Network


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

Hotspot


Dive into the research topics where Adrienne L. King is active.

Publication


Featured researches published by Adrienne L. King.


Angewandte Chemie | 2011

A Fluorescent Probe for Fast and Quantitative Detection of Hydrogen Sulfide in Blood

Hanjing Peng; Yunfeng Cheng; Chaofeng Dai; Adrienne L. King; Benjamin L. Predmore; David J. Lefer; Binghe Wang

Hydrogen sulfide (H2S), well known for its unpleasant rotten egg smell, was traditionally considered as a toxic gas. However, recent studies have demonstrated that hydrogen sulfide is an endogenously produced gaseous signaling compound (gasotransmitter) with importance on par with that of the other two known endogenous gasotransmitters, nitric oxide (NO)[1] and carbon monoxide (CO).[2] H2S has been recognized for mediating a wide range of physiological effects. Studies have shown that H2S can have an effect on the cardiovascular system[3] by acting as a K-ATP channel opener.[4] Several studies have shown the protective roles of H2S, in situations such as myocardial ischemia, most likely through a combination of antioxidant and anti-apoptotic signaling.[5] Further studies also showed that H2S may be a therapeutic benefit for the treatment of ischemia-induced heart failure.[6-7] It is also a modulator in the central nervous system,[8-10] respiratory system, gastrointestinal system, and endocrine system.[11] It seems that hydrogen sulfide exhibits almost all the beneficial effects of NO without generating the toxic reactive oxygen species (ROS). In contrast, it also acts as an anti-oxidant or scavenger of ROS. Furthermore, research has indicated that hydrogen sulfide level is related to diseases such as Down syndrome[12] and Alzheimer’s disease.[13] Therefore, recent years have seen a steady increase in the interest in understanding hydrogen sulfide ’s physiological and pathological functions.[11, 14-15] One significant limiting factor in studying hydrogen sulfide is the lack of sensors and agents that allow for its rapid and accurate detection. There have been literature methods using colorimetric,[16-18] electrochemical analysis[19-21] and gas chromatography.[22-23] However, hydrogen sulfide catabolism is known to be fast, which could result in continuous fluctuation in its concentration, leading to difficulties in accurate analysis of this important molecule. Current methods do not allow for fast, accurate, and real-time determinations. Literature reported endogenous sulfide concentrations vary substantially among publications with most publications suggesting that sulfide concentration in blood is in the 10-100 μM range.[24-29] There are other studies suggesting sulfide concentration being much lower than this.[30-31] Therefore, there is an urgent need for the development of new methods for the efficient detection of sulfide in biological systems.


Biochemical Journal | 2009

High fat diet induces dysregulation of hepatic oxygen gradients and mitochondrial function in vivo.

Sudheer K. Mantena; Denty Paul Vaughn; Kelly K. Andringa; Heather B. Eccleston; Adrienne L. King; Gary A. Abrams; Jeannette E. Doeller; David W. Kraus; Victor M. Darley-Usmar; Shannon M. Bailey

NAFLD (non-alcoholic fatty liver disease), associated with obesity and the cardiometabolic syndrome, is an important medical problem affecting up to 20% of western populations. Evidence indicates that mitochondrial dysfunction plays a critical role in NAFLD initiation and progression to the more serious condition of NASH (non-alcoholic steatohepatitis). Herein we hypothesize that mitochondrial defects induced by exposure to a HFD (high fat diet) contribute to a hypoxic state in liver and this is associated with increased protein modification by RNS (reactive nitrogen species). To test this concept, C57BL/6 mice were pair-fed a control diet and HFD containing 35% and 71% total calories (1 cal≈4.184 J) from fat respectively, for 8 or 16 weeks and liver hypoxia, mitochondrial bioenergetics, NO (nitric oxide)-dependent control of respiration, and 3-NT (3-nitrotyrosine), a marker of protein modification by RNS, were examined. Feeding a HFD for 16 weeks induced NASH-like pathology accompanied by elevated triacylglycerols, increased CYP2E1 (cytochrome P450 2E1) and iNOS (inducible nitric oxide synthase) protein, and significantly enhanced hypoxia in the pericentral region of the liver. Mitochondria from the HFD group showed increased sensitivity to NO-dependent inhibition of respiration compared with controls. In addition, accumulation of 3-NT paralleled the hypoxia gradient in vivo and 3-NT levels were increased in mitochondrial proteins. Liver mitochondria from mice fed the HFD for 16 weeks exhibited depressed state 3 respiration, uncoupled respiration, cytochrome c oxidase activity, and mitochondrial membrane potential. These findings indicate that chronic exposure to a HFD negatively affects the bioenergetics of liver mitochondria and this probably contributes to hypoxic stress and deleterious NO-dependent modification of mitochondrial proteins.


Circulation | 2013

H2S Protects Against Pressure Overload Induced Heart Failure via Upregulation of Endothelial Nitric Oxide Synthase (eNOS)

Kazuhisa Kondo; Shashi Bhushan; Adrienne L. King; Sumanth D. Prabhu; Tariq Hamid; Steven C. Koenig; Toyoaki Murohara; Benjamin L. Predmore; Gabriel Gojon; Rui Wang; Naveena Karusula; Chad K. Nicholson; John W. Calvert; David J. Lefer

Background— Cystathionine &ggr;-lyase (CSE) produces H2S via enzymatic conversion of L-cysteine and plays a critical role in cardiovascular homeostasis. We investigated the effects of genetic modulation of CSE and exogenous H2S therapy in the setting of pressure overload–induced heart failure. Methods and Results— Transverse aortic constriction was performed in wild-type, CSE knockout, and cardiac-specific CSE transgenic mice. In addition, C57BL/6J or CSE knockout mice received a novel H2S donor (SG-1002). Mice were followed up for 12 weeks with echocardiography. We observed a >60% reduction in myocardial and circulating H2S levels after transverse aortic constriction. CSE knockout mice exhibited significantly greater cardiac dilatation and dysfunction than wild-type mice after transverse aortic constriction, and cardiac-specific CSE transgenic mice maintained cardiac structure and function after transverse aortic constriction. H2S therapy with SG-1002 resulted in cardioprotection during transverse aortic constriction via upregulation of the vascular endothelial growth factor–Akt–endothelial nitric oxide synthase–nitric oxide–cGMP pathway with preserved mitochondrial function, attenuated oxidative stress, and increased myocardial vascular density. Conclusions— Our results demonstrate that H2S levels are decreased in mice in the setting of heart failure. Moreover, CSE plays a critical role in the preservation of cardiac function in heart failure, and oral H2S therapy prevents the transition from compensated to decompensated heart failure in part via upregulation of endothelial nitric oxide synthase and increased nitric oxide bioavailability.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Hydrogen sulfide cytoprotective signaling is endothelial nitric oxide synthase-nitric oxide dependent

Adrienne L. King; David J. Polhemus; Shashi Bhushan; Hiroyuki Otsuka; Kazuhisa Kondo; Chad K. Nicholson; Jessica M. Bradley; Kazi N. Islam; John W. Calvert; Ya-Xiong Tao; Tammy R. Dugas; Eric E. Kelley; John W. Elrod; Paul L. Huang; Rui Wang; David J. Lefer

Significance Physiological concentrations of hydrogen sulfide (H2S) exert potent prosurvival actions. We demonstrate that the cytoprotective actions of H2S are mediated in part via a second gaseous signaling molecule, nitric oxide (NO). We found that cystathionine γ-lyase (CSE) KO mice with reduced H2S levels exhibit increased oxidative stress and an exacerbated response to myocardial ischemia/reperfusion injury. CSE KO mice also exhibit reduced levels of NO and reduced NO synthesis via endothelial NO synthase (eNOS). Both oxidative stress and myocardial injury in CSE KO mice were attenuated by exogenous H2S therapy, with increased eNOS function and restoration of NO levels. These findings provide insight into H2S-mediated cytoprotetion and important information regarding the translation of H2S therapy to the clinic. Previous studies have demonstrated that hydrogen sulfide (H2S) protects against multiple cardiovascular disease states in a similar manner as nitric oxide (NO). H2S therapy also has been shown to augment NO bioavailability and signaling. The purpose of this study was to investigate the impact of H2S deficiency on endothelial NO synthase (eNOS) function, NO production, and ischemia/reperfusion (I/R) injury. We found that mice lacking the H2S-producing enzyme cystathionine γ-lyase (CSE) exhibit elevated oxidative stress, dysfunctional eNOS, diminished NO levels, and exacerbated myocardial and hepatic I/R injury. In CSE KO mice, acute H2S therapy restored eNOS function and NO bioavailability and attenuated I/R injury. In addition, we found that H2S therapy fails to protect against I/R in eNOS phosphomutant mice (S1179A). Our results suggest that H2S-mediated cytoprotective signaling in the setting of I/R injury is dependent in large part on eNOS activation and NO generation.


American Journal of Physiology-heart and Circulatory Physiology | 2012

The Polysulfide, Diallyl trisulfide, Protects the Ischemic Myocardium by Preservation of Endogenous Hydrogen Sulfide and Increasing Nitric Oxide Bioavailability

Benjamin L. Predmore; Kazuhisa Kondo; Shashi Bhushan; Maxim Zlatopolsky; Adrienne L. King; Juan Pablo Aragon; D. Bennett Grinsfelder; Marah E. Condit; David J. Lefer

Diallyl trisulfide (DATS), a polysulfide constituent found in garlic oil, is capable of the release of hydrogen sulfide (H(2)S). H(2)S is a known cardioprotective agent that protects the heart via antioxidant, antiapoptotic, anti-inflammatory, and mitochondrial actions. Here, we investigated DATS as a stable donor of H(2)S during myocardial ischemia-reperfusion (MI/R) injury in vivo. We investigated endogenous H(2)S levels, infarct size, postischemic left ventricular function, mitochondrial respiration and coupling, endothelial nitric oxide (NO) synthase (eNOS) activation, and nuclear E2-related factor (Nrf2) translocation after DATS treatment. Mice were anesthetized and subjected to a surgical model of MI/R injury with and without DATS treatment (200 μg/kg). Both circulating and myocardial H(2)S levels were determined using chemiluminescent gas chromatography. Infarct size was measured after 45 min of ischemia and 24 h of reperfusion. Troponin I release was measured at 2, 4, and 24 h after reperfusion. Cardiac function was measured at baseline and 72 h after reperfusion by echocardiography. Cardiac mitochondria were isolated after MI/R, and mitochondrial respiration was investigated. NO metabolites, eNOS phosphorylation, and Nrf2 translocation were determined 30 min and 2 h after DATS administration. Myocardial H(2)S levels markedly decreased after I/R injury but were rescued by DATS treatment (P < 0.05). DATS administration significantly reduced infarct size per area at risk and per left ventricular area compared with control (P < 0.001) as well as circulating troponin I levels at 4 and 24 h (P < 0.05). Myocardial contractile function was significantly better in DATS-treated hearts compared with vehicle treatment (P < 0.05) 72 h after reperfusion. DATS reduced mitochondrial respiration in a concentration-dependent manner and significantly improved mitochondrial coupling after reperfusion (P < 0.01). DATS activated eNOS (P < 0.05) and increased NO metabolites (P < 0.05). DATS did not appear to significantly induce the Nrf2 pathway. Taken together, these data suggest that DATS is a donor of H(2)S that can be used as a cardioprotective agent to treat MI/R injury.


Experimental Physiology | 2011

Cytoprotective actions of hydrogen sulfide in ischaemia–reperfusion injury

Adrienne L. King; David J. Lefer

Hydrogen sulfide (H2S) has been known as a highly toxic gas for several centuries. There have been considerable advances made in the H2S field regarding its physiological role; however, there is much more work that needs to be done. The biosynthesis of H2S has been attributed to three endogenous enzymes: cystathionine β‐synthase (CBS), cystathionine γ‐lyase (CGL or CSE) and 3‐mercaptopyruvate sulfurtransferase (3‐MST). These enzymes require further investigation to more fully elucidate the cellular expression profile, regulation and precise role of these critical enzymes in the production of H2S. In recent years, H2S has been demonstrated to have cytoprotective effects in multiple organ systems. In particular, it has been demonstrated that the administration of H2S either prior to ischaemia or at reperfusion significantly ameliorates myocardial and hepatic ischaemia–reperfusion injury. Therefore, this review focuses on the cardioprotective and hepatoprotective role of H2S. In addition, the review provides a summary of several known molecular targets of H2S protection.


Antioxidants & Redox Signaling | 2011

Chronic Exposure to a High-Fat Diet Induces Hepatic Steatosis, Impairs Nitric Oxide Bioavailability, and Modifies the Mitochondrial Proteome in Mice

Heather B. Eccleston; Kelly K. Andringa; Angela M. Betancourt; Adrienne L. King; Sudheer K. Mantena; Telisha M. Swain; Heather N. Tinsley; Ryan N. Nolte; Tim R. Nagy; Gary A. Abrams; Shannon M. Bailey

Obesity-related pathologies, such as nonalcoholic fatty liver disease, are linked to mitochondrial dysfunction and nitric oxide (NO) deficiency. Herein, we tested the hypothesis that a high-fat diet (HFD) modifies the liver mitochondrial proteome and alters proteins involved in NO metabolism, namely arginase 1 and endothelial NO synthase. Male C57BL/6 mice were fed a control or HFD and liver mitochondria were isolated for proteomics and reactive oxygen species measurements. Steatosis and hepatocyte ballooning were present in livers of HFD mice, with no pathology observed in the controls. HFD mice had increased serum glucose and decreased adiponectin. Mitochondrial reactive oxygen species was increased after 8 weeks in the HFD mice, but decreased at 16 weeks compared with the control, which was accompanied by increased uncoupling protein 2. Using proteomics, 22 proteins were altered as a consequence of the HFD. This cohort consists of oxidative phosphorylation, lipid metabolism, sulfur amino acid metabolism, and chaperone proteins. We observed a HFD-dependent increase in arginase 1 and decrease in activated endothelial NO synthase. Serum and liver nitrate + nitrite were decreased by HFD. In summary, these data demonstrate that a HFD causes steatosis, alters NO metabolism, and modifies the liver mitochondrial proteome; thus, NO may play an important role in the processes responsible for nonalcoholic fatty liver disease.


International journal of hepatology | 2012

Betaine Treatment Attenuates Chronic Ethanol-Induced Hepatic Steatosis and Alterations to the Mitochondrial Respiratory Chain Proteome

Kusum K. Kharbanda; Sandra L. Todero; Adrienne L. King; Natalia A. Osna; Benita L. McVicker; Dean J. Tuma; James L. Wisecarver; Shannon M. Bailey

Introduction. Mitochondrial damage and disruption in oxidative phosphorylation contributes to the pathogenesis of alcoholic liver injury. Herein, we tested the hypothesis that the hepatoprotective actions of betaine against alcoholic liver injury occur at the level of the mitochondrial proteome. Methods. Male Wister rats were pair-fed control or ethanol-containing liquid diets supplemented with or without betaine (10 mg/mL) for 4-5 wks. Liver was examined for triglyceride accumulation, levels of methionine cycle metabolites, and alterations in mitochondrial proteins. Results. Chronic ethanol ingestion resulted in triglyceride accumulation which was attenuated in the ethanol plus betaine group. Blue native gel electrophoresis (BN-PAGE) revealed significant decreases in the content of the intact oxidative phosphorylation complexes in mitochondria from ethanol-fed animals. The alcohol-dependent loss in many of the low molecular weight oxidative phosphorylation proteins was prevented by betaine supplementation. This protection by betaine was associated with normalization of SAM : S-adenosylhomocysteine (SAH) ratios and the attenuation of the ethanol-induced increase in inducible nitric oxide synthase and nitric oxide generation in the liver. Discussion/Conclusion. In summary, betaine attenuates alcoholic steatosis and alterations to the oxidative phosphorylation system. Therefore, preservation of mitochondrial function may be another key molecular mechanism responsible for betaine hepatoprotection.


American Journal of Physiology-gastrointestinal and Liver Physiology | 2010

Analysis of the liver mitochondrial proteome in response to ethanol and S-adenosylmethionine treatments: novel molecular targets of disease and hepatoprotection

Kelly K. Andringa; Adrienne L. King; Heather B. Eccleston; Sudheer K. Mantena; Aimee Landar; Nirag Jhala; Dale A. Dickinson; Giuseppe L. Squadrito; Shannon M. Bailey

S-adenosylmethionine (SAM) minimizes alcohol hepatotoxicity; however, the molecular mechanisms responsible for SAM hepatoprotection remain unknown. Herein, we use proteomics to determine whether the hepatoprotective action of SAM against early-stage alcoholic liver disease is linked to alterations in the mitochondrial proteome. For this, male rats were fed control or ethanol-containing liquid diets +/- SAM and liver mitochondria were prepared for proteomic analysis. Two-dimensional isoelectric focusing (2D IEF/SDS-PAGE) and blue native gel electrophoresis (BN-PAGE) were used to determine changes in matrix and oxidative phosphorylation (OxPhos) proteins, respectively. SAM coadministration minimized alcohol-dependent inflammation and preserved mitochondrial respiration. SAM supplementation preserved liver SAM levels in ethanol-fed rats; however, mitochondrial SAM levels were increased by ethanol and SAM treatments. With use of 2D IEF/SDS-PAGE, 30 proteins showed significant changes in abundance in response to ethanol, SAM, or both. Classes of proteins affected by ethanol and SAM treatments were chaperones, beta oxidation proteins, sulfur metabolism proteins, and dehydrogenase enzymes involved in methionine, glycine, and choline metabolism. BN-PAGE revealed novel changes in the levels of 19 OxPhos proteins in response to ethanol, SAM, or both. Ethanol- and SAM-dependent alterations in the proteome were not linked to corresponding changes in gene expression. In conclusion, ethanol and SAM treatment led to multiple changes in the liver mitochondrial proteome. The protective effects of SAM against alcohol toxicity are mediated, in part, through maintenance of proteins involved in key mitochondrial energy conserving and biosynthetic pathways. This study demonstrates that SAM may be a promising candidate for treatment of alcoholic liver disease.


Nature Communications | 2015

CD163 interacts with TWEAK to regulate tissue regeneration after ischaemic injury.

Hirokuni Akahori; Vinit Karmali; Rohini Polavarapu; Alicia N. Lyle; Daiana Weiss; Eric Shin; Ahsan Husain; Nawazish Naqvi; Richard Van Dam; Anwer Habib; Cheol Ung Choi; Adrienne L. King; Kimberly Pachura; W. Robert Taylor; David J. Lefer; Aloke V. Finn

Macrophages are an essential component of the immune response to ischaemic injury and play an important role in promoting inflammation and its resolution, which is necessary for tissue repair. The type I transmembrane glycoprotein CD163 is exclusively expressed on macrophages, where it acts as a receptor for haemoglobin:haptoglobin complexes. An extracellular portion of CD163 circulates in the blood as a soluble protein, for which no physiological function has so far been described. Here we show that during ischaemia, soluble CD163 functions as a decoy receptor for TWEAK, a secreted pro-inflammatory cytokine of the tumour necrosis factor family, to regulate TWEAK-induced activation of canonical nuclear factor-κB (NF-κB) and Notch signalling necessary for myogenic progenitor cell proliferation. Mice with deletion of CD163 have transiently elevated levels of TWEAK, which stimulate muscle satellite cell proliferation and tissue regeneration in their ischaemic and non-ischaemic limbs. These results reveal a role for soluble CD163 in regulating muscle regeneration after ischaemic injury.

Collaboration


Dive into the Adrienne L. King's collaboration.

Top Co-Authors

Avatar

Shannon M. Bailey

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kelly K. Andringa

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sudheer K. Mantena

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Heather B. Eccleston

University of Alabama at Birmingham

View shared research outputs
Researchain Logo
Decentralizing Knowledge