bond R, Pearte C, Mieres JH. Sex‐specific disparities in risk factors for coronary heart disease. Curr Atheroscler Rep 17(8): 523, 2015.
 695.Rosenbaum DM, Degterev A, David J, Rosenbaum PS, Roth S, Grotta JC, Cuny GD, Yuan J, Savitz SI. Necroptosis, a novel form of caspase‐independent cell death, contributes to neuronal damage in a retinal ischemia‐reperfusion injury model. J Neurosci Res 88: 1569‐1576, 2010.
 696.Rossaint J, Zarbock A. Platelets in leucocyte recruitment and function. Cardiovasc Res 107: 386‐395, 2015.
 697.Rossen RD, Michael LH, Hawkins HK, Youker K, Dreyer WJ, Baughn RE, Entman ML. Cardiolipin‐protein complexes and initiation of complement activation after coronary artery occlusion. Circ Res 75: 546‐555, 1994.
 698.Rotter D, Grinsfelder DB, Parra V, Pedrozo Z, Singh S, Sachan N, Rothermel BA. Calcineurin and its regulator, RCAN1, confer time‐of‐day changes in susceptibility of the heart to ischemia/reperfusion. J Mol Cell Cardiol 74: 103‐111, 2014.
 699.Roy S, Khanna S, Hussain SR, Biswas S, Azad A, Rink C, Gnyawali S, Shilo, S, Nuovo GJ, Sen CK. MicroRNA expression in response to murine myocardial infarction: miR‐21 regulates fibroblast metalloprotease‐2 via phosphatase and tensin homologue. Cardiovasc Res 82: 21‐29, 2009.
 700.Rudolf V, Freeman BA. Cardiovascular consequences when nitric oxide and lipid signaling converge. Circ Res 105: 511‐512, 2009.
 701.Rueda‐Clausen CF, Morton JS, Davidge ST. Effects of hypoxia‐induced intrauterine growth restriction on cardiopulmonary structure and function during adulthood. Cardiovasc Res 81: 713‐722, 2009.
 702.Rueda‐Clausen CF, Morton JS, Lopaschuk GD, Davidge ST. Long‐term effects of intrauterine growth restriction on cardiac metabolism and susceptibility to ischaemia/reperfusion. Cardiovasc Res 90: 285‐294, 2011.
 703.Rueda‐Clausen CF, Morton JS, Dolinsky VW, Dyck JR, Davidge ST. Synergistic effects of prenatal hypoxia and postnatal high‐fat diet in the development of cardiovascular pathology in young rats. Am J Physiol Regul Integr Comp Physiol 303: R418‐R426, 2012.
 704.Rueda‐Clausen CF, Morton JS, Oudit GY, Kassiri Z, Jiang Y, Davidge ST. Effects of hypoxia‐induced intrauterine growth restriction on cardiac siderosis and oxidative stress. J Dev Orig Health Dis 3(5): 350‐357, 2012.
 705.Russell J, Cooper D, Tailor A, Stokes KY, Ganger DN. Low venular shear rates promote leukocyte‐dependent recruitment of adherent platelets. Am J Physiol 284: G123‐G129, 2003.
 706.Ryan KJ, Elmes MJ, Langley‐Evans SC. The effects of prenatal protein restriction on β‐adrenergic signalling of the adult rat heart during ischaemia reperfusion. J Nutr Metab 2012: 397389.
 707.Ryter SW, Alam J, Choi AMK. Heme oxygenase‐1/carbon monoxide: From basic science to therapeutic applications. Physiol Rev 86: 583‐650, 2006.
 708.Ryter SW, Mizumura K, Choi AM. The impact of autophagy on cell death modalities. Int J Cell Biol 2014;2014:502676. doi: 10.1155/2014/502676. Epub 2014 Feb 4.
 709.Saitoh M, Nishitoh H, Fuji M, Takeda K, Tobiume K, Sawada Y, Kawabata M, Miyazono K, Ichijo H. Mammalian thioredoxin is a direct inhibitor of apoptosis signal‐regulating kinase (ASK) 1. EMBO J 17: 2596‐2606, 1998.
 710.Salehi S, Reed EF. The divergent roles of macrophages in solid organ transplantation. Curr Opin Organ Transplant 20: 446‐453. 2015.
 711.Salim SY, Young PY, Lukowski CM, Madsen KL, Sis B, Churchill TA, Khadaroo RG. VSL#3 probiotics provide protection against acute intestinal ischaemia/reperfusion injury. Benef Microbes 4: 357‐365, 2013.
 712.Salter JW, Krieglstein CF, Isserkutz AC, Granger DN. Platelets modulate ischemia/reperfusion‐induced leukocyte recruitment in the mesenteric circulation. Am J Physiol 281: G1432‐G1439, 2001.
 713.Salvemini D, Cazzocrea S. Superoxide, superoxide dismutase and ischemic injury. Curr Opin Invest Drugs 3: 886‐895, 2002.
 714.Sanada S, Komuro I, Kitakaze M. Pathophysiology of myocardial reperfusion injury: Preconditioning, postconditioning, and translational aspects of protective measures. Am J Physiol 301: H1723‐H1741, 2011.
 715.Sansbury BE, Spite M. Resolution of acute inflammation and the role of resolvins in immunity, thrombosis, and vascular biology. Circ Res 119: 113‐130, 2016.
 716.Santén S, Mihaescu A, Laschke MW, Menger MD, Wang Y, Jeppsson B, Thorlacius H. p38 MAPK regulates ischemia‐reperfusion‐induced recruitment of leukocytes in the colon. Surgery 145: 303‐12, 2009.
 717.Santen S, Wang Y, Menger MD, Jeppsson B, Thorlacius H. Mast‐cell‐dependent secretion of CXC chemokines regulates ischemia‐reperfusion‐induced leukocyte recruitment in the colon. Int J Colorectal Dis 23: 527‐534, 2008.
 718.Santora RJ, Lie ML, Grigoryev DN, Nasir O, Moore FA, Hassoun HT. Therapeutic distant organ effects of regional hypothermia during mesenteric ischemia‐reperfusion injury. J Vasc Surg 52: 1003‐1014, 2010.
 719.Sapega AA, Heppenstall RB, Chance B, Park YS, Sokolow D. Optimizing tourniquet application and release times in extremity surgery. A biochemical and ultrastructural study. J Bone Joint Surg Am 67: 303‐314, 1985.
 720.Saugstad JA. Non‐coding RNAs in stroke and neuroprotection. Front Neurol 6: 50, 2015.
 721.Savas C, Ozogul C, Karaoz E, Delibas N, Ozguner F. Splenectomy reduces remote organ damage after intestinal ischaemia‐reperfusion injury. Acta Chir Belg 103: 315‐320, 2003.
 722.Saxton NE, Barclay JL, Clouston AD, Fawcett J. Cyclosporin A pretreatment in a rat model of warm ischaemia/reperfusion injury. J Hepatol 36: 241‐247, 2002.
 723.Sayed D, Abdellatif M. MicroRNAs in development and disease. Physiol Rev 91: 827‐887, 2011.
 724.Saztpute SR, Park JM, Jang HR, Agreda P, Liu M, Gandalfo MT, Racusen L, Rabb H. The role for T cell repertoire/antigen‐specific interactions in experimental kidney ischemia reperfusion injury. J Immunol 183: 984‐992, 2009.
 725.Schinzel AC, Takeuchi O, Huang Z, Fisher JK, Zhou Z, Rubens J, Hetz C, Danial NN, Moskowitz MA, Korsmeyer SJ. Cyclophilin D is a component of mitochondrial permeability transition and mediates neuronal cell death after focal cerebral ischemia. Proc Natl Acad Sci U S A 102: 12005‐12010, 2005.
 726.Schofield ZV, Woodruff TM, Halai R, Wu MC, Cooper MA. Neutrophils–a key component of ischemia‐reperfusion injury. Shock 40: 463‐470, 2013.
 727.Schroen B, Heymans S. Small but smart–microRNAs in the centre of inflammatory processes during cardiovascular diseases, the metabolic syndrome, and ageing. Cardiovasc Res 93: 605‐6013, 2012.
 728.Schulz C, Massberg S. Platelets in atherosclerosis and thrombosis. Handb Exp Pharmacol 2012: 111‐133.
 729.Schwartz BG, Kloner RA. Coronary no reflow. J Mol Cell Cardiol 52: 873‐882, 2012.
 730.Sciarretta S, Yee D, Ammann P, Nagarajan N, Volpe M, Frati G, Sadoshima J. Role of NADPH oxidase in the regulation of autophagy in cardiomyocytes. Clin Sci (Lond) 128: 387‐403, 2015.
 731.Sciarretta S, Zhai P, Shao D, Maejima Y, Robbins J, Volpe M, Condorelli G, Sadoshima J. Rheb is a critical regulator of autophagy during myocardial ischemia: Pathophysiological implications in obesity and metabolic syndrome. Circulation 125: 1134‐1146, 2012.
 732.Sciarretta S, Zhai P, Volpe M, Sadoshima J. Pharmacological modulation of autophagy during cardiac stress. J Cardiovasc Pharmacol 60: 235‐241, 2012.
 733.Scorrano L, Oakes SA, Opferman JT, Cheng EH, Sorcinelli MD, Pozzan T, Korsmeyer SJ. BAX and BAK regulation of endoplasmic reticulum Ca2+: A control point for apoptosis. Science 300: 135‐139, 2003.
 734.Seldin MM, Meng Y, Qi H, Zhu W, Wang Z, Hazen SL, Lusis AJ, Shih DM. Trimethylamine N‐oxide promotes vascular inflammation through signaling of mitogen‐activated protein kinase and nuclear factor‐kappaB. J Am Heart Assoc 5: pii: e002767, 2016.
 735.Sepramaniam S, Armugam A, Lim KY, Karolina DS, Swaminathan P, Tan JR, Jeyaseelan K. MicroRNA 320a functions as a novel endogenous modulator of aquaporins 1 and 4 as well as a potential therapeutic target in cerebral ischemia. J Biol Chem 285: 29223‐29230, 2010.
 736.Serino M, Blasco‐Baque V, Nicolas S, Burcelin R. Far from the eyes, close to the heart: Dysbiosis of gut microbiota and cardiovascular consequences. Curr Cardiol Rep 16: 540‐547, 2014.
 737.Seronde MF, Vausort M, Gayat E, Goretti E, Ng LL, Squire IB, Vodovar N, Sadoune M, Samuel JL, Thum T, Solal AC, Laribi S, Plaisance P, Wagner DR, Mebazaa A, Devaux Y. Circulating microRNAs and outcome in patients with acute heart failure. PloS One 10, e0142237, 2015.
 738.Sessa WC. Molecular control of blood flow and angiogenesis: Role of nitric oxide. J Thromb Haemost 7(Suppl 1): 35‐37, 2009.
 739.Sesti C, Simkhovich BZ, Kalvinsh I, Kloner RA. Mildronate, a novel fatty acid oxidation inhibitor and antianginal agent, reduces myocardial infarct size without affecting hemodynamics. J Cardiovasc Pharmacol 47: 493‐499, 2006.
 740.Settergren M, Böhm F, Malmström RE, Chancon KM, Pernow J. L‐arginine and tetrahydrobiopterin protects against ischemia/reperfusion‐induced endothelial dysfunction in patients with type 2 diabetes mellitis and coronary artery disease. Atherosclerosis 204: 73‐78, 2009.
 741.Sharma AK, Laubach VE, Ramos SI, Zhao Y, Stukenborg G, Linden J, Kron IL, Yang Z. Adenosine A2A receptor activation on CD4 +T lymphocytes and neutrophils attenuates lung ischemia‐reperfusion injury. J Thorac Cardiovasc Surg 139: 474‐482, 2010.
 742.Shen X, Wang Y, Gao F, Ren F, Busuttil RW, Kupiec‐Weglinski JW, Zhai Y. CD4+ T cells promote tissue inflammation via CD40 signaling without de novo activation in a murine model of liver ischemia/reperfusion injury. Hepatology 50: 1537‐1546, 2009.
 743.Shen XD, Ke B, Zhai Y, Gao F, Anselmo D, Lassman CR, Busuttil RW, Kupiec‐Weglinski JC. Stat4 and Stat6 signaling in hepatic ischemia/reperfusion injury in mice: HO‐1 dependence of Stat4 disruption‐mediated cytoprotection. Hepatology 37: 269‐303, 2003.
 744.Sheu EG, Oakes SM, Ahmadi‐Yazdi C, Afnan J, Carroll MC, Moore FD. Restoration of skeletal muscle ischemia‐reperfusion injury in humanized immunodeficient mice. Surgery 146: 340‐346, 2009.
 745.Shi J, Fujiedo H, Kokubo Y, Wake K. Apoptosis of neutrophils and their elimination by Kupffer cells in rat liver. Hepatology 24: 1256‐1263, 1996.
 746.Shi J, Gilbert GE, Kokubo Y, Ohashi T. Role of the liver in regulating numbers of circulating neutrophils. Blood 98: 1226‐1230, 2001.
 747.Shi Y, Melnikov VY, Schrier RW, Edelstein CL. Downregulation of the calpain inhibitor protein calpastatin by caspases during renal ischemia‐reperfusion. Am J Physiol Renal Physiol 279: F509‐F517, 2000.
 748.Shichita T, Sugiyama Y, Ooboshi H, Sugimori H, Nakagawa R, Takeda I, Iwaki T, Okada Y, Iida M, Cua OJ, Iwakura Y, Yoshimura A. Pivotal role of cerebral interleukin‐17‐producing γδT cells in the delayed phase of ischemic brain injury. Nat Med 15: 946‐951, 2009.
 749.Shimamura K, Kawamura H, Nagara T, Kato T, Naito T, Kameyama H, Hatakeyama K, Abo T. Association of NKT cells and granulocytes with liver injury after reperfusion of the portal vein. Cell Immunol 234: 31‐38, 2005.
 750.Shinagawa H, Frantz S. Cellular immunity and cardiac remodeling after myocardial infarction: Role of neutrophils, monocytes, and macrophages. Curr Heart Fail Rep 12: 247‐254, 2015.
 751.Shiotani S, Shimada M, Taketomi A, Soejima Y, Yoshizumi T, Hashimoto K, Shimokawa H, Maehara Y. Rho‐kinase as a novel gene therapeutic target in treatment of cold ischemia/reperfusion‐induced acute lethal liver injury: Effect on hepatocellular NADPH oxidase system. Gene Ther 14: 1425‐1433, 2007.
 752.Shiva S, Sack MN, Greer JJ, Duranski M, Ringwoood LA, Burwell L, Wang X, MacArthur PH, Shoja A, Raghavachari N, Calvert JW, Brookes PS, Lefer DJ, Gladwin MT. Nitrite augments tolerance to ischemia/reperfusion injury via the modulation of mitochondrial electron transfer. J Exp Med 204: 2089‐2102, 2007.
 753.Sies H. Oxidative stress: Introductory remarks. In: Sies H, editor. Oxidative Stress. London: Academic Press, 1985, pp. 1‐8.
 754.Simkhovich BZ, Przyklenk K, and Kloner RA. Role of protein kinase C in ischemic “conditioning”: From first evidence to current perspectives. J Cardiovasc Pharmacol Ther 18: 525‐532, 2013.
 755.Singbartl K, Forlow SB, Ley K. Platelet, but not endothelial, P‐selectin is critical for neutrophil‐mediated acute postischemic renal failure. FASEB J 15: 2337‐2344, 2001.
 756.Singh D, Chander V, Chopra K. Cyclosporine protects against ischemia/reperfusion injury in rat kidneys. Toxicology 207: 339‐347, 2005.
 757.Sitailo LA, Tibudan SS, Denning MF. Bax activation and induction of apoptosis in human keratinocytes by the protein kinase C delta catalytic domain. J Invest Dermatol 123: 434‐443, 2004.
 758.Slezak J, Tribulova N, Okruhlicova L, Dhingra R, Bajaj A, Freed D, Singal P. Hibernating myocardium: Pathophysiology, diagnosis, and treatment. Can J Physiol Pharmacol 87: 252‐265, 2009.
 759.Smith CC, Davidson SM, Lim SY, Simpkin JC, Hothersall JS, Yellon DM. Necrostatin: A potentially novel cardioprotective agent? Cardiovasc Drugs Ther 21: 227‐233, 2007.
 760.Smith CC, Yellon DM. Necroptosis, necrostatins and tissue injury. J Cell Mol Med 15: 1797‐1806, 2011.
 761.Smith VA, Johnson T. Evaluation of an animal product‐free variant of MegaCell MEM as a storage medium for corneas destined for transplantation. Ophthalmic Res 43: 33‐42, 2010.
 762.Solaini G, Harris DA. Biochemical dysfunction in heart mitochondria exposed to ischaemia and reperfusion. Biochem J 390: 377‐394, 2005.
 763.Song MA, Paradis AN, Gay MS, Shin J, Zhang L. Differential expression of microRNAs in ischemic heart disease. Drug Disc Today 20: 223‐235, 2015.
 764.Song L, Yang H, Wang HX, Tian C, Liu Y, Zeng XJ, Gao E, Kang YM, Du J, Li HH. Inhibition of 12/15 lipoxygenase by baicalein reduces myocardial ischemia/reperfusion injury via modulation of multiple signaling pathways. Apoptosis 19: 567‐580, 2014.
 765.Sorimachi Y, Harada K, Saido TC, Ono T, Kawashima S, Yoshida K. Downregulation of calpastatin in rat heart after brief ischemia and reperfusion. J Biochem 122: 743‐748, 1997.
 766.Sorkine P, Szold O, Halpern P, Gutman M, Greemland M, Rudick V, Goldman G. Gut decontamination reduces bowel ischemia‐induced lung injury in rats. Chest 112: 491‐495, 1997.
 767.Souza DG, Mendonça VA, de A Castro MS, Poole S, Teixeira MM. Role of tachykinin NK receptors on the local and remote injuries following ischaemia and reperfusion of the superior mesenteric artery in the rat. Br J Pharmacol 135: 303‐312, 2002.
 768.Souza DG, Vieira AT, Soares AC, Pinho V, Nicoli JR, Vieira LQ, Teixeira MM. The essential role of the intestinal microbiota in facilitating acute inflammatory responses. J Immunol 173: 4137‐4146, 2004.
 769.Sozen E, Karademir B, Ozer NK. Basic mechanisms in endoplasmic reticulum stress and relation to cardiovascular diseases. Free Radic Biol Med 78: 30‐41, 2015.
 770.Spescha RD, Klohs J, Semerano A, Giacalone G, Derungs RS, Reiner MF, Rodriguez Gutierrez D, Mendez‐Carmona N, Glanzmann M, Savarese G, Krankel N, Akhmedov A, Keller S, Mocharla P, Kaufmann MR, Wenger RH, Vogel J, Kulic L, Nitsch RM, Beer JH, Peruzzotti‐Jametti L, Sessa M, Luscher TF, Camici GG. Post‐ischaemic silencing of p66Shc reduces ischaemia/reperfusion brain injury and its expression correlates to clinical outcome in stroke. Eur Heart J 36: 1590‐1600, 2015.
 771.Spescha RD, Shi Y, Wegener S, Keller S, Weber B, Wyss MM, Lauinger N, Tabatabai G, Paneni F, Cosentino F, Hock C, Weller M, Nitsch RM, Luscher TF, Camici GG. Deletion of the ageing gene p66(Shc) reduces early stroke size following ischaemia/reperfusion brain injury. Eur Heart J 34: 96‐103, 2013.
 772.Stahl GL, Xu Y, Hao L, Miller M, Buras JA, Fung M, Zhao H. Role for the alternative complement pathway in ischemia/reperfusion injury. Am J Pathol 162: 449‐455, 2003.
 773.Stallion A, Kou TD, Latfi SQ, Miller KA, Dahms BB, Dudgeon DL, Levine AD. Ischemia/reperfusion: A clinically relevant model of intestinal injury yielding systemic inflammation. J Pediatr Surg 40: 470‐477, 2005.
 774.Stark K, Eckart A, Haidari S, Tirniceriu A, Lorenz M, von Bruhl ML, Gartner F, Khandoga AG, Legate KR, Pless R, Hepper I, Lauber K, Walzog B, Massberg S. Capillary and arteriolar pericytes attract innate leukocytes exiting through venules and ‘instruct' them with pattern‐recognition and motility programs. Nat Immunol 14: 41‐51, 2013.
 775.Stefanidakis M, Newton G, Lee WY, Parkos CA, Luscinskas FW. Endothelial CD47 interaction with SIRPγ is required for human T‐cell transendothelial migration under shear flow conditions in vitro. Blood 112: 1280‐1289, 2008.
 776.Stocker R, Yamamoto Y, McDonagh A, Glazer A, Ames BN. Bilirubin is an antioxidant of possible physiological importance. Science 235: 1043‐1045, 1987.
 777.Stokes KY, Granger DN. Platelets: A critical link between inflammation and microvascular dysfunction. J Physiol 590: 1023‐1034, 2012.
 778.Stowe DF, Camara KS. Mitochondrial reactive oxygen species production in excitable cells: Modulators of mitochondrial and cell function. Antiox Redox Signal 11: 1373‐1414, 2009.
 779.Strasser RH, Simonis G, Schön SP, Braun MU, Ihl‐Vahl R, Weinbrenner C, Marquetant R, Kübler W. Two distinct mechanisms mediate a differential regulation of protein kinase C isozymes in acute and prolonged myocardial ischemia. Circ Res 85: 77‐87, 1999.
 780.Strbian D, Karailainen‐Lindsberg ML, Kovanen PT, Tatlisumak T, Lindsberg PJ. Mast cell stabilization reduces hemorrhage formation and mortality after administration of thrombolytics in experimental ischemic stroke. Circulation 116: 411‐418, 2007.
 781.Strbian D, Karialainen‐Lindsberg ML, Tatlisumak T, Lindsberg PJ. Cerebral mast cells regulate early ischemic brain swelling and neutrophil accumulation. J Cereb Blood Flow Metab 26: 605‐612, 2006.
 782.Strbian D, Kovanen PT, Karajalainen‐Lindsberg ML, Tatlisumak T, Lindsberg PJ. An emerging role of mast cells in cerebral ischemia and hemorrhage. Annals Med 41: 438‐450, 2009.
 783.Strock PE, Majno G. Vascular responses to experimental tourniquet ischemia. Surg Gynecol Obstet 129: 309‐318. 1969.
 784.Suematsu M, Ishimura Y. The heme oxygenase‐carbon monoxide system: A regulator of hepatobiliary functions. Hepatology 31: 3‐6, 2000.
 785.Summers WK, Jamison RL. The no reflow phenomenon in renal ischemia. Lab Invest 25: 635‐643, 1974.
 786.Sun J, Murphy E. Protein s‐nitrosylation and cardioprotection. Circ Res 106: 285‐296, 2010.
 787.Sun J, Wu Q, Sun H, Qiao Y. Inhibition of histone deacetylase by butyrate protects rat liver from ischemic reperfusion injury. Int J Mol Sci 15: 21069‐21079, 2014.
 788.Sutherland BA, Rahman RMA, Apleton I. Mechanisms of action of green tea catechins, with a focus on ischemia‐induced neurodegeneration. J Nutr Biochem 17: 291‐306, 2006.
 789.Swank GM, Deitch EA. Role of the gut in multiple organ failure: Bacterial translocation and permeability changes. World J Surg 20: 411‐417, 1996.
 790.Symonds ME, Pope M, Sharkey D, Budge H. Adipose tissue and fetal programming. Diabetologia 55: 1597‐1606, 2012.
 791.Syrjälä SO, Keränen MA, Tuuminen R, Nykänen AI, Tammi M, Krebs R, Lemström KB. Increased Th17 rather than Th1 alloimmune response is associated with cardiac allograft vasculopathy after hypothermic presentation in the rat. J Heart Lung Transplant 29: 1047‐1057, 2010.
 792.Szydlowska K, Tymianski M. Calcium, ischemia and excitotoxicity. Cell Calcium 47: 122‐129, 2010.
 793.Tadimalla A, Belmont PJ, Thuerauf DJ, Glassy MS, Martindale JJ, Gude N, Sussman MA, Glembotski CC. Mesencephalic astrocyte‐derived neurotrophic factor is an ischemia‐inducible secreted endoplasmic reticulum stress response protein in the heart. Circ Res 103: 1249‐1258, 2008.
 794.Tailor A, Cooper D, Granger DN. Platelet‐vessel wall interactions in the microcirculation. Microcirc 12: 275‐285, 2005.
 795.Takagi H, Matsui Y, Hirotani S, Sakoda H, Asano T, Sadoshima J. AMPK mediates autophagy during myocardial ischemia in vivo. Autophagy 3: 405‐407, 2007.
 796.Takagi Y, Nozaki K, Sugino T, Hattori I, Hashimoto N. Phosphorylation of c‐Jun NH(2)‐terminal kinase and p38 mitogen‐activated protein kinase after transient forebrain ischemia in mice. Neurosci Lett 294: 117‐120, 2000.
 797.Takahashi M. NLRP3 in myocardial ischaemia‐reperfusion injury: Inflammasome‐dependent or ‐independent role in different cell types. Cardiovasc Res 99: 4‐5, 2013.
 798.Talukdar HA, Foroughi Asl H, Jain RK, Ermel R, Ruusalepp A, Franzen O, Kidd BA, Readhead B, Giannarelli C, Kovacic JC, Ivert T, Dudley JT, Civelek M, Lusis AJ, Schadt EE, Skogsberg J, Michoel T, Bjorkegren JL. Cross‐tissue regulatory gene networks in coronary artery disease. Cell Systems 2: 196‐208, 2016.
 799.Talukder MA, Zweier JL, Periasamy M. Targeting calcium transport in ischaemic heart disease. Cardiovasc Res 84: 345‐352, 2009.
 800.Tan JR, Koo YX, Kaur P, Liu F, Armugam A, Wong PT, Jeyaseelan K. microRNAs in stroke pathogenesis. Curr Mol Med 11: 76‐92, 2011.
 801.Tan L, Yu JT, Guan HS, Reservatrol exerts pharmacological preconditioning by activating PGC‐1alpha. Med Hypoth 71: 664‐667, 2008.
 802.Tankersley CG, Moldobaeva A, Wagner EM. Strain variation in response to lung ischemia: Role of MMP‐12. Respir Res 13: 93, 2012.
 803.Taylor MJ, Baicu SC. Current state of hypothermic machine perfusion preservation of organs: The clinical perspective. Cryobiology 60: S20‐35, 2010.
 804.Taylor PB, Young BG. Effect of myocardial ischemia on uridine incorporation and histone acetylation. Can J Physiol Pharmacol 60: 313‐318, 1982.
 805.Teoh NC, Ajamieh H, Wong HJ, Croft K, Mori T, Allison AC, Farrell GC. Microparticles mediate hepatic ischemia‐reperfusion injury and are the targets of Diannexin (ASP8597). PloS One 9: e104376, 2014.
 806.Terai K, Hiramoto Y, Masaki M, Sugiyama S, Kuroda T, Hori M, Kawase I, Hirota H. AMP‐activated protein kinase protects cardiomyocytes against hypoxic injury through attenuation of endoplasmic reticulum stress. Mol Cell Biol 25: 9554‐9575, 2005.
 807.Terao S, Yilmaz G, Stokes KY, Russell J, Ishikawa M, Kawase T, Granger DN. Blood cell‐derived RANTES mediates cerebral microvascular dysfunction, inflammation, and tissue injury after focal ischemia‐reperfusion. Stroke 39: 2560‐257, 2008.
 808.Tersteeg C, Heijnen HF, Eckly A, Pasterkamp G, Urbanus RT, Maas C, Hoefer IE, Nieuwland R, Farndale RW, Gachet C, de Groot PG, Roest M. FLow‐induced PRotrusions (FLIPRs): A platelet‐derived platform for the retrieval of microparticles by monocytes and neutrophils. Circ Res 114: 780‐791, 2014.
 809.Terui K, Enosawa S, Haga S, Zhang HQ, Kuroda H, Kouchi K, Matsunaga T, Yoshida H, Engelhardt JF, Irani K, Ohnuma N, Ozaki M. Stat3 confers resistance against hypoxia/reoxygenation‐induced oxidative injury in hepatocytes through upregulation of Mn‐SOD. J Hepatol 41: 957‐965, 2004.
 810.Theoharidis TC, Kempuraj D, Tagen M, Conti P, Kalogeromitros D. Differential relased mast cell mediators and the pathogenesis of inflammation. Immunol Rev 217: 65‐78, 2007.
 811.Theruvath TP, Snoddy MC, Zhong Z, Lemasters JJ. Mitochondrial permeability transition in liver ischemia and reperfusion: Role of c‐Jun N‐terminal kinase 2. Transplantation 85: 1500‐1504, 2008.
 812.Thomas A, Lenglet S, Chaurand P, Deglon J, Mangin P, Mach F, Steffens S, Wolfender JL, Staub C. Mass spectrometry for the evaluation of cardiovascular diseases based on proteomics and lipidomics. Thromb Haemost 106: 20‐33, 2011.
 813.Thomas WS, Mori E, Copeland BR, Yu JQ, Morrissey JH, del Zappo GJ. Tissue factor contributes to microvascular defects following cerebral ischemia. Stroke 24: 847‐853, 1993.
 814.Thon L, Möhlig H, Mathieu S, Lange A, Bulanova E, Winoto‐Morbach S, Schütze S, Bulfone‐Paus S, Adam D. Ceramide mediates caspase‐independent programmed cell death. FASEB J 19: 1945‐1956, 2005.
 815.Thornburg KL, O'Tierney PF, Louey S. Review: The placenta is a programming agent for cardiovascular disease. Placenta 31(Suppl): S54‐S59, 2010.
 816.Thuerauf DJ, Marcinko M, Gude N, Rubio M, Sussman MA, Glembotski CC. Activation of the unfolded protein response in infarcted mouse heart and hypoxic cultured cardiac myocytes. Circ Res 99: 275‐282, 2006.
 817.Tiefenbacher CP, Chilian WM, Mitchell M, Defily DV. Restoration of endothelium‐dependent vasodilation after reperfusion injury by tetrahydrobiopterin. Circ 94: 1423‐1429, 1996.
 818.Toko H, Takahashi H, Kayama Y, Okada S, Minamino T, Terasaki F, Kitaura Y, Komuro I. ATF6 is important under both pathological and physiological states in the heart. J Mol Cell Cardiol 49: 113‐120, 2010.
 819.Tomita S, Li RK, Weisel RD, Mickle DA, Kim EJ, Sakai T, Jia ZQ. Autologous transplantation of bone marrow cells improves damaged heart function. Circulation 100: II247‐256, 1999.
 820.Tomiyama K, Ikeda A, Ueki S, Nakao A, Stolz DB, Koike Y, Afrazi A, Gandhi C, Tokita D, Geller DA, Murase N. Inhibition of Kupffer cell‐mediated early proinflammatory response with carbon monoxide in transplant‐induced hepatic ischemia/reperfusion injury in rats. Hepatology 48: 1608‐1620, 2008.
 821.Toth A, Jeffers JR, Nickson P, Min JY, Morgan JP, Zambetti GP, Erhardt P. Targeted deletion of Puma attenuates cardiomyocyte death and improves cardiac function during ischemia‐reperfusion. Am J Physiol Heart Circ Physiol 291: H52‐60, 2006.
 822.Tracey KJ. Physiology and immunology of the cholinergic anti‐inflammatory pathway. J Clin Invest 117: 289‐296, 2007.
 823.Trinei M, Migliaccio E, Bernardi P, Paolucci F, Pelicci P, Giorgio M. p66Shc, mitochondria, and the generation of reactive oxygen species. Methods Enzymol 528: 99‐110, 2013.
 824.Tsubokawa T, Yamaguchi‐Okada M, Calvert JW, Solaroglu I, Shimamura N, Yata K, Zhang JH. Neurovascular and neuronal protection by E64d after focal cerebral ischemia in rats. J Neurosci Res 84: 832‐840, 2006.
 825.Tsukamoto T, Chanthaphavong RS, Pape HC. Current theories on the pathophysiology of multiple organ failure after trauma. Injury 41(1): 21‐26, 2010.
 826.Tsung A, Hoffman RA, Izuishi K, Critchlow ND, Nakao A, Chan MH, Lotze MT, Geller DA, Billiar TR. Hepatic ischemia‐reperfusion involves functional TLR4 signaling in nonparenchymal cells. J Immunol 175: 7661‐7668, 2005.
 827.Tsung A, Sahai R, Tanaka H, Takao A, Fink MP, Lotze MT, Yang H, Li J, Tracey KJ, Geller DA, Billiar TR. The nuclear factor HMGB1 mediates hepatic injury after liver ischemia‐reperfusion. J Exp Med 201: 1135‐1143, 2005.
 828.Tuboly E, Futakuchi M, Varga G, Erces D, Tokes T, Meszaros A, Kaszaki J, Suzui M, Imai M, Okada A, Okada N, Boros M, Okada H. C5a inhibitor protects against ischemia/reperfusion injury in rat small intestine. Microbiol Immunol 60: 35‐46, 2016.
 829.Turer AT, Hill JA. Pathogenesis of myocardial ischemia‐reperfusion injury and rationale for therapy. Am J Cardiol 106: 360‐368, 2016.
 830.Uchida Y, Ke B, Freitas MCS, Ji H, Zhao D, Benjamin ER, Najafsan N, Yagita H, Akiba H, Busuttil RW, Kupiec‐Weglinski JW. The emerging role of T cell immunoglobulin mucin‐1 in the mechanism of liver ischemia and reperfusion injury in the mouse. Hepatology 51: 1363‐1372, 2010.
 831.Uemura A, Naito Y, Matsubara T. Dynamics of Ca(2+)/calmodulin‐dependent protein kinase II following acute myocardial ischemia‐translocation and autophosphorylation. Biochem Biophys Res Commun 297: 997‐1002, 2002.
 832.Ufnal M, Zadlo A, Ostaszewski R. TMAO: A small molecule of great expectations. Nutrition 31: 1317‐1323, 2015.
 833.Ulven T. Short‐chain free fatty acid receptors FFA2/GPR43 and FFA3/GPR41 as new potential therapeutic targets. Front Endocrin 3: 111, 2012.
 834.Urbich C, Kuehbacher A, Dimmeler S. Role of microRNAs in vascular diseases, inflammation, and angiogenesis. Cardiovasc Res 79: 581‐588, 2008.
 835.Ussher JR, Lopaschuk GD, Arduini A. Gut microbiota metabolism of L‐carnitine and cardiovascular risk. Atherosclerosis 231: 456‐461, 2013.
 836.Vaghasiya JD, Sheth NR, Bhalodia YS, Jivani NP. Exaggerated liver injury produced by renal ischemia reperfusion in diabetes: Effect of exenatide. Saudi J Gastroenterol 16: 174‐180, 2010.
 837.Vakeva A, Meri S. Complement activation and regulator expression after anoxic injury of human endothelial cells. Acta Pathol Microbiol Immunol Scand 106: 1149‐1156, 1998.
 838.Valentim L, Laurence KM, Townsend PA, Carroll CJ, Soond S, Scarabelli TM, Knight RA, Latchman DS, Stephanou A. Urocortin inhibits Beclin1‐mediated autophagic cell death in cardiac myocytes exposed to ischaemia/reperfusion injury. J Mol Cell Cardiol 40: 846‐852, 2006.
 839.Valko M, Leibfritz D, Moncol J, Cronin MTD, Mazur M, Telser J. Free radicals and antioxidants in normal functions and human disease. Int J Biochem Cell Biol 39: 44‐84, 2007.
 840.van Ampting MT, Schonewille AJ, Vink C, Brummer RJ, van der Meer R, Bovee‐Oudenhoven IM. Damage to the intestinal epithelial barrier by antibiotic pretreatment of salmonella‐infected rats is lessened by dietary calcium or tannic acid. J Nutr 140: 2167‐2172, 2010.
 841.Vanden Berghe T, Vandenabeele P, Pasparakis M, Bleich M, Weinberg JM, Reichel CA, Brasen JH, Kunzendorf U, Anders HJ, Stockwell BR, Green DR. Synchronized renal tubular cell death involves ferroptosis. Proc Nat Acad Sci 111: 16836‐16841, 2014.
 842.Vander Heide RS, Steenbergen C. Cardioprotection and myocardial reperfusion: Pitfalls to clinical application. Circ Res 113: 464‐477, 2013.
 843.Van der Vusse GJ, Reneman RS, van Bilsen M. Accumulation of arachidonic acid in ischemic/reperfused cardiac tissue: Possible causes and consequences. Prostaglandins Leukot Essent Fatty Acids 57: 85‐93, 1997.
 844.van Leyen K, Kim HY, Lee SR, Jin G, Arai K, Lo EH. Baicalein and 12/15‐lipoxygenase in the ischemic brain. Stroke 37: 3014‐3018, 2006.
 845.Vandenabeele P, Declercq W, Van Herreweghe F, Vanden Berghe T. The role of the kinases RIP1 and RIP3 in TNF‐induced necrosis. Sci Signal 3(115): re4, 2010.
 846.Vega VL, Mardones L, Maldonado M, Nicovani S, Manriquez V, Roa J, Ward PH. Xanthine oxidase released from reperfused hind limbs mediate kupffer cell activation, neutrophil sequestration, and hepatic oxidative stress in rats subjected to tourniquet shock. Shock 14: 565‐571, 2000.
 847.Vickers MH. Developmental programming and transgenerational transmission of obesity. Ann Nutr Metab 64(Suppl 1): 26‐34, 2014.
 848.Vickers MH. Early life nutrition, epigenetics and programming of later life disease. Nutrients 6(6): 2165‐2178, 2014.
 849.Vila‐Petroff M, Salas MA, Said M, Valverde CA, Sapia L, Portiansky E, Hajjar RJ, Kranias EG, Mundiña‐Weilenmann C, Mattiazzi A. CaMKII inhibition protects against necrosis and apoptosis in irreversible ischemia‐reperfusion injury. Cardiovasc Res 73: 689‐698, 2007.
 850.Voisin MB, Nourshargh S. Neutrophil transmigration: Emergence of an adhesive cascade within venular walls. J Innate Immun 5: 336‐347, 2013.
 851.Vuohelainen V, Hamalainen M, Paavonen T, Karlsson S, Moilanen E, Mennander A. Inhibition of monoamine oxidase A increases recovery after experimental cardiac arrest. Interact Cardiovasc Thorac Surg 21: 441‐449, 2015.
 852.Wagers AJ, Conboy IM. Cellular and molecular signatures of muscle regeneration: Current concepts and controversies in adult myogenesis. Cell 122: 659‐667, 2005.
 853.Wagner S, Dybkova N, Rasenack EC, Jacobshagen C, Fabritz L, Kirchhof P, Maier SK, Zhang T, Hasenfuss G, Brown JH, Bers DM, Maier LS. Ca2+/calmodulin‐dependent protein kinase II regulates cardiac Na+ channels. J Clin Invest 116: 3127‐3138, 2006.
 854.Wagner S, Maier LS. Modulation of cardiac Na(+) and Ca(2+) currents by CaM and CaMKII. J Cardiovasc Electrophysiol 17(Suppl 1): S26‐S33, 2006.
 855.Wainwright CL, McCabe C, Kane KA. Endothelin and the ischaemic heart. Curr Vasc Pharmacol 3: 333‐341, 2005.
 856.Wall TM, Sheehy R, Hartman JC. Role of bradykinin in myocardial preconditioning. J Pharmacol Exp Ther 270: 681‐689, 1994.
 857.Walsh SR, Boyle JR, Tang TY, Sadat U, Cooper DG, Lapsley M, Norden AG, Varty K, Hayes PD, Gaunt ME. Remote ischemic preconditioning for renal and cardiac protection during endovascular aneurysm repair: A randomized controlled trial. J Endovasc Ther 16: 680‐689, 2009.
 858.Wang B, Huang Q, Zhang W, Li N, Li J. Lactobacillus plantarum prevents bacterial translocation in rats following ischemia and reperfusion injury. Dig Dis Sci 56: 3187‐3194, 2011.
 859.Wang H, Zhang W, Zuo L, Zhu W, Wang B, Li Q, Li J. Bifidobacteria may be beneficial to intestinal microbiota and reduction of bacterial translocation in mice following ischaemia and reperfusion injury. Br J Nutr 109: 1990‐1998, 2013.
 860.Wang K, Zhang J, Liu J, Tian J, Wu Y, Wang X, Quan L, Xu H, Wang W, Liu H. Variations in the protein level of Omi/HtrA2 in the heart of aged rats may contribute to the increased susceptibility of cardiomyocytes to ischemia/reperfusion injury and cell death: Omi/HtrA2 and aged heart injury. Age (Dordr) 35: 733‐746, 2013.
 861.Wang Y, Dawson VL, Dawson TM. Poly(ADP‐ribose) signals to mitochondrial AIF: A key event in parthanatos. Exp Neurol 218: 193‐202, 2009.
 862.Wang Y, Ji HX, Xing SH, Pei DS, Guan QH. SP600125, a selective JNK inhibitor, protects ischemic renal injury via suppressing the extrinsic pathways of apoptosis. Life Sci 80: 2067‐2075, 2007.
 863.Wang Z, Klipfell E, Bennett BJ, Koeth R, Levison BS, Dugar B, Feldstein AE, Britt EB, Fu X, Chung YM, Wu Y, Schauer P, Smith JD, Allayee H, Tang WH, DiDonato JA, Lusis AJ, Hazen SL. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature 472: 57‐63, 2011.
 864.Wang Z, Roberts AB, Buffa JA, Levison BS, Zhu W, Org E, Gu X, Huang Y, Zamanian‐Daryoush M, Culley MK, DiDonato AJ, Fu X, Hazen JE, Krajcik D, DiDonato JA, Lusis AJ, Hazen SL. Non‐lethal inhibition of gut microbial trimethylamine production for the treatment of atherosclerosis. Cell 163: 1585‐1595, 2015.
 865.Webb A, Bond R, McLean P, Uppal R, Benjamin N, Ahluwalia A. Reduction of nitrite to nitric oxide during ischemia protects against myocardial ischemia‐reperfusion damage. Proc Natl Acad Sci U S A 101: 13683‐13688, 2004.
 866.Webster KA. Mitochondrial membrane permeabilization and cell death during myocardial infarction: Roles of calcium and reactive oxygen species. Future Cardiol 8: 863‐884, 2012.
 867.Wegener S, Gottschalk B, Jovanovic V, Knab R, Fiebach JB, Schellinger PD, Kucinski T, Jungehulsing GJ, Brunecker P, Muller B, Banasik A, Amberger N, Wehrens XH, Lehnart SE, Reiken SR, Marks AR. Ca2+/calmodulin‐dependent protein kinase II phosphorylation regulates the cardiac ryanodine receptor. Circ Res 94: e61‐70, 2014.
 868.Wegener S, Gottschalk B, Jovanovic V, Knab R, Fiebach JB, Schellinger PD, Kucinski T, Jungehulsing GJ, Brunecker P, Muller B, Banasik A, Amberger N, Wernecke KD, Siebler M, Rother J, Villringer A, Weih M. Transient ischemic attacks before ischemic stroke: Preconditioning the human brain? A multicenter magnetic resonance imaging study. Stroke 35: 616‐621, 2004.
 869.Wehrens XH, Lehnart SE, Reiken SR, Marks AR. Ca2+/calmodulin‐dependent protein kinase II phosphorylation regulates the cardiac ryanodine receptor. Circ Res 94: e61‐70, 2004.
 870.Wei Q, Yin XM, Wang MH, Dong Z. Bid deficiency ameliorates ischemic renal failure and delays animal death in C57BL/6 mice. Am J Physiol Renal Physiol 290: F35‐42, 2006.
 871.Weinbroum AA, Hochhauser E, Rudick V, Kluger Y, Sorkine P, Karchevsky E, Graf E, Boher P, Flaishon R, Fjodorov D, Niv D, Vidne BA. Direct induction of acute lung and myocardial dysfunction by liver ischemia and reperfusion. J Trauma 43: 627‐633; discussion 633‐625, 1997.
 872.Weisman HF, Bartow T, Leppo MK, Boyle MP, Marsh HC, Jr., Carson GR, Roux KH, Weisfeldt ML, Fearon DT. Recombinant soluble CR1 suppressed complement activation, inflammation, and necrosis associated with reperfusion of ischemic myocardium. Trans Assoc Am Physicians 103: 64‐72, 1990.
 873.Welborn MB, III, Moldawer LL, Seeger JM, Minter RM, Huber TS. Role of endogenous interleukin‐10 in local and distant organ injury after visceral ischemia‐reperfusion. Shock 20: 35‐40, 2003.
 874.Wells JM, Gaggar A, Blalock JE. MMP generated matrikines. Matrix Biol 44‐46: 122‐129, 2015.
 875.Wershil BK, Wang ZS, Gordon JR, Galli SJ. Recruitment of neutrophils during IgE‐dependent cutaneous late phase reactions in the mouse is mast cell‐dependent. Partial inhibition of the reaction with antiserum against tumor necrosis factor‐alpha. J Clin Invest 87: 446‐453, 1991.
 876.West JD, Marnett LJ. Endogenous reactive intermediates as modulators of cell signaling and cell death. Chem Res Toxicol 19: 173‐194, 2006.
 877.Whelan RS, Kaplinskiy V, Kitsis RN. Cell death in the pathogenesis of heart disease: Mechanisms and significance. Annu Rev Physiol 72: 19‐44, 2010.
 878.Willems IE, Havenith MG, DeMey JG, Chaponnier C, Brown RA. The alpha‐smooth muscle actin‐positive cells in healing human myocardial scars. Am J Pathol 145: 868‐875, 1994.
 879.Williams JP, Pechet TT, Weiser MR, Reid R, Kobzik L, Moore FD, Jr., Carroll MC, Hechtman HB. Intestinal reperfusion injury is mediated by IgM and complement. J Appl Physiol 86: 938‐942, 1997.
 880.Winek K, Engel O, Koduah P, Heimesaat MM, Fischer A, Bereswill S, Dames C, Kershaw O, Gruber AD, Curato C, Oyama N, Meisel C, Meisel A, Dirnagl U. Depletion of cultivatable gut microbiota by broad‐spectrum antibiotic pretreatment worsens outcome after murine stroke. Stroke 47: 1354‐1363, 2016.
 881.Winek K, Meisel A, Dirnagl U. Gut microbiota impact on stroke outcome: Fad or fact? J Cereb Blood Flow Metab 36: 891‐898, 2016.
 882.Winquist RJ, Kerr S. Cerebral ischemia‐reperfusion injury and adhesion. Neurology 49: S23‐S26, 1997.
 883.Wittnich C. Age‐related differences in myocardial metabolism affects response to ischemia. Age in heart tolerance to ischemia. Am J Cardiovasc Pathol 4: 175‐180, 1992.
 884.Wolf PS, Merry HE, Farivar AS, McCourtie AS, Mulligan MS. Stress‐activated protein kinase inhibition to ameliorate lung ischemia reperfusion injury. J Thorac Cardiovasc Surg 135: 656‐665, 2008.
 885.Woodfin A, Voisin MB, Beyrau M, Colom B, Caille D, Diapouli FM, Nash GB, Chavakis T, Albelda SM, Rainger GE, Meda P, Imhof BA, Nourshargh S. The junctional adhesion molecule JAM‐C regulates polarized transendothelial migration of neutrophils in vivo. Nat Immunol 12: 761‐769, 2011.
 886.Wu B, Qiu W, Wang P, Yu H, Cheng T, Zambetti GP, Zhang L, Yu J. p53 independent induction of PUMA mediates intestinal apoptosis in response to ischaemia‐reperfusion. Gut 56: 645‐654, 2007.
 887.Wu SY, Tang SE, Ko FC, Wu GC, Huang KL, Chu SJ, Wu J, Huang Z, Ren J, Zhang Z, He P, Li Y, Ma J, Chen W, Zhang Y, Zhou X, Yang Z, Wu SQ, Chen L, Han J. Mlkl knockout mice demonstrate the indispensable role of Mlkl in necroptosis. Cell Res 23: 994‐1006, 2013.
 888.Wu SY, Tang SE, Ko FC, Wu GC, Huang KL, Chu SJ. Valproic acid attenuates acute lung injury induced by ischemia‐reperfusion in rats. Anesthesiology 122: 1327‐1337, 2015.
 889.Wung SF, Hickey KT, Taylor JY, Gallek MJ. Cardiovascular genomics. J Nurs 45: 60‐68, 2013.
 890.Xie LH, Chen F, Karagueusian HS, Weiss JN. Oxidative stress‐induced afterdepolarizations and calmodulin kinase II signaling. Circ Res 104: 79‐86, 2009.
 891.Xiong F, Lin T, Song M, Ma Q, Martinez SR, Lv J, MataGreenwood E, Xiao D, Xu Z, Zhang L. Antenatal hypoxia induces epigenetic repression of glucocorticoid receptor and promotes ischemic‐sensitive phenotype in the developing heart. J Mol Cell Cardiol 91: 160‐171, 2016.
 892.Xu CF, Yu CH, Li YM. Regulation of hepatic microRNA expression in response to ischemic preconditioning following ischemia/reperfusion injury in mice. OMICS 13: 513‐520, 2009.
 893.Xu H, Manivannan A, Jiang HR, Liversidge J, Shazrp PF, Forester JV, Crane IJ. Recruitment of IFN‐γ‐producing (Th1‐like) cells into the inflamed retina in vivo is preferentially regulated by p‐selectin glycoprotein ligand 1: P/E‐selectin interactions. J Immunol 172: 3215‐3224, 2004.
 894.Xu J, Zhao J, Evan G, Xiao C, Cheng Y, Xiao J. Circulating microRNAs: Novel biomarkers for cardiovascular diseases. J Mol Med 90: 865‐870, 2012.
 895.Xu X, Chua KW, Chua CC, Liu CF, Hamdy RC, Chua BH. Synergistic protective effects of humanin and necrostatin‐1 on hypoxia and ischemia/reperfusion injury. Brain Res 1355: 189‐194, 2010.
 896.Xu X, Zhang XA, Wang DW. The role of CYP450 epoxygenases and metabolites, epoxysatrienoic acids, cardiovascular and malignant diseases. Adv Drug Deliv Rev 63: 597‐608, 2011.
 897.Xu Y, Huang S, Liu ZG, Han J. Poly(ADP‐ribose) polymerase‐1 signaling to mitochondria in necrotic cell death requires RIP1/TRAF2‐mediated JNK1 activation. J Biol Chem 281, 8788‐8795, 2006.
 898.Xu Y, Williams SJ, O'Brien D, Davidge ST. Hypoxia or nutrient restriction during pregnancy in rats leads to progressive cardiac remodeling and impairs postischemic recovery in adult male offspring. FASEB J 20: 1251‐1253, 2006.
 899.Xu Z, Zhang J, David KK, Yang ZJ, Li X, Dawson TM, Dawson VL, Koehler RC. Endonuclease G does not play an obligatory role in poly(ADP‐ribose) polymerase‐dependent cell death after transient focal cerebral ischemia. Am J Physiol Regul Integr Comp Physiol 299: R215‐R221, 2010.
 900.Xue Q, Zhang L. Prenatal hypoxia causes a sex‐dependent increase in heart susceptibility to ischemia and reperfusion injury in adult male offspring: Role of protein kinase C epsilon. J Pharmacol Exp Ther 330: 624‐632, 2009.
 901.Xue Q, Dasgupta C, Chen M, Zhang L. Foetal hypoxia increases cardiac AT(2)R expression and subsequent vulnerability to adult ischaemic injury. Cardiovasc Res 89: 300‐308, 2011.
 902.Xue Q, Chen P, Li X, Zhang G, Patterson AJ, Luo J. Maternal high‐fat diet causes a sex‐dependent increase in AGTR2 expression and cardiac dysfunction in adult male rat offspring. Biol Reprod 93: 49, 2015.
 903.Yamashiro S, Noguchi K, Kuniyoshi Y, Koja K, Sakanashi M. Role of tetrahydrobiopterin on ischemia‐reperfusion injury in isolated injury in isolated perfused rat hearts. J Cardiovasc Surg (Torino) 44: 37‐49, 2003.
 904.Yang JS, Lai EC. Alternative miRNA biogenesis pathways and the interpretation of core miRNA pathway mutants. Mol Cell 43: 892‐903, 2011.
 905.Yan L, Yang H, Li Y, Duan H, Wu J, Qian P, Li B, Wang S. Regulator of calcineurin 1‐1L protects cardiomyocytes against hypoxia‐induced apoptosis via mitophagy. J Cardiovasc Pharmacol 64: 310‐317, 2014.
 906.Yang M, Stowe DF, Udoh KB, Heisner JS, Camara AK. Reversible blockade of complex I or inhibition of PKCbeta reduces activation and mitochondria translocation of p66Shc to preserve cardiac function after ischemia. PLoS One 9: e113534, 2014.
 907.Yang MQ, Ma YY, Ding J, Li JY. The role of mast cells in ischemia and reperfusion injury. Inflamm Res 63: 899‐905, 2014.
 908.Yang W, Guastella J, Huang JC, Wang Y, Zhang L, Xue D, Tran M, Woodward R, Kasibhatla S, Tseng B, Drewe J, Cai SX. MX1013, a dipeptide caspase inhibitor with potent in vivo antiapoptotic activity. Br J Pharmacol 140: 402‐412, 2003.
 909.Yang Y, Duan W, Li Y, Yan J, Yi W, Liang Z, Wang N, Yi D, Jin Z. New role of silent information regulator 1 in cerebral ischemia. Neurobiol Aging 34: 2879‐2888, 2013.
 910.Yang Y, Rosenberg GA. Matrix metalloproteinases as therapeutic targets for stroke. Brain Res 1623: 30‐38, 2015.
 911.Yang Z, Sharma AK, Linden J, Kron IL, Laubach VE. CD4+ T lymphocytes mediate acute pulmonary ischemia‐reperfusion injury. J Thorac Cardiovasc Surg 137: 695‐702, 2009.
 912.Yang ZJ, Carter EL, Kibler KK, Kwansa H, Crafa DA, Martin LJ, Roman RJ, Harder DR, Koehler RC. Attenuation of neonatal ischemic brain damage using a 20‐HETE synthesis inhibitor. J Neurochem 121: 168‐179, 2012.
 913.Yaoita H, Ogawa K, Maehara K, Maruyama Y. Attenuation of ischemia/reperfusion injury in rats by a caspase inhibitor. Circulation 97: 276‐281, 1998.
 914.Yasojima K, Kilgore KS, Washington RA, Lucchesi BR, McGeer PL. Complement gene expression by rabbit heart: Upregulation by ischemia and reperfusion. Circ Res 82: 1224‐1230, 1998.
 915.Ye Y, Hu Z, Lin Y, Zhang C, Perez‐Polo JR. Downregulation of microRNA‐29 by antisense inhibitors and a PPAR‐gamma agonist protects against myocardial ischaemia‐reperfusion injury. Cardiovasc Res 87: 535‐544, 2010.
 916.Ye Y, Perez‐Polo JR, Aguilar D, Birnbaum Y. The potential effects of anti‐diabetic medications on myocardial ischemia‐reperfusion injury. Basic Res Cardiol 106: 925‐952, 2011.
 917.Ye Y, Perez‐Polo JR, Qian J, Birnbaum Y. The role of microRNA in modulating myocardial ischemia‐reperfusion injury. Physiol Genomics 43: 534‐542, 2011.
 918.Yellon DM, Downey JM. Preconditioning the myocardium: From cellular physiology to clinical cardiology. Physiol Rev 83: 1113‐1151, 2003.
 919.Yellon DM, Hausenloy DJ. Myocardial reperfusion injury. N Engl J Med 357: 1121‐1135, 2007.
 920.Yemisci M, Gursoy‐Ozdemir Y, Vural A, Can A, Topalkara K, Dalkara T. Pericyte contraction induced by oxidative‐nitrosative stress impairs capillary reflow despite successful opening of an occluded cerebral artery. Nat Med 15: 1031‐1038, 2009.
 921.Yigitkanli K, Pekcec A, Karatas H, Pallast S, Mandeville E, Joshi N, Smirnova N, Gazaryan I, Ratan RR, Witztum JL, Montaner J, Holman TR, Lo EH, van Leyen K. Inhibition of 12/15‐lipoxygenase as therapeutic strategy to treat stroke. Ann Neurol 73: 129‐135, 2013.
 922.Yilmaz G, Granger DN. Cell adhesion molecules and ischemic stroke. Neurol Res 30: 783‐793, 2008.
 923.Yilmaz G, Granger DN. Cell adhesion molecules and ischemic stroke. Neurol Res 30: 783‐793, 2008.
 924.Yilmaz G, Granger DN. Leukocyte recruitment and ischemic brain injury. Neuromol Med 12: 193‐204, 2010.
 925.Yin KJ, Deng Z, Huang H, Hamblin M, Xie C, Zhang J, Chen YE. miR‐497 regulates neuronal death in mouse brain after transient focal cerebral ischemia. Neurobiol Dis 38: 17‐26, 2010.
 926.Yin KJ, Hamblin M, Chen YE. Non‐coding RNAs in cerebral endothelial pathophysiology: Emerging roles in stroke. Neurochem Int 77: 9‐16, 2014.
 927.Yokota N, Burne‐Taney M, Racusen L, Rabb H. Contrasting roles for STAT4 and STAT6 signal transduction pathways in murine renal ischemia‐reperfusion injury. Am J Physiol 285: F319‐F325, 2003.
 928.Yoshiya K, Lapchak PH, Thai TH, Kannan L, Rani P, Lucca JJ, Tsokos GC. Depletion of gut commensal bacteria attenuates intestinal ischemia/reperfusion injury. Am J Physiol Gastrointest Liver Physiol 301: G1020‐G1030, 2011.
 929.Youle RJ, Narendra DP. Mechanisms of mitophagy. Nat Rev Mol Cell Biol 12: 9‐14, 2011.
 930.Young GW, Wang Y, Ping P. Understanding proteasome assembly and regulation: Importance to cardiovascular medicine. Trends Cardiovasc Med 18: 93‐98, 2008.
 931.Yu X, Kem DC. Proteasome inhibition during myocardial infarction. Cardiovasc Res 85: 312‐320, 2010.
 932.Yuan Y, Wang JY, Xu LY, Cai R, Chen Z, Luo BY. MicroRNA expression changes in the hippocampi of rats subjected to global ischemia. J Clin Neurosci 17: 774‐778, 2010.
 933.Yusof M, Kamad K, Gaskin FS, Korthuis RJ. Angiotensin II mediates postischemic leukocyte‐endothelial interactions: Role of calcitonin gene‐related peptide. Am J Physiol Heart Circ Physiol 292: H3032‐3037, 2007.
 934.Yusof M, Kamada K, Kalogeris T, Gaskin FS, Korthuis RJ. Hydrogen sulfide triggers late‐phase preconditioning in postischemic small intestine by an NO‐ and p38 MAPK‐dependent mechanism. Am J Physiol Heart Circ Physiol 296: H868‐H876, 2009.
 935.Zaccagnini G, Martelli F, Fasanaro P, Magenta A, Gaetano C, Di Carlo A, Biglioli P, Giorgio M, Martin‐Padura I, Pelicci PG, Capogrossi MC. p66ShcA modulates tissue response to hindlimb ischemia. Circulation 109: 2917‐2923, 2004.
 936.Zaman AK, French CJ, Spees JL, Binbrek AS, Sobel BE. Vascular rhexis in mice subjected to non‐sustained myocardial ischemia and its therapeutic implications. Exp Biol Med (Maywood) 236: 598‐603, 2011.
 937.Zernecke A, Preissner KT. Extracellular ribonucleic acids (RNA) enter the stage in cardiovascular disease. Circ Res 118: 469‐479, 2016.
 938.Zhai P, Sciarretta S, Galeotti J, Volpe M, Sadoshima J. Differential roles of GSK‐3beta during myocardial ischemia and ischemia/reperfusion. Circ Res 109: 502‐511, 2011.
 939.Zhang C. MicroRNAs in vascular biology and vascular disease. J Cardiovasc Transl Res 3: 235‐240, 2010.
 940.Zhang M, Alicot EM, Carroll MC. Human natural IgM can induce ischemia/reperfusion injury in a murine intestinal model. Mol Immunol 44: 103‐110, 2008.
 941.Zhang M, Austen WG, Jr., Chiu I, Alicot EM, Hung R, Ma M, Verna N, Xu M, Hechtman HB, Moore FD, Jr., Carroll MC. Identification of a specific self‐reactive IgM antibody that initiates intestinal ischemia/reperfusion injury. Proc Nat Acad Sci 101: 3886‐3891, 2004.
 942.Zhang M, Carroll MC. Natural antibody mediated innate autoimmune response. Mol Immunol 44: 103‐110, 2007.
 943.Zhang ZG, Chopp M. Neurorestorative therapies for stroke: Underlying mechanisms and translation to the clinic. Lancet Neurol 8: 491‐500, 2009.
 944.Zhang Z, Liang D, Gao X, Zhao C, Qin X, Xu Y, Su T, Sun D, Li W, Wang H, Liu B, Cao F. Selective inhibition of inositol hexakisphosphate kinases (IP6Ks) enhances mesenchymal stem cell engraftment and improves therapeutic efficacy for myocardial infarction. Basic Res Cardiol 109: 417, 2014.
 945.Zhang Y, Ramos BF, Jakschik BA. Neutrophil recruitment by tumor necrosis factor from mast cells in immune complex peritonitis. Science 258: 1957‐1959, 1992.
 946.Zhang C, Wa J, Xu X, Potter BJ, Gao X. Direct relationship between levels of TNFα expression and endothelial dysfunction in reperfusion injury. Basic Res Cardiol 105: 453‐464, 2010.
 947.Zhang T, Zhang Y, Cui M, Jin L, Wang Y, Lv F, Liu Y, Zheng W, Shang H, Zhang J, Zhang M, Wu H, Guo J, Zhang X, Hu X, Cao CM, Xiao RP. CaMKII is a RIP3 substrate mediating ischemia‐ and oxidative stress‐induced myocardial necroptosis. Nat Med 22: 175‐182, 2016.
 948.Zhang XF, Zhang R, Huang L, Wang PX, Zhang Y, Jiang DS, Zhu LH, Tian S, Zhang XD, Li H. TRAF1 is a key mediator for hepatic ischemia/reperfusion injury. Cell Death Dis 5: e1467, 2014.
 949.Zhang Y, Zhao J, Lau WB, Jiao LY, Liu B, Yuan Y, Wang X, Gao E, Koch WJ, Ma XL, Wang Y. Tumor necrosis factor‐alpha and lymphotoxin‐alpha mediate myocardial ischemic injury via TNF receptor 1, but are cardioprotective when activating TNF receptor 2. PLoS One 8: e60227, 2013.
 950.Zhao BQ, Chauhan AK, Canau HM, Patten IS, Yang JJ, Dockal M, Scheiflinger F, Wagner DD. Von Willebrand factor‐cleaving protease ADAMTS13 reduces ischemic brain injury in experimental stroke. Blood 114: 3329‐3334, 2009.
 951.Zhao H, Ning J, Lemaire A, Koumpa FS, Sun JJ, Fung A, Gu J, Yi B, Lu K, Ma D. Necroptosis and parthanatos are involved in remote lung injury after receiving ischemic renal allografts in rats. Kidney Int 87: 738‐748, 2015.
 952.Zhao H, Ren C, Chen X, Shen J. From rapid to delayed and remote postconditioning: The evolving concept of ischemic postconditioning in brain ischemia. Curr Drug Targets 13: 173‐187, 2012.
 953.Zhao ZQ, Nakamura M, Wang NP, Velez DA, Hewan‐Lowe KO, Guyton RA, Vinten‐Johansen J. Dynamic progression of contractile and endothelial dysfunction and infarct extension in the late phase of reperfusion. J Surg Res 94: 133‐144, 2000.
 954.Zhao ZQ, Nakamura M, Wang NP, Wilcox JN, Shearer S, Ronson RS, Guyton RA, Vinten‐Johansen J. Reperfusion induces myocardial apoptotic cell death. Cardiovasc Res 45: 651‐660, 2000.
 955.Zheng SY, Fu XB, Xu JG, Zhao JY, Sun TZ, Chen W. Inhibition of p38 mitogen‐activated protein kinase may decrease intestinal epithelial cell apoptosis and improve intestinal epithelial barrier function after ischemia‐reperfusion injury. World J Gastroenterol 11: 656‐660, 2005.
 956.Zheng X, Zhang X, Sun H, Feng B, Li M, Chen G, Vladau C, Chen D, Suzuki M, Min L, Liu W, Zhong R, Garcia B, Jevnikar A, Min WP. Protection of renal ischemia injury using combination gene silencing of complement 3 and caspase 3 genes. Transplantation 82: 1781‐1786, 2006.
 957.Zhou Q, Lam PY, Han D, Cadenas E. c‐Jun N‐terminal kinase regulates mitochondrial bioenergetics by modulating pyruvate dehydrogenase activity in primary cortical neurons. J Neurochem 104: 325‐335, 2008.
 958.Zhu Q, Wani AA. Histone modifications: Crucial elements for damage response and chromatin restoration. J Cell Physiol 223: 283‐288, 2010.
 959.Zhu W, Gregory JC, Org E, Buffa JA, Gupta N, Wang Z, Li L, Fu X, Wu Y, Mehrabian M, Sartor RB, McIntyre TM, Silverstein RL, Tang WH, DiDonato JA, Brown JM, Lusis AJ, Hazen SL. Gut microbial metabolite TMAO enhances platelet hyperreactivity and thrombosis risk. Cell 165: 111‐124, 2016.
 960.Zhuang S, Demirs JT, Kochevar IE. p38 mitogen‐activated protein kinase mediates bid cleavage, mitochondrial dysfunction, and caspase‐3 activation during apoptosis induced by singlet oxygen but not by hydrogen peroxide. J Biol Chem 275: 25939‐25948, 2000.
 961.Zile MR, Mehurg SM, Arroyo JE, Stroud RE, DeSantis SM, Spinale FG. Relationship between the temporal profile of plasma microRNA and left ventricular remodeling in patients after myocardial infarction. Circ Cardiovasc Genet 4: 614‐619, 2011.
 962.Zuidema MY, Korthuis RJ. Intravital microscopic methods to evaluate anti‐inflammatory effects and signaling mechanisms evoked by hydrogen sulfide. Methods Enzymol 555: 93‐125, 2015.
 963.Zweier JL, Flaherty JT, Weisfeldt ML. Direct measurement of free radical generation following reperfusion of ischemic myocardium. Proc Natl Acad Sci U S A 84: 1404‐1407, 1987.

Contact Editor

Submit a note to the editor about this article by filling in the form below.

* Required Field

How to Cite

Theodore Kalogeris, Christopher P. Baines, Maike Krenz, Ronald J. Korthuis. Ischemia/Reperfusion. Compr Physiol 2016, 7: 113-170. doi: 10.1002/cphy.c160006