Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Arch Anim Breed ; 66(1): 103-111, 2023.
Article in English | MEDLINE | ID: mdl-37312865

ABSTRACT

Due to the legislation of antibiotic usage, natural substances are required for application in the poultry industry. Because of their potential anti-inflammatory and immunomodulatory effects, carotenoids are great sources. Capsanthin, a major carotenoid giving the red color of pepper, is a promising feed additive, as it can reduce chronic inflammation. This study was conducted to determine the effects of capsanthin supplementation at 80 mg kg-1 in feed on the immune response of broiler chickens under Escherichia coli O55:B5 lipopolysaccharide (LPS) challenge. Ross 308 male broilers were divided into treatments: control (basal diet) and feed-supplemented groups. At 42 d of age, chickens were weighed and then challenged with 1 mg LPS per kilogram of body weight intraperitoneally. Four hours after injection, birds were euthanized, and then spleen and blood samples were collected. Capsanthin supplement at 80 mg kg-1 did not change the growth parameters and the relative spleen weight. LPS immunization resulted in higher splenic interleukin-1ß (IL-1ß), interleukin-6 (IL-6), and interferon-γ (IFN-γ) mRNA expressions. Capsanthin addition reached lower gene expression levels of IL-6 and IFN-γ compared to the LPS-injected birds. At plasma level, dietary capsanthin resulted in lower IL-1ß and IL-6 levels. These results may indicate the potential anti-inflammatory effect of capsanthin supplementation in broiler chickens.

2.
J Exp Med ; 219(7)2022 07 04.
Article in English | MEDLINE | ID: mdl-35616614

ABSTRACT

Given the clinical, economic, and societal impact of obesity, unraveling the mechanisms of adipose tissue expansion remains of fundamental significance. We previously showed that white adipose tissue (WAT) levels of 3-mercaptopyruvate sulfurtransferase (MPST), a mitochondrial cysteine-catabolizing enzyme that yields pyruvate and sulfide species, are downregulated in obesity. Here, we report that Mpst deletion results in fat accumulation in mice fed a high-fat diet (HFD) through transcriptional and metabolic maladaptation. Mpst-deficient mice on HFD exhibit increased body weight and inguinal WAT mass, reduced metabolic rate, and impaired glucose/insulin tolerance. At the molecular level, Mpst ablation activates HIF1α, downregulates subunits of the translocase of outer/inner membrane (TIM/TOM) complex, and impairs mitochondrial protein import. MPST deficiency suppresses the TCA cycle, oxidative phosphorylation, and fatty acid oxidation, enhancing lipid accumulation. Sulfide donor administration to obese mice reverses the HFD-induced changes. These findings reveal the significance of MPST for white adipose tissue biology and metabolic health and identify a potential new therapeutic target for obesity.


Subject(s)
Glucose Intolerance , Sulfurtransferases , Animals , Diet, High-Fat , Energy Metabolism , Mice , Mice, Inbred C57BL , Mitochondrial Proteins/metabolism , Obesity/metabolism , Sulfides , Sulfurtransferases/metabolism
3.
Animals (Basel) ; 12(3)2022 Feb 05.
Article in English | MEDLINE | ID: mdl-35158709

ABSTRACT

Heat stress (HS) can have detrimental effects on intestinal integrity and can jeopardize the digestibility performance in pigs. With prolonged exposure to heat, some thermoregulatory processes in pigs are potential causes for electrolyte imbalance. The adverse effects of HS on mineral digestibility and electrolyte balance are not widely studied and information on its abatement through vitamin and micro-mineral supplementation in combinations above the recommended level in pigs is limited. The aim of this study is to research this area. Thirty-six Danbred hybrid barrows (65.1 ± 2.81kg) were distributed among the four treatments (n = 9 per treatment): (1) thermo-neutral (19.5 ± 0.9 °C, RH- 85.9 ± 7.3%)+ control diet (TC) (NRC, 2012), (2) HS (28.9 ± 0.9 °C, RH- 60.4 ± 4.3%) + control diet (HC), (3) HS +diet with elevated levels of vitamins (vitamin E and C) and micro-minerals (Zn and Se) (HT1), and (4) HS + diet with further elevation of vitamins and micro-minerals (HT2). Plasma samples were collected on days 7 and 21 of the experiment to investigate electrolyte concentration. During the experimental period, feces samples were collected from pigs placed in digestibility cages (six pigs from each treatment) to investigate the digestibility of Ca, P, Na, Se, and Zn. HS did not decrease the digestibility of minerals, but elevated supplementation of the selected vitamins and trace minerals improved it significantly. HS caused a significant decrease of Cl- (p < 0.01) in plasma, indicating an imbalance. In conclusion, pigs can have some resilience against heat stress in terms of mineral digestibility. Proper vitamin and trace mineral supplementation are key factors in the ability of pigs to overcome the negative effects of HS.

4.
Proc Natl Acad Sci U S A ; 118(20)2021 05 18.
Article in English | MEDLINE | ID: mdl-33972444

ABSTRACT

In mammalian cells, cyanide is viewed as a cytotoxic agent, which exerts its effects through inhibition of mitochondrial Complex IV (Cytochrome C oxidase [CCOx]). However, the current report demonstrates that cyanide's effect on CCOx is biphasic; low (nanomolar to low-micromolar) concentrations stimulate CCOx activity, while higher (high-micromolar) concentrations produce the "classic" inhibitory effect. Low concentrations of cyanide stimulated mitochondrial electron transport and elevated intracellular adenosine triphosphate (ATP), resulting in the stimulation of cell proliferation. The stimulatory effect of cyanide on CCOx was associated with the removal of the constitutive, inhibitory glutathionylation on its catalytic 30- and 57-kDa subunits. Transfer of diluted Pseudomonas aeruginosa (a cyanide-producing bacterium) supernatants to mammalian cells stimulated cellular bioenergetics, while concentrated supernatants were inhibitory. These effects were absent with supernatants from mutant Pseudomonas lacking its cyanide-producing enzyme. These results raise the possibility that cyanide at low, endogenous levels serves regulatory purposes in mammals. Indeed, the expression of six putative mammalian cyanide-producing and/or -metabolizing enzymes was confirmed in HepG2 cells; one of them (myeloperoxidase) showed a biphasic regulation after cyanide exposure. Cyanide shares features with "classical" mammalian gasotransmitters NO, CO, and H2S and may be considered the fourth mammalian gasotransmitter.


Subject(s)
Cyanides/pharmacology , Electron Transport Complex IV/drug effects , Energy Metabolism/drug effects , Mitochondria/drug effects , Cyanides/metabolism , Electron Transport Complex IV/physiology , HCT116 Cells , HT29 Cells , Humans , Mitochondria/metabolism
5.
Mol Med ; 26(1): 102, 2020 11 09.
Article in English | MEDLINE | ID: mdl-33167881

ABSTRACT

Clinical observations and preclinical studies both suggest that Down syndrome (DS) may be associated with significant metabolic and bioenergetic alterations. However, the relevant scientific literature has not yet been systematically reviewed. The aim of the current study was to conduct a meta-analysis of metabolites involved in bioenergetics pathways in DS to conclusively determine the difference between DS and control subjects. We discuss these findings and their potential relevance in the context of pathogenesis and experimental therapy of DS. Articles published before July 1, 2020, were identified by using the search terms "Down syndrome" and "metabolite name" or "trisomy 21" and "metabolite name". Moreover, DS-related metabolomics studies and bioenergetics literature were also reviewed. 41 published reports and associated databases were identified, from which the descriptive information and the relevant metabolomic parameters were extracted and analyzed. Mixed effect model revealed the following changes in DS: significantly decreased ATP, CoQ10, homocysteine, serine, arginine and tyrosine; slightly decreased ADP; significantly increased uric acid, succinate, lactate and cysteine; slightly increased phosphate, pyruvate and citrate. However, the concentrations of AMP, 2,3-diphosphoglycerate, glucose, and glutamine were comparable in the DS vs. control populations. We conclude that cells of subjects with DS are in a pseudo-hypoxic state: the cellular metabolic and bio-energetic mechanisms exhibit pathophysiological alterations that resemble the cellular responses associated with hypoxia, even though the supply of the cells with oxygen is not disrupted. This fundamental alteration may be, at least in part, responsible for a variety of functional deficits associated with DS, including reduced exercise difference, impaired neurocognitive status and neurodegeneration.


Subject(s)
Down Syndrome/metabolism , Energy Metabolism , Metabolic Networks and Pathways , Oxygen/metabolism , Animals , Biomarkers , Disease Management , Disease Susceptibility , Down Syndrome/diagnosis , Down Syndrome/etiology , Down Syndrome/therapy , Gene Expression Profiling/methods , Gene Expression Regulation , Glucose/metabolism , Humans , Hypoxia/metabolism , Mammals , Metabolomics/methods , Mitochondria/genetics , Mitochondria/metabolism , Oxidative Phosphorylation , Proteomics/methods , Treatment Outcome
6.
Animals (Basel) ; 10(8)2020 Aug 11.
Article in English | MEDLINE | ID: mdl-32796555

ABSTRACT

Determination of wool mineral content to assess the animal' mineral status has been extensively used, but the results are controversial. One of the possible contributing factors is that the sampling material in previous studies was collected from a long staple, a fact that could mask the response to recent differences in mineral intake. Therefore, the aim of the present study was to test the sensitiveness of newly grown wool to different dietary mineral intake. Twenty Tsigai ewes were allocated into five dietary treatments with similar hay and concentrate intake but different premix inclusion rates in the concentrate (3, 4, 5, 6, and 7%). Wool was sampled on the left side from a 5 × 5 cm area using bent scissors at the beginning of the trial and from the very same area 28 days later. Samples after cleaning and mineralization were analyzed with ICP-OES (Perkin-Elmer, Optima 3300 DV) for calcium, phosphorus, magnesium, sodium, selenium, zinc, copper, and sulfur content. Long fleeces had significantly lower Ca and Se content compared to the newly grown wool samples of the group at the premix manufacturer's suggested level of supplementation (5%). Macrominerals in fresh wool did not respond to increased dietary supplementation. Se and Zn content of wool had a strong relationship with the daily intake (R2 = 0.95 and R2 = 0.97, respectively.) In conclusion, the mineral content of long fleeces can be different compared to recently developed wool fiber. This indicates that, in some cases, analyzing long staples for mineral status can be misleading. Our results showed that wool could be a sensitive indicator of low selenium and high zinc intake. Mineral interactions can significantly affect the actual availability of trace minerals; therefore, a more careful design of premixes is needed. The described method seems to be applicable in livestock farming, but the mineral interactions that may alter the results need to be further explored.

7.
Br J Pharmacol ; 177(4): 866-883, 2020 02.
Article in English | MEDLINE | ID: mdl-30644090

ABSTRACT

BACKGROUND AND PURPOSE: During angiogenesis, quiescent endothelial cells (ECs) are activated by various stimuli to form new blood vessels from pre-existing ones in physiological and pathological conditions. Many research groups have shown that hydrogen sulfide (H2 S), the newest member of the gasotransmitter family, acts as a proangiogenic factor. To date, very little is known about the regulatory role of 3-mercaptopyruvate sulfurtransferase (3-MST), an important H2 S-producing enzyme in ECs. The aim of our study was to explore the potential role of 3-MST in human EC bioenergetics, metabolism, and angiogenesis. EXPERIMENTAL APPROACH: To assess in vitro angiogenic responses, we used EA.hy926 human vascular ECs subjected to shRNA-mediated 3-MST attenuation and pharmacological inhibition of proliferation, migration, and tube-like network formation. To evaluate bioenergetic parameters, cell respiration, glycolysis, glucose uptake, and mitochondrial/glycolytic ATP production were measured. Finally, global metabolomic profiling was performed to determine the level of 669 metabolic compounds. KEY RESULTS: 3-MST-attenuated ECs subjected to shRNA or pharmacological inhibition of 3-MST significantly reduced EC proliferation, migration, and tube-like network formation. 3-MST silencing also suppressed VEGF-induced EC migration. From bioenergetic and metabolic standpoints, 3-MST attenuation decreased mitochondrial respiration and mitochondrial ATP production, increased glucose uptake, and perturbed the entire EC metabolome. CONCLUSION AND IMPLICATIONS: 3-MST regulates bioenergetics and morphological angiogenic functions in human ECs. The data presented in the current report support the view that 3-MST pathway may be a potential candidate for therapeutic modulation of angiogenesis. LINKED ARTICLES: This article is part of a themed section on Hydrogen Sulfide in Biology & Medicine. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.4/issuetoc.


Subject(s)
Endothelial Cells , Hydrogen Sulfide , Sulfurtransferases/metabolism , Endothelial Cells/metabolism , Energy Metabolism , Humans
8.
Geroscience ; 41(4): 419-439, 2019 08.
Article in English | MEDLINE | ID: mdl-31463647

ABSTRACT

Understanding molecular mechanisms involved in vascular aging is essential to develop novel interventional strategies for treatment and prevention of age-related vascular pathologies. Recent studies provide critical evidence that vascular aging is characterized by NAD+ depletion. Importantly, in aged mice, restoration of cellular NAD+ levels by treatment with the NAD+ booster nicotinamide mononucleotide (NMN) exerts significant vasoprotective effects, improving endothelium-dependent vasodilation, attenuating oxidative stress, and rescuing age-related changes in gene expression. Strong experimental evidence shows that dysregulation of microRNAs (miRNAs) has a role in vascular aging. The present study was designed to test the hypothesis that age-related NAD+ depletion is causally linked to dysregulation of vascular miRNA expression. A corollary hypothesis is that functional vascular rejuvenation in NMN-treated aged mice is also associated with restoration of a youthful vascular miRNA expression profile. To test these hypotheses, aged (24-month-old) mice were treated with NMN for 2 weeks and miRNA signatures in the aortas were compared to those in aortas obtained from untreated young and aged control mice. We found that protective effects of NMN treatment on vascular function are associated with anti-aging changes in the miRNA expression profile in the aged mouse aorta. The predicted regulatory effects of NMN-induced differentially expressed miRNAs in aged vessels include anti-atherogenic effects and epigenetic rejuvenation. Future studies will uncover the mechanistic role of miRNA gene expression regulatory networks in the anti-aging effects of NAD+ booster treatments and determine the links between miRNAs regulated by NMN and sirtuin activators and miRNAs known to act in the conserved pathways of aging and major aging-related vascular diseases.


Subject(s)
Aging/metabolism , Aorta/metabolism , Dietary Supplements , Endothelium, Vascular/metabolism , MicroRNAs/metabolism , Nicotinamide Mononucleotide/pharmacology , Aging/genetics , Animals , Atherosclerosis , Epigenesis, Genetic , Gene Expression Profiling , Mice, Inbred C57BL , Rejuvenation
9.
Br J Pharmacol ; 175(2): 284-300, 2018 01.
Article in English | MEDLINE | ID: mdl-27723079

ABSTRACT

BACKGROUND AND PURPOSE: We hypothesized that an in vitro, stretch-based model of neural injury may be useful to identify compounds that decrease the cellular damage in neurotrauma. EXPERIMENTAL APPROACH: We screened three neural cell lines (B35, RN33B and SH-SY5Y) subjected to two differentiation methods and selected all-trans-retinoic acid-differentiated B35 rat neuroblastoma cells subjected to rapid stretch injury, coupled with a subthreshold concentration of H2 O2 , for the screen. The model induced marked alterations in gene expression and proteomic signature of the cells and culminated in delayed cell death (LDH release) and mitochondrial dysfunction [reduced 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) conversion]. Follow-up studies utilized human stem cell-derived neurons subjected to rapid stretch injury. KEY RESULTS: From screening of a composite library of 3500 drugs, five drugs (when applied in a post-treatment regimen relative to stretch injury) improved both LDH and MTT responses. The effects of rifampicin were investigated in further detail. Rifampicin reduced cell necrosis and apoptosis and improved cellular bioenergetics. In a second model (stretch injury in human stem cell-derived neurons), rifampicin pretreatment attenuated LDH release, protected against the loss of neurite length and maintained neuron-specific class III ß-tubulin immunoreactivity. CONCLUSIONS AND IMPLICATIONS: We conclude that the current model is suitable for medium-throughput screening to identify compounds with neuroprotective potential. Rifampicin, when applied either in pre- or post-treatment, improves the viability of neurons subjected to stretch injury and protects against neurite loss. Rifampicin may be a candidate for repurposing for the therapy of traumatic brain injury. LINKED ARTICLES: This article is part of a themed section on Inventing New Therapies Without Reinventing the Wheel: The Power of Drug Repurposing. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.2/issuetoc.


Subject(s)
Brain Injuries, Traumatic/drug therapy , Rifampin/pharmacology , Rifampin/therapeutic use , Animals , Apoptosis/drug effects , Brain Injuries, Traumatic/metabolism , Cell Death/drug effects , Cell Line, Tumor , Disease Models, Animal , Drug Evaluation, Preclinical/methods , Humans , Hydrogen Peroxide , L-Lactate Dehydrogenase/metabolism , Mitochondria/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Stress, Mechanical , Tetrazolium Salts/metabolism
10.
Mol Pharmacol ; 92(6): 718-730, 2017 12.
Article in English | MEDLINE | ID: mdl-29030392

ABSTRACT

Hydrogen sulfide (H2S) exhibits beneficial effects in the cardiovascular system, many of which depend on nitric oxide (NO). Proline-rich tyrosine kinase 2 (PYK2), a redox-sensitive tyrosine kinase, directly phosphorylates and inhibits endothelial NO synthase (eNOS). We investigated the ability of H2S to relieve PYK2-mediated eNOS inhibition and evaluated the importance of the H2S/PYK2/eNOS axis on cardiomyocyte injury in vitro and in vivo. Exposure of H9c2 cardiomyocytes to H2O2 or pharmacologic inhibition of H2S production increased PYK2 (Y402) and eNOS (Y656) phosphorylation. These effects were blocked by treatment with Na2S or by overexpression of cystathionine γ-lyase (CSE). In addition, PYK2 overexpression reduced eNOS activity in a H2S-reversible manner. The viability of cardiomyocytes exposed to Η2Ο2 was reduced and declined further after the inhibition of H2S production. PYK2 downregulation, l-cysteine supplementation, or CSE overexpression alleviated the effects of H2O2 on H9c2 cardiomyocyte survival. Moreover, H2S promoted PYK2 sulfhydration and inhibited its activity. In vivo, H2S administration reduced reactive oxygen species levels, as well as PYK2 (Y402) and eNOS (Y656) phosphorylation. Pharmacologic blockade of PYK2 or inhibition of PYK2 activation by Na2S reduced myocardial infarct size in mice. Coadministration of a PYK2 inhibitor and Na2S did not result in additive effects on infarct size. We conclude that H2S relieves the inhibitory effect of PYK2 on eNOS, allowing the latter to produce greater amounts of NO, thereby affording cardioprotection. Our results unravel the existence of a novel H2S-NO interaction and identify PYK2 as a crucial target for the protective effects of H2S under conditions of oxidative stress.


Subject(s)
Cardiotonic Agents/pharmacology , Focal Adhesion Kinase 2/antagonists & inhibitors , Hydrogen Sulfide/pharmacology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Nitric Oxide Synthase Type III/metabolism , Proline/metabolism , Animals , Cell Line , Cystathionine gamma-Lyase/metabolism , Cysteine/metabolism , HEK293 Cells , Humans , Hydrogen Peroxide/pharmacology , Male , Mice , Mice, Inbred C57BL , Nitric Oxide/metabolism , Rats
11.
ACS Chem Biol ; 12(8): 2117-2123, 2017 08 18.
Article in English | MEDLINE | ID: mdl-28613823

ABSTRACT

Hydrogen sulfide (H2S) is an important biological mediator, and synthetic H2S donating molecules provide an important class of investigative tools for H2S research. Here, we report esterase-activated H2S donors that function by first releasing carbonyl sulfide (COS), which is rapidly converted to H2S by the ubiquitous enzyme carbonic anhydrase (CA). We report the synthesis, self-immolative decomposition, and H2S release profiles of the developed scaffolds. In addition, the developed esterase-triggered COS/H2S donors exhibit higher levels of cytotoxicity than equivalent levels of Na2S or the common H2S donors GYY4137 and AP39. Using cellular bioenergetics measurements, we establish that the developed donors reduce cellular respiration and ATP synthesis in BEAS 2B human lung epithelial cells, which is consistent with COS/H2S inhibition of cytochrome c oxidase in the mitochondrial respiratory chain although not observed with common H2S donors at the same concentrations. Taken together, these results may suggest that COS functions differently than H2S in certain biological contexts or that the developed donors are more efficient at delivering H2S than other common H2S-releasing motifs.


Subject(s)
Benzyl Compounds/pharmacology , Energy Metabolism/drug effects , Esterases/pharmacology , Hydrogen Sulfide/metabolism , Mitochondria/drug effects , Sulfur Oxides/metabolism , Thiocarbamates/pharmacology , Benzyl Compounds/chemistry , Cell Survival/drug effects , Epithelial Cells/drug effects , Esterases/metabolism , Humans , Lung/drug effects , Magnetic Resonance Spectroscopy , Molecular Structure , Thiocarbamates/chemistry
12.
Biochem Pharmacol ; 136: 86-98, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28404377

ABSTRACT

Cystathionine-ß-synthase (CBS) is upregulated and hydrogen sulfide (H2S) production is increased in colon cancer cells. The functional consequence of this response is stimulation of cellular bioenergetics and tumor growth and proliferation. Lactate dehydrogenase A (LDHA) is also upregulated in various colon cancer cells and has been previously implicated in tumor cell bioenergetics and proliferation. In the present study, we sought to determine the potential interaction between the H2S pathway and LDH activity in the control of bioenergetics and proliferation of colon cancer, using the colon cancer line HCT116. Low concentrations of GYY4137 (a slow-releasing H2S donor) enhanced mitochondrial function (oxygen consumption, ATP production, and spare respiratory capacity) and glycolysis in HCT116 cells. SiRNA-mediated transient silencing of LDHA attenuated the GYY4137-induced stimulation of mitochondrial respiration, but not of glycolysis. H2S induced the S-sulfhydration of Cys163 in recombinant LDHA, and stimulated LDHA activity. The H2S-induced stimulation of LDHA activity was absent in C163A LDHA. As shown in HCT116 cell whole extracts, in addition to LDHA activation, GYY4137 also stimulated LDHB activity, although to a smaller extent. Total cellular lactate and pyruvate measurements showed that in HCT116 cells LDHA catalyzes the conversion of pyruvate to lactate. Total cellular lactate levels were increased by GYY4137 in wild-type cells (but not in cells with LDHA silencing). LDHA silencing sensitized HCT116 cells to glucose oxidase (GOx)-induced oxidative stress; this was further exacerbated with GYY4137 treatment. Treatment with low concentrations of GYY4137 (0.3mM) or GOx (0.01U/ml) significantly increased the proliferation rate of HCT116 cells; the effect of GOx, but not the effect of GYY4137 was attenuated by LDHA silencing. The current report points to the involvement of LDHA in the stimulatory effect of H2S on mitochondrial respiration in colon cancer cells and characterizes some of the functional interactions between LDHA and H2S-stimulated bioenergetics under resting conditions, as well as during oxidative stress.


Subject(s)
Colonic Neoplasms/metabolism , Energy Metabolism/drug effects , Energy Metabolism/physiology , Hydrogen Sulfide/pharmacology , L-Lactate Dehydrogenase/metabolism , Cell Proliferation/drug effects , Cell Proliferation/physiology , Dose-Response Relationship, Drug , HCT116 Cells , Humans , Isoenzymes/metabolism , Lactate Dehydrogenase 5 , Morpholines/pharmacology , Organothiophosphorus Compounds/pharmacology , Oxygen Consumption/drug effects , Oxygen Consumption/physiology
13.
Sci Rep ; 6: 36125, 2016 11 03.
Article in English | MEDLINE | ID: mdl-27808278

ABSTRACT

Therapeutic manipulation of the gasotransmitter hydrogen sulfide (H2S) has recently been proposed as a novel targeted anticancer approach. Here we show that human lung adenocarcinoma tissue expresses high levels of hydrogen sulfide (H2S) producing enzymes, namely, cystathionine beta-synthase (CBS), cystathionine gamma lyase (CSE) and 3-mercaptopyruvate sulfurtransferase (3-MST), in comparison to adjacent lung tissue. In cultured lung adenocarcinoma but not in normal lung epithelial cells elevated H2S stimulates mitochondrial DNA repair through sulfhydration of EXOG, which, in turn, promotes mitochondrial DNA repair complex assembly, thereby enhancing mitochondrial DNA repair capacity. In addition, inhibition of H2S-producing enzymes suppresses critical bioenergetics parameters in lung adenocarcinoma cells. Together, inhibition of H2S-producing enzymes sensitize lung adenocarcinoma cells to chemotherapeutic agents via induction of mitochondrial dysfunction as shown in in vitro and in vivo models, suggesting a novel mechanism to overcome tumor chemoresistance.


Subject(s)
Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Antineoplastic Agents/therapeutic use , DNA Repair , Energy Metabolism , Hydrogen Sulfide/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Mitochondria/metabolism , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Animals , Antineoplastic Agents/pharmacology , DNA Repair/drug effects , DNA, Mitochondrial/genetics , Disease Models, Animal , Energy Metabolism/drug effects , Lung Neoplasms/pathology , Mitochondria/drug effects , Models, Biological , Tumor Cells, Cultured
14.
Pharmacol Res ; 113(Pt A): 175-185, 2016 11.
Article in English | MEDLINE | ID: mdl-27569706

ABSTRACT

Recent studies have implicated endogenously produced H2S in the angiogenic process. On one hand, pharmacological inhibition and silencing of the enzymes involved in H2S synthesis attenuate the angiogenic properties of endothelial cells, including proliferation, migration and tube-like structure network formation. On the other hand, enhanced production of H2S by substrate supplementation or over-expression of H2S-producing enzymes leads to enhanced angiogenic responses in cultured endothelial cells. Importantly, H2S up-regulates expression of the key angiogenic factor vascular endothelial growth factor (VEGF) and contributes to the angiogenic signaling in response to VEGF. The signaling pathways mediating H2S-induced angiogenesis include mitogen-activated protein kinases, phosphoinositide-3 kinase, nitric oxide/cGMP-regulated cascades and ATP-sensitive potassium channels. Endogenously produced H2S has also been shown to facilitate neovascularization in prototypical model systems in vivo, and to contribute to wound healing, post-ischemic angiogenesis in the heart and other tissues, as well as in tumor angiogenesis. Targeting of H2S synthesizing enzymes might offer novel therapeutic opportunities for angiogenesis-related diseases.


