Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Anna K. Hsu is active.

Publication


Featured researches published by Anna K. Hsu.


Circulation Research | 1996

Morphine Mimics the Cardioprotective Effect of Ischemic Preconditioning via a Glibenclamide-Sensitive Mechanism in the Rat Heart

Jo El J. Schultz; Anna K. Hsu; Garrett J. Gross

Previous results from our laboratory have suggested that opioid receptors are involved in ischemic preconditioning (PC) in rat heart. Furthermore, other investigators have suggested that mu- and delta-opioid receptors mediate analgesia and hypoxic cerebral vasodilatation via opening of ATP-sensitive K+ (KATP) channels. Thus, the purpose of the present study was to test the hypothesis that activation of opioid receptors mimics the cardioprotective effect of ischemic PC and that this effect is produced by activation of KATP channels in the rat heart. Anesthetized open-chest Wistar rats were subjected to six different protocols. All groups were subjected to 30 minutes of occlusion and 2 hours of reperfusion. Ischemic PC was elicited by three 5-minute occlusion periods interspersed with 5 minutes of reperfusion. Similarly, morphine-induced PC was elicited by three 5-minute drug infusions (100 micrograms/kg i.v. ) interspersed with 5-minute drug-free periods before the prolonged 30-minute occlusion. Infarct size (IS) as a percentage of the area at risk (AAR) was determined by triphenyltetrazolium staining. Ischemic PC and morphine infusions resulted in similar reductions in IS/AAR from 56 +/- 5% to 11 +/- 3% and 12 +/- 5%, respectively (P < .05). Administration of glibenclamide (0.3 mg/kg i.v.), a KATP channel antagonist, or naloxone (3 mg/kg i.v.), a nonselective opioid receptor antagonist, both blocked the cardioprotective effects of morphine. These results indicate that opioid receptor stimulation results in a reduction in infarct size similar to that produced by ischemic PC. The effect of morphine is most likely mediated via an opioid receptor-KATP channel-linked mechanism in the rat heart, since glibenclamide abolished its protection.


Circulation Research | 2004

Opioid-Induced Cardioprotection Occurs via Glycogen Synthase Kinase β Inhibition During Reperfusion in Intact Rat Hearts

Eric R. Gross; Anna K. Hsu; Garrett J. Gross

Abstract— Glycogen synthase kinase (GSK) inhibition produced by ischemic preconditioning has been previously shown to be regulated through phosphatidylinositol-3 kinase (PI3K). Therefore, we determined whether opioid-induced cardioprotection (OIC) occurs during reperfusion by altering GSK phosphorylation through PI3K and target of rapamycin (TOR). Furthermore, we determined if selective GSK inhibitors, SB216763(SB21) or SB415286(SB41), emulate OIC. Rats were treated with the nonselective opioid agonist, morphine (MOR, 0.3 mg/kg), the δ-selective opioid agonist BW373U86 (BW, 1 mg/kg), or the GSK inhibitors, SB21 (0.6 mg/kg) or SB41(1.0 mg/kg), either 10 minutes before ischemia or 5 minutes before reperfusion. Five minutes before opioid or SB21 treatment, some rats received either the PI3K inhibitor wortmannin (15 μg/kg) or LY294002 (0.3 mg/kg) or the TOR inhibitor rapamycin (3 μg/kg). After 30 minutes of ischemia followed by 2 hours of reperfusion, infarct size was assessed. MOR, BW, SB41, and SB21 reduced infarct size compared with vehicle when administered before ischemia (42.9±2.6, 40.3±2.3, 46.6±1.6, 42.2±1.8 versus 60.0±1.1%, respectively; P <0.001) and showed similar protection when administered 5 minutes before reperfusion (43.6±2.3, 40.2±2.6, 44.8±2.8, 39.4±0.8%, respectively; P <0.001). Wortmannin, LY294002, and rapamycin were found to inhibit OIC; however, they did not abrogate SB21-induced infarct size reduction. At 5 minutes of reperfusion, both MOR and BW increased P-GSKβ at Ser9 in the ischemic zone compared with vehicle (181±20, 178±15 versus 75±17 DU, respectively; P <0.05), and this effect was abrogated by prior administration of wortmannin or rapamycin in MOR-treated rats. Furthermore, no differences were seen in phosphorylation of GSK&agr; (Ser21 or Tyr279) or phosphorylation of GSKβ (Tyr216). These data indicate that OIC occurs via the phosphorylation of GSKβ at Ser9 during reperfusion.


Circulation | 1998

Ischemic Preconditioning in the Intact Rat Heart Is Mediated by δ1- But Not μ- or κ-Opioid Receptors

Jo El J. Schultz; Anna K. Hsu; Garrett J. Gross

Background—Our laboratory has previously shown that δ-opioid receptors are involved in the cardioprotective effect of ischemic preconditioning in the rat heart. However, this class of receptors consists of two subtypes, δ1 and δ2, and μ- or κ-opioid receptors may also exist in the heart. Therefore, the purpose of the present study was to test the hypothesis that ischemic preconditioning is mediated through stimulation of one or both δ-opioid receptor subtypes. Methods and Results—Anesthetized, open chest, male Wistar rats were assigned to 1 of 14 groups. All animals were subjected to 30 minutes of occlusion and 2 hours of reperfusion. Ischemic preconditioning was elicited by three 5-minute occlusion periods interspersed with 5 minutes of reperfusion. Two doses of 7-benzylidenenaltrexone (BNTX; 1 and 3 mg/kg IV), a selective δ1-opioid receptor antagonist, or naltriben (NTB; 1 and 3 mg/kg IV), a selective δ2-opioid receptor antagonist, were given before ischemic preconditioning. To test for a role of μ-opio...


American Journal of Physiology-heart and Circulatory Physiology | 1998

TAN-67, a δ1-opioid receptor agonist, reduces infarct size via activation of Gi/o proteins and KATP channels

Jo El J. Schultz; Anna K. Hsu; Hiroshi Nagase; Garrett J. Gross

We have previously shown that delta (δ)-opioid receptors, most notably δ1, are involved in the cardioprotective effect of ischemic preconditioning (PC) in rats; however, the mechanism by which δ-opioid receptor-induced cardioprotection is mediated remains unknown. Therefore, we hypothesized that several of the known mediators of ischemic PC such as the ATP-sensitive potassium (KATP) channel and Gi/o proteins are involved in the cardioprotective effect produced by δ1-opioid receptor activation. To address these possibilities, anesthetized, open-chest Wistar rats were randomly assigned to five groups. Control animals were subjected to 30 min of coronary artery occlusion and 2 h of reperfusion. To demonstrate that stimulating δ1-opioid receptors produces cardioprotection, TAN-67, a new selective δ1-agonist, was infused for 15 min before the long occlusion and reperfusion periods. In addition, one group received 7-benzylidenenaltrexone (BNTX), a selective δ1-antagonist, before TAN-67. To study the involvement of KATPchannels or Gi/o proteins in δ1-opioid receptor-induced cardioprotection, glibenclamide (Glib), a KATP channel antagonist, or pertussis toxin (PTX), an inhibitor of Gi/o proteins, was administered before TAN-67. Infarct size (IS) as a percentage of the area at risk (IS/AAR) was determined by tetrazolium stain. TAN-67 significantly reduced IS/AAR as compared with control (56 ± 2 to 27 ± 5%, n = 5, P < 0.05). The cardioprotective effect of TAN-67 was completely abolished by BNTX, Glib, and PTX (51 ± 3, 53 ± 5, and 61 ± 4%, n = 6 for each group, respectively). These results are the first to suggest that stimulating the δ1-opioid receptor elicits a cardioprotective effect that is mediated via Gi/o proteins and KATP channels in the intact rat heart.We have previously shown that delta (delta)-opioid receptors, most notably delta 1, are involved in the cardioprotective effect of ischemic preconditioning (PC) in rats; however, the mechanism by which delta-opioid receptor-induced cardioprotection is mediated remains unknown. Therefore, we hypothesized that several of the known mediators of ischemic PC such as the ATP-sensitive potassium (KATP) channel and Gi/o proteins are involved in the cardioprotective effect produced by delta 1-opioid receptor activation. To address these possibilities, anesthetized, open-chest Wistar rats were randomly assigned to five groups. Control animals were subjected to 30 min of coronary artery occlusion and 2 h of reperfusion. To demonstrate that stimulating delta 1-opioid receptors produces cardioprotection, TAN-67, a new selective delta 1-agonist, was infused for 15 min before the long occlusion and reperfusion periods. In addition, one group received 7-benzylidenenaltrexone (BNTX), a selective delta 1-antagonist, before TAN-67. To study the involvement of KATP channels or Gi/o proteins in delta 1-opioid receptor-induced cardioprotection, glibenclamide (Glib), a KATP channel antagonist, or pertussis toxin (PTX), an inhibitor of Gi/o proteins, was administered before TAN-67. Infarct size (IS) as a percentage of the area at risk (IS/AAR) was determined by tetrazolium stain. TAN-67 significantly reduced IS/AAR as compared with control (56 +/- 2 to 27 +/- 5%, n = 5, P < 0.05). The cardioprotective effect of TAN-67 was completely abolished by BNTX, Glib, and PTX (51 +/- 3, 53 +/- 5, and 61 +/- 4%, n = 6 for each group, respectively). These results are the first to suggest that stimulating the delta 1-opioid receptor elicits a cardioprotective effect that is mediated via Gi/o proteins and KATP channels in the intact rat heart.


American Journal of Physiology-heart and Circulatory Physiology | 1999

Importance of PKC and tyrosine kinase in single or multiple cycles of preconditioning in rat hearts.

Ryan M. Fryer; Jo El J. Schultz; Anna K. Hsu; Garrett J. Gross

Both tyrosine kinase (TK) and protein kinase C (PKC) inhibitors have been shown individually to completely abolish the cardioprotective effects of ischemic preconditioning (IPC) in rabbits; however, blockade of both enzymes is necessary to totally abolish IPC in pigs. Recently, we have shown that TK inhibition partially attenuates the cardioprotective effect of IPC in intact rat hearts. Therefore, the present study was designed to test the hypothesis that inhibition of both TK and PKC is necessary to completely abolish IPC in the intact rat and that this effect is dependent on the intensity of the preconditioning stimulus. All animals were subjected to 30 min of coronary artery occlusion and 2 h of reperfusion. In series 1, multiple-cycle-induced IPC was produced via three 5-min occlusions interspersed with 5 min of reperfusion (3 × 5 IPC). Genistein (5 mg/kg), a TK inhibitor infused 30 min before IPC, and chelerythrine chloride (5 mg/kg), a PKC inhibitor infused 5 min before the prolonged ischemic insult, were administered alone or in combination in the absence or presence of 3 × 5 IPC. 3 × 5 IPC produced a marked reduction in infarct size as a percentage of area at risk compared with control (8.0 ± 0.8 vs. 56.1 ± 0.8%). The effects of 3 × 5 IPC were partially blocked by pretreatment with genistein (34.0 ± 2.0%) or chelerythrine (26.4 ± 2.8%) alone; however, combined administration of genistein and chelerythrine completely abolished the effects of 3 × 5 IPC (50.7 ± 3.6%). In series 2, single-cycle IPC was elicited by one 5-min occlusion followed by 10 min of reperfusion (1 × 5 IPC). Compared with control, 1 × 5 IPC also significantly reduced infarct size (15.4 ± 3.0%). Genistein or chelerythrine administered alone completely abolished 1 × 5 IPC-induced cardioprotection. These results suggest that the efficacy of TK and PKC inhibition to block IPC depends on the intensity of the preconditioning stimulus and that these kinases may work through parallel pathways.


Diabetes | 2007

Diabetes Abolishes Morphine-Induced Cardioprotection via Multiple Pathways Upstream of Glycogen Synthase Kinase-3β

Eric R. Gross; Anna K. Hsu; Garrett J. Gross

The cardioprotective effect of opioids or glycogen synthase kinase (GSK) inhibitors given at reperfusion has not been investigated in diabetes models. Therefore, nondiabetic (NDBR) or streptozotocin-induced diabetic (DBR) rat hearts were subjected to 30 min of ischemia and 2 h of reperfusion. Groups of NDBR or DBR were administered either vehicle, morphine (0.3 mg/kg), or the GSK inhibitor SB216763 (0.6 mg/kg) 5 min before reperfusion. SB216763 (but not morphine) reduced infarct size in DBRs (44 ± 1* and 55 ± 2%, respectively), while both agents reduced infarct size in NDBRs versus untreated NDBRs or DBRs (44 ± 3*, 42 ± 3*, 60 ± 2, and 56 ± 2%, respectively, *P < 0.001). Morphine-induced phospho- (P-)GSK3β was reduced 5 min after reperfusion in DBRs compared with NDBRs (0.83 ± 0.29 and 1.94 ± 0.12 [P < 0.05] pg/μg tissue, respectively). The GSK3β mediators, P-Akt, P–extracellular signal–related kinase (ERK)1, and P–signal transducer and activator of transcription (STAT)3, were also significantly reduced in untreated DBR compared with NDBR rats. Morphine-induced elevations of P-Akt, P-ERK1, P-p70s6, P–janus-activated kinase-2, and P-STAT3 in NDBRs were also blunted in DBRs. H9C2 cells raised in 25 mmol/l compared with 5.56 mmol/l glucose media also demonstrated reduced morphine-induced P-GSK3β, P-Akt, P-STAT3, and P-ERK1 after 15 min. Hence, acute GSK inhibition may provide a novel therapeutic strategy for diabetic patients during an acute myocardial infarction, whereas morphine is less effective due to signaling events that adversely affect GSK3β.


Basic Research in Cardiology | 2001

ERK and p38 MAP kinase activation are components of opioid-induced delayed cardioprotection

Ryan M. Fryer; Anna K. Hsu; Garrett J. Gross

Abstract Opioids have been previously shown to confer acute and delayed cardioprotection against a prolonged ischemic insult. We have extensively characterized the signal transduction pathway mediating acute cardioprotection and have suggested a role for extracellular signal regulated kinase (ERK) in this cardioprotection. Therefore, we attempted to determine a role for ERK and the stress activated MAP kinase, p38, in opioid-induced delayed cardioprotection by using selective inhibitors of these pathways. All rats were subjected to 30 min of ischemia and 2 h of reperfusion (I/R). Control animals, injected with saline 48 h prior to I/R, had an infarct size/area at risk (IS/AAR) of 61.6 ± 1.6. 48-h pretreatment with TAN-67 (30 mg/kg), a δ1-opioid receptor agonist, maximally reduced IS/AAR (31.2 ± 6.5). The involvement of ERK was examined with PD 098059, a selective pharmacological antagonist which inhibits the upstream kinase, MEK-1, that phosphorylates and activates ERK. PD 098059 (0.3 mg/kg) did not alter IS/AAR when administered alone (60.7 ± 4.9). However, PD 098059 (0.3 mg/kg) administration 30 min prior to TAN-67 (30 mg/kg) completely abolished cardioprotection (61.0 ± 7.6). The selective p38 inhibitor, SB 203580 (1.0 mg/kg), had no effect on IS/AAR in the absence of TAN-67 (53.1 ± 2.3). Additionally, SB 203580 (1.0 mg/kg) when administered prior to TAN-67 (30 mg/kg) partially abolished cardioprotection (51.3 ± 6.4). These results suggest that both ERK and p38 are integral components of opioid-induced delayed cardioprotection and may act via parallel pathways.


Basic Research in Cardiology | 2007

GSK3β inhibition and KATP channel opening mediate acute opioid-induced cardioprotection at reperfusion

Eric R. Gross; Anna K. Hsu; Garrett J. Gross

Both glycogen synthase kinase 3β (GSK3β) and the ATP-dependant potassium channel (KATP) mediate opioid-induced cardioprotection (OIC). However, whether direct KATP channel openers induce cardioprotection prior to reperfusion and their signaling cascade position with respect to GSK3β inhibition is unknown. Therefore, we investigated the role of KATP channel opening at reperfusion in OIC, and the interaction between the GSK signaling axis and KATP channels in cardioprotection.Male Sprague-Dawley rats underwent 30 minutes ischemia with 2 hours of reperfusion and infarct size was determined. Rats given the nonselective opioid agonist, morphine (0.3 mg/kg), or the selective delta opioid agonist, BW373U86 (1.0 mg/kg), 5 minutes prior to reperfusion reduced infarct size (40.3±1.6*, 39.7±1.9* versus 60.0±1.1%, respectively, * P<0.001%). This protection was abrogated with prior administration of the putative sarcolemmal KATP antagonist, HMR-1098 (6 mg/kg), or the putative mitochondrial KATP antagonist, 5-HD (10 mg/kg). The putative sKATP channel opener, P-1075 (1μg/kg) or the putative mKATP channel opener, BMS-191095 (1 mg/kg) given 5 minutes prior to reperfusion also reduced infarct size (41.8±2.4*, 43.4±1.4*) and protection was abrogated by prior administration of the PI3k inhibitor wortmannin (60.0±1.7, 64.0±2.6%, respectively, * P<0.001). Cardioprotection afforded by the GSK inhibitor SB216763 (0.6 mg/kg) given 5 minutes prior to reperfusion was also partially blocked by either HMR or 5-HD and completely blocked when HMR and 5-HD were given in combination (40.8±1.6*, 50.4±1.6^; 49.4±1.7^, 61.6±1.6%, respectively, * or ^ P<0.001). These data indicate that both the sKATP and mKATP channel are involved in acute OIC and the GSK signaling axis regulates cardioprotection via KATP channel opening.


Basic Research in Cardiology | 2001

Production and metabolism of ceramide in normal and ischemic-reperfused myocardium of rats.

David X. Zhang; Ryan M. Fryer; Anna K. Hsu; Ai-Ping Zou; Garrett J. Gross; William B. Campbell; Pin-Lan Li

Abstract Ceramide has been shown to be a key signaling molecule involved in the apoptotic effect of tumor necrosis factor α (TNF-α) and other cytokines. Given the importance of cytokines such as TNF-α in myocardial ischemia-reperfusion injury, we hypothesize that ceramide is increased during ischemia or reperfusion, and that the activity of enzymes responsible for its production or breakdown should be increased and/or decreased, respectively. Therefore, in the present study, we characterized the enzymatic activities responsible for ceramide production and metabolism in the myocardium of rats, and determined the contribution of these enzymes to altered ceramide levels during myocardial ischemia and reperfusion. The basal ceramide concentration in the myocardium of rats was 34.0 pmol/mg tissue. As determined by the conversion of 14C-sphingomyelin into ceramide and 14C-choline phosphate, both neutral (N-) and acidic (A-) SMase were detected in the myocardium, with a conversion rate of 0.09 ± 0.008 and 0.32 ± 0.05 nmol/min per mg protein, respectively. The activity of A-SMase (78 % of total cellular activity) was significantly higher in microsomes than in cytosol, while the activity of N-SMase was similar in both fractions. Ceramidase, a ceramide-metabolizing enzyme, was also detected in the myocardium of rats. It metabolized ceramide into sphingosine at a rate of 9.94 ± 0.42 pmol/min per mg protein. In anesthetized rats, 30 min of ischemia had no apparent effect on ceramide concentrations in the myocardium, while 30 min of ischemia followed by 3 h of reperfusion resulted in a significant increase in ceramide by 48 %. The activities of both N- and A-SMase were reduced by 44 % and 32 %, respectively, in the myocardium subjected to ischemia followed by reperfusion, but unaltered in the ischemic myocardium. It was also found that myocardial ischemia followed by reperfusion produced a marked inhibition of ceramidase (by 29 %). These results demonstrate that the myocardium of rats expresses N- and A-SMase and ceramidase, which contribute to the production and metabolism of ceramide, respectively. Tissue ceramide concentrations increased in reperfused myocardium. These increases in ceramide were not associated with enhanced SMase activity, but rather with reduced ceramidase activity.


The FASEB Journal | 2002

Delta opioid agonists and volatile anesthetics facilitate cardioprotection via potentiation of KATP channel opening

Hemal H. Patel; Lynda M. Ludwig; Ryan M. Fryer; Anna K. Hsu; David C. Warltier; Garrett J. Gross

Opioids and volatile anesthetics produce marked cardioprotective effects against myocardial infarction via the activation of ATP‐sensitive potassium (KATP) channels, however, the effect of combined treatment with both drugs is unknown. We examined the hypothesis that opioids and volatile anesthetics potentiate cardiac KATP channel opening, thereby enhancing cardioprotection. Rats were treated with the delta opioid agonists, TAN‐67 or BW373U86, or isoflurane, together or alone with and without diazoxide, a mitochondrial KATP channel opener. Glibenclamide, a non‐selective KATP channel blocker, was used to further characterize the signaling mechanism involved. Myocardial infarct size (IS) was determined by tetrazolium staining and was expressed as a percent of the area at risk (AAR). High doses of TAN‐67 (10 mg/kg), diazoxide (10 mg/kg), and isoflurane (1 MAC) produced a significant reduction in IS compared with the control group (30±3%, 36±5%, and 42±2 vs. 58±2%, respectively), whereas lower doses of the drugs had no effect except for the low dose of isoflurane (0.5 MAC). The combination of TAN‐67 and diazoxide or isoflurane and diazoxide resulted in a marked reduction in IS compared with controls in the presence of high (9±3% and 14±3%) and low (17±7% and 31±7%) dose combinations, respectively. The combination of TAN‐67 or BW373U86 and isoflurane also caused a striking reduction in IS/AAR (16±7% and 7±2%, respectively). To date, this is the first demonstration that opioids and volatile anesthetics work in conjunction to confer protection against myocardial infarction through potentiation of cardiac KATP channel opening.

Collaboration


Dive into the Anna K. Hsu's collaboration.

Top Co-Authors

Avatar

Garrett J. Gross

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ryan M. Fryer

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Hemal H. Patel

University of California

View shared research outputs
Top Co-Authors

Avatar

Jo El J. Schultz

University of Cincinnati Academic Health Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Janis T. Eells

University of Wisconsin–Milwaukee

View shared research outputs
Top Co-Authors

Avatar

Yigang Wang

University of Cincinnati

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge