Network


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

Hotspot


Dive into the research topics where Jidong Su is active.

Publication


Featured researches published by Jidong Su.


Basic Research in Cardiology | 2005

Erythropoietin protects the infant heart against ischemia–reperfusion injury by triggering multiple signaling pathways

Parvaneh Rafiee; Yang Shi; Jidong Su; Kirkwood A. Pritchard; James S. Tweddell; John E. Baker

Abstract The immediate protective effect of erythropoietin (EPO) against ischemia in heart suggests a role beyond hematopoiesis and the treatment of anemia. We determined the role of JAK/STAT and Ras/Rac/MAPK in the protective effect of EPO against ischemia–reperfusion injury in infant rabbit heart. EPO (1.0 U/ml) administered 15 minutes prior to 30–minutes global ischemia and 35 minutes reperfusion resulted in increased recovery of postischemic ventricular developed pressure in rabbit hearts. EPO exerted its immediate cardioprotective effect via activation of multiple signaling pathways by: 1) phosphorylation and activation of JAK1/2, STAT3 and STAT5A but not of STAT1α and STAT5B, 2) phosphorylation and activation of PI3 kinase and its downstream kinases Akt and Rac, 3) activation of PKCε, Raf, MEK1/2, p42/44 MAPK and p38 MAPK. Pretreatment with Wortmannin abolished EPO–induced Akt activation and phosphorylation. Pretreatment with Chelerythrine followed by EPO treatment resulted in partial inhibition of Raf activation, and abolished PKCε and p38 MAPK activation without any effect on Akt, MEK1/2 and p42/44 MAPK. PD98059 abolished MEK1/2 and p42/44 MAPK activation with no effect on Akt, Raf and p38 MAPK activation. SB203580 inhibited only p38 MAPK activation by EPO. We can conclude EPO increases immediate cardioprotection through the activation of multiple signal transduction pathways.


The FASEB Journal | 2012

Intestinal microbiota determine severity of myocardial infarction in rats

Vy Lam; Jidong Su; Stacy Koprowski; Anna Hsu; James S. Tweddell; Parvaneh Rafiee; Garrett J. Gross; Nita H. Salzman; John E. Baker

Signals from the intestinal microbiota are important for normal host physiology; alteration of the microbiota (dysbiosis) is associated with multiple disease states. We determined the effect of antibiotic‐induced intestinal dysbiosis on circulating cytokine levels and severity of ischemia/reperfusion injury in the heart. Treatment of Dahl S rats with a minimally absorbed antibiotic vancomycin, in the drinking water, decreased circulating leptin levels by 38%, resulted in smaller myocardial infarcts (27% reduction), and improved recovery of postischemic mechanical function (35%) as compared with untreated controls. Vancomycin altered the abundance of intestinal bacteria and fungi, measured by 16S and 18S ribosomal DNA quantity. Pretreatment with leptin (0.12 μg/kg i.v.) 24 h before ischemia/reperfusion abolished cardioprotection produced by vancomycin treatment. Dahl S rats fed the commercially available probiotic product Goodbelly, which contains the leptin‐suppressing bacteria Lactobacillus plantarum 299v, also resulted in decreased circulating leptin levels by 41%, smaller myocardial infarcts (29% reduction), and greater recovery of postischemic mechanical function (23%). Pretreatment with leptin (0.12 μg/kg i.v.) abolished cardioprotection produced by Goodbelly. This proof‐of‐concept study is the first to identify a mechanistic link between changes in intestinal microbiota and myocardial infarction and demonstrates that a probiotic supplement can reduce myocardial infarct size.—Lam, V., Su, J., Koprowski, S., Hsu, A., Tweddell, J. S., Rafiee, P., Gross, G. J., Salzman, N. H., Baker, J. E. Intestinal microbiota determine severity of myocardial infarction in rats. FASEB J. 26, 1727‐1735 (2012). www.fasebj.org


Circulation Research | 2002

Chronic Hypoxia Increases Endothelial Nitric Oxide Synthase Generation of Nitric Oxide by Increasing Heat Shock Protein 90 Association and Serine Phosphorylation

Yang Shi; John E. Baker; Chenyang Zhang; James S. Tweddell; Jidong Su; Kirkwood A. Pritchard

Abstract— Chronic hypoxia increases endothelial nitric oxide synthase (eNOS) production of nitric oxide (·NO) and cardioprotection in neonatal rabbit hearts. However, the mechanism by which this occurs remains unclear. Recent studies suggest that heat shock protein 90 (hsp90) alters eNOS function. In the present study, we examined the role of hsp90 in eNOS-dependent cardioprotection in neonatal rabbit hearts. Chronic hypoxia increased recovery of postischemic left ventricular developed pressure (LVDP). Geldanamycin (GA), which inhibits hsp90 and increases oxidative stress, decreased functional recovery in normoxic and hypoxic hearts. To determine if a loss in ·NO, afforded by GA, decreased recovery, GA-treated hearts were perfused with S-nitrosoglutathione (GSNO) as a source of ·NO. GSNO increased recovery of postischemic LVDP in GA-treated normoxic and hypoxic hearts to baseline levels. Although chronic hypoxia decreased phosphorylated eNOS (S1177) levels by ≈4- to 5-fold and total Akt and phosphorylated Akt by 4- and 5-fold, it also increased hsp90 association with eNOS by more than 3-fold. Using hydroethidine (HEt), a fluorescent probe for superoxide, we found that hypoxic hearts contained less ethidine (Et) staining than normoxic hearts. Normoxic hearts generated 3 times more superoxide by an N&ohgr;-nitro-l-arginine methyl ester (L-NAME)-inhibitable mechanism than hypoxic hearts. Taken together, these data indicate that the association of hsp90 with eNOS is important for increasing ·NO production and limiting eNOS-dependent superoxide anion generation. Such changes in eNOS function appear to play a critical role in protecting the myocardium against ischemic injury.


Basic Research in Cardiology | 2007

SCH 79797, a selective PAR1 antagonist, limits myocardial ischemia/reperfusion injury in rat hearts

Jennifer L. Strande; Anna Hsu; Jidong Su; Xiangping Fu; Garrett J. Gross; John E. Baker

Myocardial ischemia/reperfusion (I/R) injury is partly mediated by thrombin. In support, the functional inhibition of thrombin has been shown to decrease infarct size after I/R. Several cellular responses to thrombin are mediated by a G-protein coupled protease-activated receptor 1 (PAR1).However, the role of PAR1 in myocardial I/R injury has not been well characterized. Therefore, we hypothesized that PAR1 inhibition will reduce the amount of myocardial I/R injury. After we detected the presence of PAR1 mRNA and protein in the rat heart by RT-PCR and immunoblot analysis,we assessed the potential protective role of SCH 79797, a selective PAR1 antagonist, in two rat models of myocardial I/R injury. SCH 79797 treatment immediately before or during ischemia reduced myocardial necrosis following I/R in the intact rat heart. This response was dose-dependent with the optimal dose being 25 μg/kg IV. Likewise, SCH 79797 treatment before ischemia in the isolated heart model reduced infarct size and increased ventricular recovery following I/R in the isolated heart model with an optimal concentration of 1 μM. This reduction was abolished by a PAR1 selective agonist. SCH 79797-induced resistance to myocardial ischemia was abolished by wortmannin, an inhibitor of PI3 kinase; L-NMA, a NOS inhibitor; and glibenclamide, a nonselective KATP channel blocker. PAR1 activating peptide,wortmannin, L-NMA and glibenclamide alone had no effect on functional recovery or infarct size. A single treatment of SCH 79797 administered prior to or during ischemia confers immediate cardioprotection suggesting a potential therapeutic role of PAR1 antagonist in the treatment of injury resulting from myocardial ischemia and reperfusion.


Circulation | 2002

Activation of protein kinases in chronically hypoxic infant human and rabbit hearts: role in cardioprotection.

Parvaneh Rafiee; Yang Shi; Xiangrong Kong; Kirkwood A. Pritchard; James S. Tweddell; S. Bert Litwin; Kathleen A. Mussatto; Robert D.B. Jaquiss; Jidong Su; John E. Baker

Background—Many infants who undergo heart surgery have a congenital cyanotic defect in which the heart is chronically perfused with hypoxic blood. However, the signaling pathways by which infant hearts adapt to chronic hypoxia and resist subsequent surgical ischemia is unknown. Method and Results—We determined the activation and translocation of protein kinase C (PKC) isoforms and mitogen activated protein kinases (MAP kinases) in 15 infants with cyanotic (Sao2<85%) or acyanotic (Sao2>95%) heart defects undergoing surgical repair and in 80 rabbits raised from birth in a hypoxic (Sao2<85%) or normoxic (Sao2>95%) environment. Tissues from infant human and rabbit hearts were processed for Western and in vitro kinase analysis. In human infants with cyanotic heart defects, PKC&egr;, p38 MAP kinase, and JUN kinase but not p42/44 MAP kinase were activated and translocated from the cytosolic to the particulate fraction compared with acyanotic heart defects. In rabbit infants there was a parallel response for PKC&egr;, p38 MAP kinase, and JUN kinase similar to humans. In infant rabbit hearts inhibition of PKC&egr; with chelerythrine, p38 MAP kinase, with SB203580 and JUN kinase with curcumin abolished the cardioprotective effects of chronic hypoxia but had no effects on normoxic hearts. Conclusions—Infant human and rabbit hearts adapt to chronic hypoxia through activation of PKC&egr;, p38 MAP kinase, and JUN kinase signal transduction pathways. These pathways may be responsible for cardioprotection in the chronically hypoxic infant rabbit heart.


International Journal of Radiation Biology | 2009

10 GY TOTAL BODY IRRADIATION INCREASES RISK OF CORONARY SCLEROSIS, DEGENERATION OF HEART STRUCTURE AND FUNCTION IN A RAT MODEL

John E. Baker; Brian L. Fish; Jidong Su; Steven T. Haworth; Jennifer L. Strande; Richard A. Komorowski; Raymond Q. Migrino; Anil Doppalapudi; Leanne Harmann; X. Allen Li; J.W. Hopewell; John E. Moulder

Purpose: To determine the impact of 10 Gy total body irradiation (TBI) or local thorax irradiation, a dose relevant to a radiological terrorist threat, on lipid and liver profile, coronary microvasculature and ventricular function. Materials and methods: WAG/RijCmcr rats received 10 Gy TBI followed by bone marrow transplantation, or 10 Gy local thorax irradiation. Age-matched, non-irradiated rats served as controls. The lipid profile and liver enzymes, coronary vessel morphology, nitric oxide synthase (NOS) isoforms, protease activated receptor (PAR)-1 expression and fibrinogen levels were compared. Two-dimensional strain echocardiography assessed global radial and circumferential strain on the heart. Results: TBI resulted in a sustained increase in total and low density lipoprotein (LDL) cholesterol (190 ± 8 vs. 58 ± 6; 82 ± 8 vs. 13 ± 3 mg/dl, respectively). The density of small coronary arterioles was decreased by 32%. Histology revealed complete blockage of some vessels while cardiomyocytes remained normal. TBI resulted in cellular peri-arterial fibrosis whereas control hearts had symmetrical penetrating vessels with less collagen and fibroblasts. TBI resulted in a 32 ± 4% and 28 ± 3% decrease in endothelial NOS and inducible NOS protein, respectively, and a 21 ± 4% and 35 ± 5% increase in fibrinogen and PAR-1 protein respectively, after 120 days. TBI reduced radial strain (19 ± 8 vs. 46 ± 7%) and circumferential strain (−8 ± 3 vs. −15 ± 3%) compared to controls. Thorax-only irradiation produced no changes over the same time frame. Conclusions: TBI with 10 Gy, a dose relevant to radiological terrorist threats, worsened lipid profile, injured coronary microvasculature, altered endothelial physiology and myocardial mechanics. These changes were not manifest with local thorax irradiation. Non-thoracic circulating factors may be promoting radiation-induced injury to the heart.


Protein Science | 2009

Monomeric structure of the cardioprotective chemokine SDF-1/CXCL12

Christopher T. Veldkamp; Joshua J. Ziarek; Jidong Su; Harihar Basnet; Richard C. Lennertz; Joshua J. Weiner; Francis C. Peterson; John E. Baker; Brian F. Volkman

The chemokine stromal cell‐derived factor‐1 (SDF‐1/CXCL12) directs leukocyte migration, stem cell homing, and cancer metastasis through activation of CXCR4, which is also a coreceptor for T‐tropic HIV‐1. Recently, SDF‐1 was shown to play a protective role after myocardial infarction, and the protein is a candidate for development of new anti‐ischemic compounds. SDF‐1 is monomeric at nanomolar concentrations but binding partners promote self‐association at higher concentrations to form a typical CXC chemokine homodimer. Two NMR structures have been reported for the SDF‐1 monomer, but only one matches the conformation observed in a series of dimeric crystal structures. In the other model, the C‐terminal helix is tilted at an angle incompatible with SDF‐1 dimerization. Using a rat heart explant model for ischemia/reperfusion injury, we found that dimeric SDF‐1 exerts no cardioprotective effect, suggesting that the active species is monomeric. To resolve the discrepancy between existing models, we solved the NMR structure of the SDF‐1 monomer in different solution conditions. Irrespective of pH and buffer composition, the C‐terminal helix remains tilted at an angle with no evidence for the perpendicular arrangement. Furthermore, we find that phospholipid bicelles promote dimerization that necessarily shifts the helix to the perpendicular orientation, yielding dipolar couplings that are incompatible with the NOE distance constraints. We conclude that interactions with the alignment medium biased the previous structure, masking flexibility in the helix position that may be essential for the distinct functional properties of the SDF‐1 monomer.


Journal of Biological Chemistry | 2013

Heparin Oligosaccharides Inhibit Chemokine (CXC Motif) Ligand 12 (CXCL12) Cardioprotection by Binding Orthogonal to the Dimerization Interface, Promoting Oligomerization, and Competing with the Chemokine (CXC Motif) Receptor 4 (CXCR4) N Terminus

Joshua J. Ziarek; Christopher T. Veldkamp; Fuming Zhang; Nathan J. Murray; Gabriella A. Kartz; Xinle Liang; Jidong Su; John E. Baker; Robert J. Linhardt; Brian F. Volkman

Background: GAG/CXCL12 interactions are critical for chemokine function but co-administration may abrogate their individual cardioprotective effects in a clinical setting. Results: Biophysical studies distinguish CXCL12 residues involved in dimerization from those likely to contact heparin directly. Conclusion: CXCL12 dimerization is required for high affinity heparin binding and protects N-terminal degradation. Significance: Knowledge of the GAG-binding site will enable future development of heparin-insensitive CXCL12 therapeutics. The ability to interact with cell surface glycosaminoglycans (GAGs) is essential to the cell migration properties of chemokines, but association with soluble GAGs induces the oligomerization of most chemokines including CXCL12. Monomeric CXCL12, but not dimeric CXCL12, is cardioprotective in a number of experimental models of cardiac ischemia. We found that co-administration of heparin, a common treatment for myocardial infarction, abrogated the protective effect of CXCL12 in an ex vivo rat heart model for myocardial infarction. The interaction between CXCL12 and heparin oligosaccharides has previously been analyzed through mutagenesis, in vitro binding assays, and molecular modeling. However, complications from heparin-induced CXCL12 oligomerization and studies using very short oligosaccharides have led to inconsistent conclusions as to the residues involved, the orientation of the binding site, and whether it overlaps with the CXCR4 N-terminal site. We used a constitutively dimeric variant to simplify the NMR analysis of CXCL12-binding heparin oligosaccharides of varying length. Biophysical and mutagenic analyses reveal a CXCL12/heparin interaction surface that lies perpendicular to the dimer interface, does not involve the chemokine N terminus, and partially overlaps with the CXCR4-binding site. We further demonstrate that heparin-mediated enzymatic protection results from the promotion of dimerization rather than direct heparin binding to the CXCL12 N terminus. These results clarify the structural basis for GAG recognition by CXCL12 and lend insight into the development of CXCL12-based therapeutics.


Journal of Pharmacology and Experimental Therapeutics | 2007

Inhibiting Protease-Activated Receptor 4 Limits Myocardial Ischemia/Reperfusion Injury in Rat Hearts by Unmasking Adenosine Signaling

Jennifer L. Strande; Anna Hsu; Jidong Su; Xiangping Fu; Garrett J. Gross; John E. Baker

Harnessing endogenous cardioprotectants is a novel therapeutic strategy to combat ischemia/reperfusion (I/R) injury. Thrombin causes I/R injury, whereas exogenous adenosine prevents I/R injury. We hypothesized that blocking thrombin receptor activation with a protease-activated receptor (PAR) 4 antagonist would unmask the cardioprotective effects of endogenous adenosine. The protective role of two structurally unrelated PAR4 antagonists, trans-cinnamoyl-YPGKF-amide (tc-Y-NH2) and palmitoyl-SGRRYGHALR-amide (P4pal10), were evaluated in two rat models of myocardial I/R injury. P4pal10 (10 μg/kg) treatment before ischemia significantly decreased infarct size (IS) by 31, 21, and 19% when given before, during, and after ischemia in the in vivo model. tc-Y-NH2 (5 μM) treatment before ischemia decreased IS by 51% in the in vitro model and increased recovery of ventricular function by 26%. To assess whether the cardioprotective effects of PAR4 blockade were due to endogenous adenosine, isolated hearts were treated with a nonselective adenosine receptor blocker, 8-sulfaphenyltheophylline (8-SPT), and tc-Y-NH2 before ischemia. 8-SPT abolished the protective effects of tc-Y-NH2 but did not affect IS when given alone. Adenosine-mediated survival pathways were then explored. The cardioprotective effects of tc-Y-NH2 were abolished by inhibition of Akt (wortmannin), extracellular signal-regulated kinase 1/2 [PD98059 (2′-amino-3′-methoxyflavone)], nitric-oxide synthase [NG-monomethyl-l-arginine (l-NMA)], and KATP channels (glibenclamide). PD98059, l-NMA, and glibenclamide alone had no effect on cardioprotection in vitro. Furthermore, inhibition of mitochondrial KATP channels [5-hydroxydecanoic acid (5-HD)] and sarcolemmal KATP channels (sodium (5-(2-(5-chloro-2-methoxybenzamido)ethyl)-2-methoxyphenylsulfonyl)(methylcarbamothioyl)amide; HMR 1098) abolished P4pal10-induced cardioprotection in vivo. Thrombin receptor blockade by PAR4 inhibition provides protection against injury from myocardial I/R by unmasking adenosine receptor signaling and supports the hypothesis of a coupling between thrombin receptors and adenosine receptors.


Radiation Research | 2013

Cardiac Injury after 10 Gy Total Body Irradiation: Indirect Role of Effects on Abdominal Organs

Marek Lenarczyk; Vy Lam; Eric S. Jensen; Brian L. Fish; Jidong Su; Stacy Koprowski; Richard A. Komorowski; Leanne Harmann; Raymond Q. Migrino; X. Allen Li; J.W. Hopewell; John E. Moulder; John E. Baker

The objective of this study was to determine whether radiation-induced injury to the heart after 10 Gy total body irradiation (TBI) is direct or indirect. Young male WAG/RijCmcr rats received a 10 Gy single dose using TBI, upper hemi-body (UHB) irradiation, lower hemi-body (LHB) irradiation, TBI with the kidneys shielded or LHB irradiation with the intestines shielded. Age-matched, sham-irradiated rats served as controls. The lipid profile, kidney injury, heart and liver morphology and cardiac function were determined up to 120 days after irradiation. LHB, but not UHB irradiation, increased the risk factors for cardiac disease as well as the occurrence of cardiac and kidney injury in a way that was quantitatively and qualitatively similar to that observed after TBI. Shielding of the kidneys prevented the increases in risk factors for cardiac disease. Shielding of the intestines did not prevent the increases in risk factors for cardiac disease. There was no histological evidence of liver injury 120 days after irradiation. Injury to the heart from irradiation appears to be indirect, supporting the notion that injury to abdominal organs, principally the kidneys, is responsible for the increased risk factors for and the occurrence of cardiac disease after TBI and LHB irradiation.

Collaboration


Dive into the Jidong Su's collaboration.

Top Co-Authors

Avatar

John E. Baker

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Garrett J. Gross

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

James S. Tweddell

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Anna Hsu

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Yang Shi

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Jennifer L. Strande

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Xiangping Fu

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nita H. Salzman

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Parvaneh Rafiee

Medical College of Wisconsin

View shared research outputs
Researchain Logo
Decentralizing Knowledge