Subject(s)
Hydrogen Sulfide/metabolism , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Neovascularization, Physiologic/physiology , Animals , Cell Movement/physiology , Cell Proliferation/physiology , Endothelial Cells/metabolism , Humans , Signal Transduction/physiology
15.
Shock ; 45(1): 88-97, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26513708

ABSTRACT

This study evaluated the effects of AP39 [(10-oxo-10-(4-(3-thioxo-3H-1,2-dithiol-5yl) phenoxy)decyl) triphenyl phosphonium bromide], a mitochondrially targeted donor of hydrogen sulfide (H2S) in an in vitro model of hypoxia/oxidative stress injury in NRK-49F rat kidney epithelial cells (NRK cells) and in a rat model of renal ischemia-reperfusion injury. Renal oxidative stress was induced by the addition of glucose oxidase, which generates hydrogen peroxide in the culture medium at a constant rate. Glucose oxidase (GOx)-induced oxidative stress led to mitochondrial dysfunction, decreased intracellular ATP content, and, at higher concentrations, increased intracellular oxidant formation (estimated by the fluorescent probe 2, 7-dichlorofluorescein, DCF) and promoted necrosis (estimated by the measurement of lactate dehydrogenase release into the medium) of the NRK cells in vitro. Pretreatment with AP39 (30-300 nM) exerted a concentration-dependent protective effect against all of the above effects of GOx. Most of the effects of AP39 followed a bell-shaped concentration-response curve; at the highest concentration of GOx tested, AP39 was no longer able to afford cytoprotective effects. Rats subjected to renal ischemia/reperfusion responded with a marked increase (over four-fold over sham control baseline) blood urea nitrogen and creatinine levels in blood, indicative of significant renal damage. This was associated with increased neutrophil infiltration into the kidneys (assessed by the myeloperoxidase assay in kidney homogenates), increased oxidative stress (assessed by the malondialdehyde assay in kidney homogenates), and an increase in plasma levels of IL-12. Pretreatment with AP39 (0.1, 0.2, and 0.3 mg/kg) provided a dose-dependent protection against these pathophysiological alterations; the most pronounced protective effect was observed at the 0.3 mg/kg dose of the H2S donor; nevertheless, AP39 failed to achieve a complete normalization of any of the injury markers measured. The partial protective effects of AP39 correlated with a partial improvement of kidney histological scores and reduced TUNEL staining (an indicator of DNA damage and apoptosis). In summary, the mitochondria-targeted H2S donor AP39 exerted dose-dependent protective effects against renal epithelial cell injury in vitro and renal ischemia-reperfusion injury in vivo. We hypothesize that the beneficial actions of AP39 are related to the reduction of cellular oxidative stress, and subsequent attenuation of various positive feed-forward cycles of inflammatory and oxidative processes.


Subject(s)
Acute Kidney Injury/prevention & control , Cytoprotection/physiology , Hydrogen Sulfide/metabolism , Organophosphorus Compounds/pharmacology , Oxidative Stress/drug effects , Thiones/pharmacology , Animals , Cell Hypoxia/drug effects , Cell Hypoxia/physiology , Cells, Cultured , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Epithelial Cells/drug effects , Male , Mitochondria/drug effects , Mitochondria/metabolism , Organophosphorus Compounds/administration & dosage , Organophosphorus Compounds/therapeutic use , Rats, Sprague-Dawley , Thiones/administration & dosage , Thiones/therapeutic use
16.
Br J Pharmacol ; 172(20): 4817-32, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26218637

ABSTRACT

BACKGROUND AND PURPOSE: Ischaemic heart disease can lead to serious, life-threatening complications. Traditional therapies for ischaemia aim to increase oxygen delivery and reduce the myocardial ATP consumption by increasing the coronary perfusion and by suppressing cardiac contractility, heart rate or blood pressure. An adjunctive treatment option for ischaemia is to improve or optimize myocardial metabolism. EXPERIMENTAL APPROACH: Metabolic suppression in the ischaemic heart is characterized by reduced levels of high-energy molecules: ATP and NAD(+) . Because NAD(+) is required for most metabolic processes that generate ATP, we hypothesized that restoration of NAD(+) would be a prerequisite for ATP regeneration and examined the role of the major NAD(+) anabolic and catabolic pathways in the bioenergetic restoration process following oxygen-glucose deprivation injury in a cardiomyocyte cell line (H9c2 cells). KEY RESULTS: Salvage of NAD(+) via nicotinamide phosphoribosyl transferase was essential for bioenergetic recovery in cardiomyocytes. Blockade of nicotinamide phosphoribosyl transferase prevented the restoration of the cellular ATP pool following oxygen-glucose deprivation injury by inhibiting both the aerobic and anaerobic metabolism in the cardiomyocytes. NAD(+) consumption by PARP-1 also undermined the recovery processes, and PARP inhibition significantly improved the metabolism and increased cellular ATP levels in cardiomyocytes. CONCLUSIONS AND IMPLICATIONS: We conclude that the NAD(+) salvage pathway is essential for bioenergetic recovery in post-hypoxic cardiomyocytes and PARP inhibition may represent a potential future therapeutic intervention in ischaemic heart disease.


Subject(s)
Hypoxia/metabolism , Myocytes, Cardiac/metabolism , NAD/metabolism , Adenosine Triphosphate/metabolism , Animals , Cell Line , Energy Metabolism , Glucose/deficiency , Hydrogen Peroxide/pharmacology , Membrane Potential, Mitochondrial/drug effects , Oxidants/pharmacology , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Rats , Superoxides/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
17.
Mol Med ; 21: 1-14, 2015 Feb 18.
Article in English | MEDLINE | ID: mdl-25715337

ABSTRACT

Hydrogen sulfide (H2S), as a reducing agent and an antioxidant molecule, exerts protective effects against hyperglycemic stress in the vascular endothelium. The mitochondrial enzyme 3-mercaptopyruvate sulfurtransferase (3-MST) is an important biological source of H2S. We have recently demonstrated that 3-MST activity is inhibited by oxidative stress in vitro and speculated that this may have an adverse effect on cellular homeostasis. In the current study, given the importance of H2S as a vasorelaxant, angiogenesis stimulator and cellular bioenergetic mediator, we first determined whether the 3-MST/H2S system plays a physiological regulatory role in endothelial cells. Next, we tested whether a dysfunction of this pathway develops during the development of hyperglycemia and µmol/L to diabetes-associated vascular complications. Intraperitoneal (IP) 3-MP (1 mg/kg) raised plasma H2S levels in rats. 3-MP (10 1 mmol/L) promoted angiogenesis in vitro in bEnd3 microvascular endothelial cells and in vivo in a Matrigel assay in mice (0.3-1 mg/kg). In vitro studies with bEnd3 cell homogenates demonstrated that the 3-MP-induced increases in H2S production depended on enzymatic activity, although at higher concentrations (1-3 mmol/L) there was also evidence for an additional nonenzymatic H2S production by 3-MP. In vivo, 3-MP facilitated wound healing in rats, induced the relaxation of dermal microvessels and increased mitochondrial bioenergetic function. In vitro hyperglycemia or in vivo streptozotocin diabetes impaired angiogenesis, attenuated mitochondrial function and delayed wound healing; all of these responses were associated with an impairment of the proangiogenic and bioenergetic effects of 3-MP. The antioxidants DL-α-lipoic acid (LA) in vivo, or dihydrolipoic acid (DHLA) in vitro restored the ability of 3-MP to stimulate angiogenesis, cellular bioenergetics and wound healing in hyperglycemia and diabetes. We conclude that diabetes leads to an impairment of the 3-MST/H2S pathway, and speculate that this may contribute to the pathogenesis of hyperglycemic endothelial cell dysfunction. We also suggest that therapy with H2S donors, or treatment with the combination of 3-MP and lipoic acid may be beneficial in improving angiogenesis and bioenergetics in hyperglycemia.


Subject(s)
Endothelium, Vascular/physiology , Energy Metabolism/physiology , Hydrogen Sulfide/metabolism , Metabolic Networks and Pathways , Neovascularization, Physiologic , Sulfurtransferases/metabolism , Animals , Cell Line , Cyclic GMP-Dependent Protein Kinases/metabolism , Cysteine/administration & dosage , Cysteine/analogs & derivatives , Cysteine/pharmacology , Diabetes Mellitus/metabolism , Disease Models, Animal , Endothelial Cells , Endothelium, Vascular/drug effects , Energy Metabolism/drug effects , Hydrogen Sulfide/blood , Hyperglycemia/drug therapy , Hyperglycemia/metabolism , Male , Mice , Mitochondria/metabolism , Neovascularization, Physiologic/drug effects , Oxygen Consumption , Proto-Oncogene Proteins c-akt/metabolism , Rats , Sulfurtransferases/genetics , Thioctic Acid/pharmacology , Vasodilator Agents/administration & dosage , Vasodilator Agents/pharmacology
18.
Nitric Oxide ; 41: 120-30, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-24755204

ABSTRACT

The purpose of the current study was to investigate the effect of the recently synthesized mitochondrially-targeted H2S donor, AP39 [(10-oxo-10-(4-(3-thioxo-3H-1,2-dithiol-5yl)phenoxy)decyl) triphenylphosphonium bromide], on bioenergetics, viability, and mitochondrial DNA integrity in bEnd.3 murine microvascular endothelial cells in vitro, under normal conditions, and during oxidative stress. Intracellular H2S was assessed by the fluorescent dye 7-azido-4-methylcoumarin. For the measurement of bioenergetic function, the XF24 Extracellular Flux Analyzer was used. Cell viability was estimated by the combination of the MTT and LDH methods. Oxidative protein modifications were measured by the Oxyblot method. Reactive oxygen species production was monitored by the MitoSOX method. Mitochondrial and nuclear DNA integrity were assayed by the Long Amplicon PCR method. Oxidative stress was induced by addition of glucose oxidase. Addition of AP39 (30-300 nM) to bEnd.3 cells increased intracellular H2S levels, with a preferential response in the mitochondrial regions. AP39 exerted a concentration-dependent effect on mitochondrial activity, which consisted of a stimulation of mitochondrial electron transport and cellular bioenergetic function at lower concentrations (30-100 nM) and an inhibitory effect at the higher concentration of 300 nM. Under oxidative stress conditions induced by glucose oxidase, an increase in oxidative protein modification and an enhancement in MitoSOX oxidation was noted, coupled with an inhibition of cellular bioenergetic function and a reduction in cell viability. AP39 pretreatment attenuated these responses. Glucose oxidase induced a preferential damage to the mitochondrial DNA; AP39 (100 nM) pretreatment protected against it. In conclusion, the current paper documents antioxidant and cytoprotective effects of AP39 under oxidative stress conditions, including a protection against oxidative mitochondrial DNA damage.


Subject(s)
DNA, Mitochondrial/drug effects , Hydrogen Sulfide/pharmacology , Organophosphates/pharmacology , Organophosphorus Compounds/pharmacology , Oxidative Stress/drug effects , Protective Agents/pharmacology , Thiones/pharmacology , Animals , Cell Line , DNA Repair/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Glucose Oxidase/metabolism , Hydrogen Peroxide/analysis , Hydrogen Peroxide/metabolism , Intracellular Space/drug effects , Mice
19.
Nitric Oxide ; 41: 146-56, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-24667534

ABSTRACT

Recent data show that colon cancer cells selectively overexpress cystathionine-ß-synthase (CBS), which produces hydrogen sulfide (H2S), to maintain cellular bioenergetics, support tumor growth and stimulate angiogenesis and vasorelaxation in the tumor microenvironment. The purpose of the current study was to investigate the effect of the allosteric CBS activator S-adenosyl-L-methionine (SAM) on the proliferation and bioenergetics of the CBS-expressing colon cancer cell line HCT116. The non-transformed, non-tumorigenic colon epithelial cell line NCM356 was used as control. For assessment of cell proliferation, the xCELLigence system was used. Bioenergetic function was measured by Extracellular Flux Analysis. Experiments using human recombinant CBS or HCT116 homogenates complemented the cell-based studies. SAM markedly enhanced CBS-mediated H2S production in vitro, especially when a combination of cysteine and homocysteine was used as substrates. Addition of SAM (0.1-3 mM) to HCT116 cells induced a concentration-dependent increase H2S production. SAM exerted time- and concentration-dependent modulatory effects on cell proliferation. At 0.1-1 mM SAM increased HCT116 proliferation between 0 and 12 h, while the highest SAM concentration (3 mM) inhibited proliferation. Over a longer time period (12-24 h), only the lowest concentration of SAM used (0.1 mM) stimulated cell proliferation; higher SAM concentrations produced a concentration-dependent inhibition. The short-term stimulatory effects of SAM were attenuated by the CBS inhibitor aminooxyacetic acid (AOAA) or by stable silencing of CBS. In contrast, the inhibitory effects of SAM on cell proliferation was unaffected by CBS inhibition or CBS silencing. In contrast to HCT116 cells, the lower rate of proliferation of the low-CBS expressor NCM356 cells was unaffected by SAM. Short-term (1 h) exposure of HCT116 cells to SAM induced a concentration-dependent increase in oxygen consumption and bioenergetic function at 0.1-1 mM, while 3 mM was inhibitory. Longer-term (72 h) exposure of HCT116 cells to all concentrations of SAM tested suppressed mitochondrial oxygen consumption rate, cellular ATP content and cell viability. The stimulatory effect of SAM on bioenergetics was attenuated in cells with stable CBS silencing, while the inhibitory effects were unaffected. In NCM356 cells SAM exerted smaller effects on cellular bioenergetics than in HCT116 cells. We have also observed a downregulation of CBS in response to prolonged exposure of SAM both in HCT116 and NCM356 cells. Taken together, the results demonstrate that H2S production in HCT116 cells is stimulated by the allosteric CBS activator, SAM. At low-to intermediate levels and early time periods the resulting H2S serves as an endogenous cancer cell growth and bioenergetic factor. In contrast, the inhibition of cell proliferation and bioenergetic function by SAM does not appear to relate to adverse autocrine effects of H2S resulting from CBS over-stimulation but, rather to CBS-independent pharmacological effects.


Subject(s)
Cell Proliferation/drug effects , Colorectal Neoplasms/metabolism , Cystathionine beta-Synthase/metabolism , Energy Metabolism/drug effects , S-Adenosylmethionine/pharmacology , Cell Line , Cystathionine beta-Synthase/drug effects , Cystathionine beta-Synthase/genetics , Dose-Response Relationship, Drug , Gene Silencing , HCT116 Cells , Humans , Hydrogen Sulfide/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , RNA, Small Interfering
20.
Br J Pharmacol ; 171(8): 2123-46, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23991749

ABSTRACT

Emerging work demonstrates the dual regulation of mitochondrial function by hydrogen sulfide (H2 S), including, at lower concentrations, a stimulatory effect as an electron donor, and, at higher concentrations, an inhibitory effect on cytochrome C oxidase. In the current article, we overview the pathophysiological and therapeutic aspects of these processes. During cellular hypoxia/acidosis, the inhibitory effect of H2 S on complex IV is enhanced, which may shift the balance of H2 S from protective to deleterious. Several pathophysiological conditions are associated with an overproduction of H2 S (e.g. sepsis), while in other disease states H2 S levels and H2 S bioavailability are reduced and its therapeutic replacement is warranted (e.g. diabetic vascular complications). Moreover, recent studies demonstrate that colorectal cancer cells up-regulate the H2 S-producing enzyme cystathionine ß-synthase (CBS), and utilize its product, H2 S, as a metabolic fuel and tumour-cell survival factor; pharmacological CBS inhibition or genetic CBS silencing suppresses cancer cell bioenergetics and suppresses cell proliferation and cell chemotaxis. In the last chapter of the current article, we overview the field of H2 S-induced therapeutic 'suspended animation', a concept in which a temporary pharmacological reduction in cell metabolism is achieved, producing a decreased oxygen demand for the experimental therapy of critical illness and/or organ transplantation.


Subject(s)
Diabetes Complications/physiopathology , Energy Metabolism/physiology , Gasotransmitters/physiology , Hydrogen Sulfide/metabolism , Mitochondria/physiology , Animals , Energy Metabolism/drug effects , Gasotransmitters/adverse effects , Gasotransmitters/metabolism , Gasotransmitters/pharmacology , Gasotransmitters/therapeutic use , Hibernation/physiology , Humans , Hydrogen Sulfide/adverse effects , Hydrogen Sulfide/pharmacology , Hydrogen Sulfide/therapeutic use , Mitochondria/metabolism , Neoplasms/physiopathology , Shock/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL