Network


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

Hotspot


Dive into the research topics where Xinjun Teng is active.

Publication


Featured researches published by Xinjun Teng.


Journal of Clinical Investigation | 2007

Inhaled NO accelerates restoration of liver function in adults following orthotopic liver transplantation

John D. Lang; Xinjun Teng; Phillip Chumley; Jack H. Crawford; T. Scott Isbell; Balu K. Chacko; Yuliang Liu; Nirag Jhala; D. Ralph Crowe; Alvin B. Smith; Richard C. Cross; Luc Frenette; Eric E. Kelley; Diana W. Wilhite; Cheryl R. Hall; Grier P. Page; Michael B. Fallon; J. Steven Bynon; Devin E. Eckhoff; Rakesh P. Patel

Ischemia/reperfusion (IR) injury in transplanted livers contributes to organ dysfunction and failure and is characterized in part by loss of NO bioavailability. Inhalation of NO is nontoxic and at high concentrations (80 ppm) inhibits IR injury in extrapulmonary tissues. In this prospective, blinded, placebo-controlled study, we evaluated the hypothesis that administration of inhaled NO (iNO; 80 ppm) to patients undergoing orthotopic liver transplantation inhibits hepatic IR injury, resulting in improved liver function. Patients were randomized to receive either placebo or iNO (n = 10 per group) during the operative period only. When results were adjusted for cold ischemia time and sex, iNO significantly decreased hospital length of stay, and evaluation of serum transaminases (alanine transaminase, aspartate aminotransferase) and coagulation times (prothrombin time, partial thromboplastin time) indicated that iNO improved the rate at which liver function was restored after transplantation. iNO did not significantly affect changes in inflammatory markers in liver tissue 1 hour after reperfusion but significantly lowered hepatocyte apoptosis. Evaluation of circulating NO metabolites indicated that the most likely candidate transducer of extrapulmonary effects of iNO was nitrite. In summary, this study supports the clinical use of iNO as an extrapulmonary therapeutic to improve organ function following transplantation.


Proceedings of the National Academy of Sciences of the United States of America | 2008

Chronic sodium nitrite therapy augments ischemia-induced angiogenesis and arteriogenesis

Dinesh Kumar; Billy Glynn Branch; Christopher B. Pattillo; Jay Hood; Stephen Thoma; Stephen Simpson; Sandra Illum; Neeraj Arora; John H. Chidlow; Will Langston; Xinjun Teng; David J. Lefer; Rakesh P. Patel; Christopher G. Kevil

Chronic tissue ischemia due to defective vascular perfusion is a hallmark feature of peripheral artery disease for which minimal therapeutic options exist. We have reported that sodium nitrite therapy exerts cytoprotective effects against acute ischemia/reperfusion injury in both heart and liver, consistent with the model of bioactive NO formation from nitrite during ischemic stress. Here, we test the hypothesis that chronic sodium nitrite therapy can selectively augment angiogenic activity and tissue perfusion in the murine hind-limb ischemia model. Various therapeutic doses (8.25–3,300 μg/kg) of sodium nitrite or PBS were administered. Sodium nitrite significantly restored ischemic hind-limb blood flow in a time-dependent manner, with low-dose sodium nitrite being most effective. Nitrite therapy significantly increased ischemic limb vascular density and stimulated endothelial cell proliferation. Remarkably, the effects of sodium nitrite therapy were evident within 3 days of the ischemic insult demonstrating the potency and efficacy of chronic sodium nitrite therapy. Sodium nitrite therapy also increased ischemic tissue nitrite and NO metabolites compared to nonischemic limbs. Use of the NO scavenger carboxy PTIO completely abolished sodium nitrite-dependent ischemic tissue blood flow and angiogenic activity consistent with nitrite reduction to NO being the proangiogenic mechanism. These data demonstrate that chronic sodium nitrite therapy is a recently discovered therapeutic treatment for peripheral artery disease and critical limb ischemia.


Nature Medicine | 2008

SNO-hemoglobin is not essential for red blood cell-dependent hypoxic vasodilation

T. Scott Isbell; Chiao Wang Sun; Li Chen Wu; Xinjun Teng; Dario A. Vitturi; Billy Glynn Branch; Christopher G. Kevil; Ning Peng; Jm Wyss; Namasivayam Ambalavanan; Lisa M. Schwiebert; Jinxiang Ren; Kevin M. Pawlik; Matthew B. Renfrow; Rakesh P. Patel; Tim M. Townes

The coupling of hemoglobin sensing of physiological oxygen gradients to stimulation of nitric oxide (NO) bioactivity is an established principle of hypoxic blood flow. One mechanism proposed to explain this oxygen-sensing–NO bioactivity linkage postulates an essential role for the conserved Cys93 residue of the hemoglobin β-chain (βCys93) and, specifically, for S-nitrosation of βCys93 to form S-nitrosohemoglobin (SNO-Hb). The SNO-Hb hypothesis, which conceptually links hemoglobin and NO biology, has been debated intensely in recent years. This debate has precluded a consensus on physiological mechanisms and on assessment of the potential role of SNO-Hb in pathology. Here we describe new mouse models that exclusively express either human wild-type hemoglobin or human hemoglobin in which the βCys93 residue is replaced with alanine to assess the role of SNO-Hb in red blood cell–mediated hypoxic vasodilation. Substitution of this residue, precluding hemoglobin S-nitrosation, did not change total red blood cell S-nitrosothiol abundance but did shift S-nitrosothiol distribution to lower molecular weight species, consistent with the loss of SNO-Hb. Loss of βCys93 resulted in no deficits in systemic or pulmonary hemodynamics under basal conditions and, notably, did not affect isolated red blood cell–dependent hypoxic vasodilation. These results demonstrate that SNO-Hb is not essential for the physiologic coupling of erythrocyte deoxygenation with increased NO bioactivity in vivo.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2007

Sildenafil Promotes Ischemia-Induced Angiogenesis Through a PKG-Dependent Pathway

Annamalai Senthilkumar; Ray D. Smith; Jayant Khitha; Neeraj Arora; Srikar Veerareddy; Will Langston; John H. Chidlow; Shayne C. Barlow; Xinjun Teng; Rakesh P. Patel; David J. Lefer; Christopher G. Kevil

Background—Peripheral artery disease (PAD) is a prevalent cardiovascular disorder that results in tissue ischemia which can progress to critical limb ischemia. Restoration of tissue perfusion in the setting of chronic ischemia through stimulation of arteriogenesis and angiogenesis remains a key therapeutic target for PAD. However, experimental therapeutics, including growth factor and gene therapy, have had little clinical success indicating the need for a better understanding of molecular pathways required for therapeutic angiogenesis. Methods and Results—Here we report that phosphodiesterase-5 inhibition by sildenafil significantly increases vascular perfusion, tissue blood flow, and vascular density during chronic ischemia of the mouse hind limb. Importantly, sildenafil therapy did not alter any of these parameters in nonischemic limbs. Sildenafil increased tissue cGMP levels independently of increases in nitric oxide production, and sildenafil therapy stimulated angiogenesis in ischemic limbs of eNOS−/− and iNOS−/− mice. Lastly, sildenafil-mediated angiogenic activity was blocked by inhibition of protein kinase G using the PKG antagonist DT-3. Conclusions—These data demonstrate that sildenafil therapy results in increased angiogenic activity through a PKG-dependent pathway that is independent of nitric oxide production or NOS activity and identify the angiogenic therapeutic potential of sildenafil for critical limb ischemia.


Journal of Biological Chemistry | 2006

Mechanisms of Cystic Fibrosis Transmembrane Conductance Regulator Activation by S-Nitrosoglutathione

Lan Chen; Rakesh P. Patel; Xinjun Teng; Charles A. Bosworth; Jack R. Lancaster; Sadis Matalon

We investigated the mechanisms by which S-nitrosoglutathione (GSNO) alters cystic fibrosis transmembrane conductance regulator (CFTR) mediated chloride (Cl–) secretion across Calu-3 cells, an extensively used model of human airway gland serous cells. Confluent monolayers of Calu-3 cells, grown under an air-liquid interface, were mounted in Ussing chambers for the measurements of chloride short circuit current (Isc) and trans-epithelial resistance (Rt). Addition of GSNO into the apical compartment of these chambers resulted in significant and sustained increase of Isc with an IC50 of 3.2 ± 1 μm (mean ± 1 S.E.; n = 6). Addition of either glibenclamide or pre-treatment of Calu-3 cells with the soluble guanylate cyclase inhibitor 1H-(1,2,4)-oxadiazolo[4,3-a]quinoxalin-1-one totally prevented the GSNO-induced increase of Isc. Conversely, BAY 41-2272, a sGC stimulator, increased Isc in a dose-response fashion. The GSNO increase of Isc was reversed by addition of two phosphatases (PP2A1, PP2A2) into the apical compartment of Ussing chambers containing Calu-3 monolayers. Oxy-myoglobin (oxy-Mb, 300 μm) added into the apical compartment of Ussing chambers either prior or after GSNO either completely prevented or immediately reversed the increase of Isc. However, smaller concentrations of oxy-Mb (1–10 μm), sufficient to scavenge NO in the medium (as assessed by direct measurement of NO in the Ussing chamber using an ISO-NO meter) decreased Isc partially. Oxy-Mb did not reverse the increase of Isc following addition of GSNO and cysteine (50 μm). These findings indicate that GSNO stimulates Cl secretion via both cGMP-dependent and cGMP-independent mechanisms.


American Journal of Physiology-heart and Circulatory Physiology | 2010

Erythrocyte-dependent regulation of human skeletal muscle blood flow: role of varied oxyhemoglobin and exercise on nitrite, S-nitrosohemoglobin, and ATP

Stéphane P. Dufour; Rakesh P. Patel; Angela Brandon; Xinjun Teng; James Pearson; Horace Barker; Leena Ali; Ada H.Y. Yuen; Ryszard T. Smolenski; José González-Alonso

The erythrocyte is proposed to play a key role in the control of local tissue perfusion via three O(2)-dependent signaling mechanisms: 1) reduction of circulating nitrite to vasoactive NO, 2) S-nitrosohemoglobin (SNO-Hb)-dependent vasodilatation, and 3) release of the vasodilator and sympatholytic ATP; however, their relative roles in vivo remain unclear. Here we evaluated each mechanism to gain insight into their roles in the regulation of human skeletal muscle blood flow during hypoxia and hyperoxia at rest and during exercise. Arterial and femoral venous hemoglobin O(2) saturation (O(2)Hb), plasma and erythrocyte NO and ATP metabolites, and leg and systemic hemodynamics were measured in 10 healthy males exposed to graded hypoxia, normoxia, and graded hyperoxia both at rest and during submaximal one-legged knee-extensor exercise. At rest, leg blood flow and NO and ATP metabolites in plasma and erythrocytes remained unchanged despite large alterations in O(2)Hb. During exercise, however, leg and systemic perfusion and vascular conductance increased in direct proportion to decreases in arterial and venous O(2)Hb (r(2) = 0.86-0.98; P = 0.01), decreases in venous plasma nitrite (r(2) = 0.93; P < 0.01), increases in venous erythrocyte nitroso species (r(2) = 0.74; P < 0.05), and to a lesser extent increases in erythrocyte SNO (r(2) = 0.59; P = 0.07). No relationship was observed with plasma ATP (r(2) = 0.01; P = 0.99) or its degradation compounds. These in vivo data indicate that, during low-intensity exercise and hypoxic stress, but not hypoxic stress alone, plasma nitrite consumption and formation of erythrocyte nitroso species are associated with limb vasodilatation and increased blood flow in the human skeletal muscle vasculature.


Cardiovascular Research | 2010

Dipyridamole enhances ischaemia-induced arteriogenesis through an endocrine nitrite/nitric oxide-dependent pathway

Prasanna Venkatesh; Christopher B. Pattillo; Billy G. Branch; Jay Hood; Steven Thoma; Sandra Illum; Sibile Pardue; Xinjun Teng; Rakesh P. Patel; Christopher G. Kevil

AIMS Anti-platelet agents, such as dipyridamole, have several clinical benefits for peripheral artery disease with the speculation of angiogenic potential that could preserve ischaemic tissue viability, yet the effect of dipyridamole on ischaemic arteriogenesis or angiogenesis is unknown. Here we test the hypothesis that dipyridamole therapy augments arteriolar vessel development and function during chronic ischaemia. METHODS AND RESULTS Mice were treated with 200 mg/kg dipyridamole twice daily to achieve therapeutic plasma levels (0.8-1.2 microg/mL). Chronic hindlimb ischaemia was induced by permanent femoral artery ligation followed by measurement of tissue perfusion using laser Doppler blood flow along with quantification of vascular density, cell proliferation, and activation of nitric oxide (NO) metabolism. Dipyridamole treatment quickly restored ischaemic hindlimb blood flow, increased vascular density and cell proliferation, and enhanced collateral artery perfusion compared with control treatments. The beneficial effects of dipyridamole on blood flow and vascular density were dependent on NO production as dipyridamole did not augment ischaemic tissue reperfusion, vascular density, or endothelial cell proliferation in endothelial NO synthase (eNOS)-deficient mice. Blood and tissue nitrite levels were significantly higher in dipyridamole-treated mice compared with controls and eNOS(-/-) mice, verifying increased NO production that was regulated in a PKA-dependent manner. CONCLUSION Dipyridamole augments nitrite/NO production, leading to enhanced arteriogenesis activity and blood perfusion in ischaemic limbs. Together, these data suggest that dipyridamole can augment ischaemic vessel function and restore blood flow, which may be beneficial in peripheral artery disease.


American Journal of Physiology-heart and Circulatory Physiology | 2009

Regulation of nitrite transport in red blood cells by hemoglobin oxygen fractional saturation.

Dario A. Vitturi; Xinjun Teng; Jose C. Toledo; Sadis Matalon; Jack R. Lancaster; Rakesh P. Patel

Allosteric regulation of nitrite reduction by deoxyhemoglobin has been proposed to mediate nitric oxide (NO) formation during hypoxia. Nitrite is predominantly an anion at physiological pH, raising questions about the mechanism by which it enters the red blood cell (RBC) and whether this is regulated and coupled to deoxyhemoglobin-mediated reduction. We tested the hypothesis that nitrite transport by RBCs is regulated by fractional saturation. Using human RBCs, nitrite consumption was faster at lower fractional saturations, consistent with faster reactions with deoxyheme. A membrane-based regulation was suggested by slower nitrite consumption with intact versus lysed RBCs. Interestingly, upon nitrite addition, intracellular nitrite concentrations attained a steady state that, despite increased rates of consumption, did not change with decreasing oxygen tensions, suggesting a deoxygenation-sensitive step that either increases nitrite import or decreases the rate of nitrite export. A role for anion exchanger (AE)-1 in the control of nitrite export was suggested by increased intracellular nitrite concentrations in RBCs treated with DIDS. Moreover, deoxygenation decreased steady-state levels of intracellular nitrite in AE-1-inhibited RBCs. Based on these data, we propose a model in which deoxyhemoglobin binding to AE-1 inhibits nitrite export under low oxygen tensions allowing for the coupling between deoxygenation and nitrite reduction to NO along the arterial-to-venous gradient.


Methods in Enzymology | 2008

Novel Method for Measuring S-Nitrosothiols Using Hydrogen Sulfide

Xinjun Teng; T. Scott Isbell; Jack H. Crawford; Charles A. Bosworth; Gregory I. Giles; Jeffrey R. Koenitzer; Jack R. Lancaster; Jeannette E. Doeller; David W. Kraus; Rakesh P. Patel

Recent advances in techniques that allow sensitive and specific measurement of S-nitrosothiols (RSNOs) have provided evidence for a role for these compounds in various aspects of nitric oxide (NO) biology. The most widely used approach is to couple reaction chemistry that selectively reduces RSNOs by one electron to produce NO, with the sensitive detection of the latter under anaerobic conditions using ozone based chemiluminescence in NO analyzers. Herein, we report a novel reaction that is readily adaptable for commercial NO analyzers that utilizes hydrogen sulfide (H2S), a gas that can reduce RSNO to NO and, analogous to NO, is produced by endogenous metabolism and has effects on diverse biological functions. We discuss factors that affect H2S based methods for RSNO measurement and discuss the potential of H2S as an experimental tool to measure RSNO.


Kidney International | 2012

Sodium nitrite protects against kidney injury induced by brain death and improves post-transplant function

Stacey S. Kelpke; Bo Chen; Kelley M. Bradley; Xinjun Teng; Phillip Chumley; Angela Brandon; Brett Yancey; Brandon Moore; Hughston Head; Liliana Viera; John A. Thompson; David K. Crossman; Molly S. Bray; Devin E. Eckhoff; Anupam Agarwal; Rakesh P. Patel

Renal injury induced by brain death is characterized by ischemia and inflammation and limiting it is a therapeutic goal that could improve outcomes in kidney transplantation. Brain death resulted in decreased circulating nitrite levels and increased infiltrating inflammatory cell infiltration into the kidney. Since nitrite stimulates nitric oxide signaling in ischemic tissues, we tested whether nitrite therapy was beneficial in a rat model of brain death followed by kidney transplantation. Nitrite, administered over 2 hours of brain death, blunted the increased inflammation without affecting brain death-induced alterations in hemodynamics. Kidneys were transplanted after 2 hours of brain death and renal function followed over 7 days. Allografts collected from nitrite-treated brain dead rats showed significant improvement in function over the first 2 to 4 days post transplantation compared to untreated brain dead animals. Gene microarray analysis after 2 hours of brain death without or with nitrite therapy showed the latter significantly altered the expression of about 400 genes. Ingenuity Pathway analysis indicated multiple signaling pathways were affected by nitrite, including those related to hypoxia, transcription and genes related to humoral immune responses. Thus, nitrite-therapy attenuates brain death-induced renal injury by regulating responses to ischemia and inflammation, ultimately leading to better post-transplant kidney function.

Collaboration


Dive into the Xinjun Teng's collaboration.

Top Co-Authors

Avatar

Rakesh P. Patel

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

T. Scott Isbell

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Balu K. Chacko

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Devin E. Eckhoff

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Phillip Chumley

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Angela Brandon

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Anupam Agarwal

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jack H. Crawford

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Jack R. Lancaster

University of Alabama at Birmingham

View shared research outputs
Researchain Logo
Decentralizing Knowledge