Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Biomolecules ; 11(8)2021 07 21.
Article in English | MEDLINE | ID: mdl-34439739

ABSTRACT

Upregulation of hydrogen sulfide (H2S) biosynthesis, at least in part related to the upregulation of cystathionine ß-synthetase (CBS) in cancer cells, serves as a tumor-promoting factor and has emerged as a possible molecular target for antitumor drug development. To facilitate future clinical translation, we have synthesized a variety of novel CBS-targeting, esterase-cleavable prodrugs based on the structure of the prototypical CBS inhibitor aminooxyacetic acid (AOAA). The pharmacological properties of these compounds were evaluated in cell-free assays with recombinant human CBS protein, the human colon cancer cell line HCT116, and in vivo using various tumor-bearing mice models. The prodrug YD0251 (the isopropyl ester derivative of AOAA) was selected for detailed characterization. YD0251 exhibits improved antiproliferative efficacy in cell culture models when compared to AOAA. It is up to 18 times more potent than AOAA at suppressing HCT116 tumor growth in vivo and is effective when administered to tumor-bearing mice either via subcutaneous injection or oral gavage. Patient-derived xenografts (PDTXs) with higher levels of CBS protein grew significantly larger than tumors with lower levels, and YD0251 treatment inhibited the growth of PDTXs with elevated CBS, whereas it had no significant effect on PDTXs with low CBS protein levels. The toxicity of YD0251 was assessed in mice subjected to subchronic administration of supratherapeutic doses the inhibitor; no significant alteration in circulating markers of organ injury or histopathological alterations were noted, up to 60 mg/kg/day × 5 days. In preparation to a future theranostic concept (to match CBS inhibitor therapy to high-CBS expressors), we identified a potential plasma marker of CBS-expressing tumors. Colon cancer cells produced significant levels of lanthionine, a rare metabolic intermediate of CBS-mediated H2S biosynthesis; forced expression of CBS into non-transformed epithelial cells increased lanthionine biogenesis in vitro and in vivo (measured in the urine of tumor-bearing mice). These current results may be useful to facilitate the translation of a CBS inhibition-based antitumor concept into the clinical space.


Subject(s)
Aminooxyacetic Acid/pharmacology , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Colonic Neoplasms/drug therapy , Cystathionine beta-Synthase/antagonists & inhibitors , Prodrugs/pharmacology , Animals , HCT116 Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Nude
2.
Diabetes Obes Metab ; 22(11): 2021-2031, 2020 11.
Article in English | MEDLINE | ID: mdl-32558194

ABSTRACT

AIM: To examine the mechanism of action of γ-aminobutyric acid (GABA) on ß-cell proliferation and investigate if co-treatment with Ly49, a novel GABA type A receptor positive allosteric modulator (GABAA -R PAM), amplifies this effect. METHODS: Human or mouse islets were co-treated for 4-5 days with GABA and selected receptor or cell signalling pathway modulators. Immunofluorescence was used to determine protein co-localization, cell number or proliferation, and islet size. Osmotic minipumps were surgically implanted in mice to assess Ly49 effects on pancreatic ß-cells. RESULTS: Amplification of GABAA -R signalling enhanced GABA-stimulated ß-cell proliferation in cultured mouse islets. Co-treatment of GABA with an inhibitor specific for PI3K, mTORC1/2, or p70S6K, abolished GABA-stimulated ß-cell proliferation in mouse and human islets. Nuclear p-AktSer473 and p-p70S6KThr421/Ser424 expression in pancreatic ß-cells was increased in GABA-treated mice compared with vehicle-treated mice, an effect augmented with GABA and Ly49 co-treatment. Mice co-treated with GABA and Ly49 exhibited enhanced ß-cell area and proliferation compared with GABA-treated mice. Furthermore, S961 injection (an insulin receptor antagonist) resulted in enhanced plasma insulin in GABA and Ly49 co-treated mice compared with GABA-treated mice. Importantly, GABA co-treated with Ly49 increased ß-cell proliferation in human islets providing a potential application for human subjects. CONCLUSIONS: We show that GABA stimulates ß-cell proliferation via the PI3K/mTORC1/p70S6K pathway in both mouse and human islets. Furthermore, we show that Ly49 enhances the ß-cell regenerative effects of GABA, showing potential in the intervention of diabetes.


Subject(s)
Receptors, GABA , Ribosomal Protein S6 Kinases, 70-kDa , Animals , Cell Proliferation , Mechanistic Target of Rapamycin Complex 1 , Mice , gamma-Aminobutyric Acid
3.
FASEB J ; 33(3): 3968-3984, 2019 03.
Article in English | MEDLINE | ID: mdl-30509117

ABSTRACT

γ-Aminobutyric acid (GABA) administration has been shown to increase ß-cell mass, leading to a reversal of type 1 diabetes in mice. Whether GABA has any effect on ß cells of healthy and prediabetic/glucose-intolerant obese mice remains unknown. In the present study, we show that oral GABA administration ( ad libitum) to mice indeed increased pancreatic ß-cell mass, which led to a modest enhancement in insulin secretion and glucose tolerance. However, GABA treatment did not further increase insulin-positive islet area in high fat diet-fed mice and was unable to prevent or reverse glucose intolerance and insulin resistance. Mechanistically, whether in vivo or in vitro, GABA treatment increased ß-cell proliferation. In vitro, the effect was shown to be mediated via the GABAA receptor. Single-cell RNA sequencing analysis revealed that GABA preferentially up-regulated pathways linked to ß-cell proliferation and simultaneously down-regulated those networks required for other processes, including insulin biosynthesis and metabolism. Interestingly, single-cell differential expression analysis revealed GABA treatment gave rise to a distinct subpopulation of ß cells with a unique transcriptional signature, including urocortin 3 ( ucn3), wnt4, and hepacam2. Taken together, this study provides new mechanistic insight into the proliferative nature of GABA but suggests that ß-cell compensation associated with prediabetes overlaps with, and negates, its proliferative effects.-Untereiner, A., Abdo, S., Bhattacharjee, A., Gohil, H., Pourasgari, F., Ibeh, N., Lai, M., Batchuluun, B., Wong, A., Khuu, N., Liu, Y., Al Rijjal, D., Winegarden, N., Virtanen, C., Orser, B. A., Cabrera, O., Varga, G., Rocheleau, J., Dai, F. F., Wheeler, M. B. GABA promotes ß-cell proliferation, but does not overcome impaired glucose homeostasis associated with diet-induced obesity.


Subject(s)
Cell Proliferation , Glucose/metabolism , Insulin-Secreting Cells/metabolism , Obesity/metabolism , Transcriptome , gamma-Aminobutyric Acid/pharmacology , Animals , Cell Line , Cells, Cultured , Diet, High-Fat/adverse effects , Homeostasis , Humans , Insulin/metabolism , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/physiology , Male , Mice , Mice, Inbred C57BL , Obesity/etiology , Receptors, GABA-A/metabolism , Urocortins/metabolism
4.
Biochem Pharmacol ; 149: 174-185, 2018 03.
Article in English | MEDLINE | ID: mdl-29061341

ABSTRACT

Hydrogen sulfide (H2S) production in colon cancer cells supports cellular bioenergetics and proliferation. The aim of the present study was to investigate the alterations in H2S homeostasis during the development of resistance to 5-fluorouracil (5-FU), a commonly used chemotherapeutic agent. A 5-FU-resistant HCT116 human colon cancer cell line was established by serial passage in the presence of increasing 5-FU concentrations. The 5-FU-resistant cells also demonstrated a partial resistance to an unrelated chemotherapeutic agent, oxaliplatin. Compared to parental cells, the 5-FU-resistant cells rely more on oxidative phosphorylation than glycolysis for bioenergetic function. There was a significant increase in the expression of the drug-metabolizing cytochrome P450 enzymes CYP1A2 and CYP2A6 in 5-FU-resistant cells. The CYP450 inhibitor phenylpyrrole enhanced 5-FU-induced cytotoxicity in 5-FU-resistant cells. Two major H2S-generating enzymes, cystathionine-ß-synthase (CBS) and 3-mercaptopyruvate sulfurtransferase (3-MST) were upregulated in the 5-FU-resistant cells. 5-FU-resistant cells exhibited decreased sensitivity to the CBS inhibitor aminooxyacetate (AOAA) in terms of suppression of cell viability, inhibition of cell proliferation and inhibition of oxidative phosphorylation. However, 5FU-resistant cells remained sensitive to the antiproliferative effect of benserazide (a recently identified, potentially repurposable CBS inhibitor). Taken together, the current data suggest that 5-FU resistance in HCT116 cells is associated with the upregulation of drug-metabolizing enzymes and an enhancement of endogenous H2S production. The anticancer effect of prototypical H2S biosynthesis inhibitor AOAA is impaired in 5-FU-resistant cells, but benserazide remains efficacious. Pharmacological approaches aimed at restoring the sensitivity of 5-FU-resistant cells to chemotherapeutic agents may be useful in the formulation of novel therapeutic strategies against colorectal cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Colonic Neoplasms/enzymology , Hydrogen Sulfide/metabolism , Up-Regulation , Aminooxyacetic Acid/pharmacology , Cell Proliferation/drug effects , Cell Survival/drug effects , Colonic Neoplasms/metabolism , Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/metabolism , Drug Resistance, Neoplasm , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , Humans , Phosphorylation , Sulfurtransferases/genetics , Sulfurtransferases/metabolism
5.
Antioxid Redox Signal ; 28(16): 1463-1482, 2018 06 01.
Article in English | MEDLINE | ID: mdl-28699407

ABSTRACT

SIGNIFICANCE: Among many endogenous mediators, the gasotransmitter hydrogen sulfide (H2S) plays an important role in the regulation of glucose homeostasis. In this article we discuss different functional roles of H2S in several metabolic organs/tissues required in the maintenance of glucose homeostasis. Recent Advances: New evidence has emerged revealing the insulin sensitizing role of H2S in adipose tissue and skeletal muscle biology. In addition, H2S was demonstrated to be a potent stimulator of gluconeogenesis via the induction and stimulation of various glucose-producing pathways in the liver. CRITICAL ISSUES: Similar to its other physiological effects, H2S exhibits paradoxical characteristics in the regulation of glucose homeostasis: (1) H2S stimulates glucose production via activation of gluconeogenesis and glycogenolysis in hepatocytes, yet inhibits lipolysis in adipocytes; (2) H2S stimulates glucose uptake into adipocytes and skeletal muscle but inhibits glucose uptake into hepatocytes; (3) H2S inhibits insulin secretion from pancreatic ß cells, yet sensitizes insulin signaling and insulin-triggered response in adipose tissues and skeletal muscle. It is also unclear the impact H2S may have on glucose metabolism and utilization by other vital organs, such as the brain. FUTURE DIRECTIONS: Recent reports and ongoing studies lay the foundation for a general, although highly incomplete, understanding of the effect of H2S on regulating glucose homeostasis. In this review, we describe the molecular mechanisms and physiological outcomes of the gasotransmitter H2S on organs and tissues required for homeostatic maintenance of blood glucose. Future directions highlighting the H2S-mediated homeostatic control of glucose metabolism under physiological and insulin-resistant conditions are also discussed. Antioxid. Redox Signal. 28, 1463-1482.


Subject(s)
Glucose/metabolism , Homeostasis , Hydrogen Sulfide/metabolism , Animals , Glucose/antagonists & inhibitors , Homeostasis/drug effects , Humans , Hydrogen Sulfide/pharmacology , Insulin/metabolism
6.
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
7.
Pharmacol Res ; 113(Pt A): 116-124, 2016 11.
Article in English | MEDLINE | ID: mdl-27553984

ABSTRACT

Mammalian cells can utilize hydrogen sulfide (H2S) to support mitochondrial respiration. The aim of our study was to explore the potential role of S-sulfhydration (a H2S-induced posttranslational modification, also known as S-persulfidation) of the mitochondrial inner membrane protein ATP synthase (F1F0 ATP synthase/Complex V) in the regulation of mitochondrial bioenergetics. Using a biotin switch assay, we have detected S-sulfhydration of the α subunit (ATP5A1) of ATP synthase in response to exposure to H2S in vitro. The H2S generator compound NaHS induced S-sulfhydration of ATP5A1 in HepG2 and HEK293 cell lysates in a concentration-dependent manner (50-300µM). The activity of immunocaptured mitochondrial ATP synthase enzyme isolated from HepG2 and HEK293 cells was stimulated by NaHS at low concentrations (10-100nM). Site-directed mutagenesis of ATP5A1 in HEK293 cells demonstrated that cysteine residues at positions 244 and 294 are subject to S-sulfhydration. The double mutant ATP synthase protein (C244S/C294S) showed a significantly reduced enzyme activity compared to control and the single-cysteine-mutated recombinant proteins (C244S or C294S). To determine whether endogenous H2S plays a role in the basal S-sulfhydration of ATP synthase in vivo, we compared liver tissues harvested from wild-type mice and mice deficient in cystathionine-gamma-lyase (CSE, one of the three principal mammalian H2S-producing enzymes). Significantly reduced S-sulfhydration of ATP5A1 was observed in liver homogenates of CSE-/- mice, compared to wild-type mice, suggesting a physiological role for CSE-derived endogenous H2S production in the S-sulfhydration of ATP synthase. Various forms of critical illness (including burn injury) upregulate H2S-producing enzymes and stimulate H2S biosynthesis. In liver tissues collected from mice subjected to burn injury, we detected an increased S-sulfhydration of ATP5A1 at the early time points post-burn. At later time points (when systemic H2S levels decrease) S-sulfhydration of ATP5A1 decreased as well. In conclusion, H2S induces S-sulfhydration of ATP5A1 at C244 and C294. This post-translational modification may be a physiological mechanism to maintain ATP synthase in a physiologically activated state, thereby supporting mitochondrial bioenergetics. The sulfhydration of ATP synthase may be a dynamic process, which may be regulated by endogenous H2S levels under various pathophysiological conditions.


Subject(s)
Adenosine Triphosphate/metabolism , Energy Metabolism/physiology , Hydrogen Sulfide/metabolism , Mitochondria/metabolism , Mitochondria/physiology , Mitochondrial Proton-Translocating ATPases/metabolism , Animals , Cell Line , Cell Line, Tumor , Cystathionine gamma-Lyase/metabolism , Cysteine/metabolism , HEK293 Cells , Hep G2 Cells , Humans , Liver/metabolism , Liver/physiology , Male , Mice , Mutagenesis, Site-Directed/methods , Protein Processing, Post-Translational/physiology
8.
Nitric Oxide ; 58: 67-76, 2016 08 31.
Article in English | MEDLINE | ID: mdl-27364855

ABSTRACT

We previously showed that hydrogen sulfide (H2S) upregulates peroxisome proliferator-activated receptor-γ coactivator (PGC)-1α in primary hepatocytes. PGC-1α is a crucial regulator of mitochondrial biogenesis, a process required to maintain cellular energy homeostasis. We investigated the regulation of hepatic mitochondrial biogenesis by cystathionine γ-lyase (CSE)-generated H2S under physiological conditions. Primary hepatocytes isolated from CSE knockout (KO) and wild-type (WT) mice were used in all experiments. Mitochondrial DNA (mtDNA) and mRNA levels were measured via real-time PCR. Protein S-sulfhydration was determined via a modified biotin switch assay. MitoTracker Green was used to quantify mitochondrial content and distribution. CSE-KO hepatocytes produced less mtDNA compared to WT hepatocytes. Mitochondrial content was reduced in CSE-KO hepatocytes compared to WT hepatocytes, which was restored with NaHS (an H2S donor) treatment. CSE-KO hepatocytes exhibited lower levels of mitochondrial transcription factors and the mitochondrial transcription coactivator, peroxisome proliferator-activated receptor-γ coactivator-related protein (PPRC) compared to WT hepatocytes. NaHS administration upregulated PPRC, yet downregulated PGC-1ß protein level in mouse hepatocytes. Exogenous H2S induced the S-sulfhydration of PPRC, which was lower in untreated CSE-KO hepatocytes, but not that of PGC-1ß. Finally, knockdown of either PGC-1α or PPRC significantly decreased NaHS-stimulated mitochondrial biogenesis in hepatocytes, where knockdown of both genes were required to abolish NaHS-induced mitochondrial biogenesis. Endogenous H2S-induced liver mitochondrial biogenesis is dependent upon PGC-1α and PPRC signaling in primary hepatocytes. This study may offer clues to the regulation of energy homeostasis under physiological conditions as well as mitochondrial dysregulation.


Subject(s)
Cystathionine gamma-Lyase/metabolism , Hepatocytes/physiology , Hydrogen Sulfide/metabolism , Liver/physiology , Mitochondria/physiology , Organelle Biogenesis , Animals , Cystathionine gamma-Lyase/genetics , DNA-Binding Proteins/metabolism , Hepatocytes/ultrastructure , High Mobility Group Proteins/metabolism , Liver/ultrastructure , Male , Mice, Knockout , NF-E2-Related Factor 1/metabolism , NF-E2-Related Factor 2/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Proton-Translocating ATPases/metabolism , Transcription Factors/metabolism
9.
Mol Med ; 22: 361-379, 2016 09.
Article in English | MEDLINE | ID: mdl-27257787

ABSTRACT

Colon cancer cells contain high levels of cystathionine-beta-synthase (CBS). Its product, hydrogen sulfide (H2S) promotes the growth and proliferation of colorectal tumor cells. In order to improve the antitumor efficacy of the prototypical CBS inhibitor aminooxyacetic acid (AOAA), we have designed and synthesized YD0171, a methyl ester derivative of AOAA. The antiproliferative effect of YD0171 exceeded the antiproliferative potency of AOAA in HCT116 human colon cancer cells. The esterase inhibitor paraoxon prevented the cellular inhibition of CBS activity by YD0171. YD0171 suppressed mitochondrial respiration and glycolytic function and induced G0/G1 arrest, but did not induce tumor cell apoptosis or necrosis. Metabolomic analysis in HCT116 cells showed that YD0171 affects multiple pathways of cell metabolism. The efficacy of YD0171 as an inhibitor of tumor growth was also tested in nude mice bearing subcutaneous HCT116 cancer cell xenografts. Animals were treated via subcutaneous injection of vehicle, AOAA (1, 3 or 9 mg/kg/day) or YD0171 (0.1, 0.5 or 1 mg/kg/day) for 3 weeks. Tumor growth was significantly reduced by 9 mg/kg/day AOAA, but not at the lower doses. YD0171 was more potent: tumor volume was significantly inhibited at 0.5 and 1 mg/kg/day. Thus, the in vivo efficacy of YD0171 is 9-times higher than that of AOAA. YD0171 (1 mg/kg/day) attenuated tumor growth and metastasis formation in the intracecal HCT116 tumor model. YD0171 (3 mg/kg/day) also reduced tumor growth in patient-derived tumor xenograft (PDTX) bearing athymic mice. YD0171 (3 mg/kg/day) induced the regression of established HCT116 tumors in vivo. A 5-day safety study in mice demonstrated that YD0171 at 20 mg/kg/day (given in two divided doses) does not increase plasma markers of organ injury, nor does it induce histological alterations in the liver or kidney. YD0171 caused a slight elevation in plasma homocysteine levels. In conclusion, the prodrug approach improves the pharmacological profile of AOAA; YD0171 represents a prototype for CBS inhibitory anticancer prodrugs. By targeting colorectal cancer bioenergetics, an emerging important hallmark of cancer, the approach exemplified herein may offer direct translational opportunities.

10.
Antioxid Redox Signal ; 24(3): 129-40, 2016 Jan 20.
Article in English | MEDLINE | ID: mdl-26401978

ABSTRACT

AIMS: To investigate the regulation of hepatic glucose production by cystathionine γ-lyase (CSE)-generated hydrogen sulfide (H2S) in hepatic glucose production under physiological conditions. RESULTS: We found that CSE knockout (KO) mice had a reduced rate of gluconeogenesis, which was reversed by administration of NaHS (an H2S donor) (i.p.). Interestingly, isolated CSE KO hepatocytes exhibited a reduced glycemic response to chemical-induced activation of the cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA) and glucocorticoid pathways compared with wild-type (WT) hepatocytes. Treatment with the inhibitors for PKA (KT5720) or glucocorticoid receptor (GR) (RU-486) significantly reduced H2S-stimulated glucose production from both WT and CSE KO mouse hepatocytes. NaHS treatment upregulated the protein levels of key gluconeogenic transcription factors, such as peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) and CCAAT-enhancer-binding protein-ß (C/EBP-ß). Moreover, exogenous H2S augmented the S-sulfhydration of the rate-limiting gluconeogenic enzymes and PGC-1α and increased their activities, which were lower in untreated CSE KO hepatocytes. Finally, knockdown of PGC-1α, but not C/EBP-ß, significantly decreased NaHS-induced glucose production from the primary hepatocytes. INNOVATION: This study demonstrates the stimulatory effect of endogenous H2S on liver glucose production and reveals three underlying mechanisms; that is, H2S upregulates the expression levels of PGC-1α and phosphoenolpyruvate carboxykinase via the GR pathway; H2S upregulates the expression level of PGC-1α through the activation of the cAMP/PKA pathway as well as PGC-1α activity via S-sulfhydration; and H2S upregulates the expression and the activities (by S-sulfhydration) of glucose-6-phosphatase and fructose-1,6-bisphosphatase. CONCLUSION: This study may offer clues for the homeostatic regulation of glucose metabolism under physiological conditions and its dysregulation in metabolic syndrome.


Subject(s)
Cystathionine gamma-Lyase/metabolism , Gluconeogenesis/genetics , Glucose/metabolism , Liver/metabolism , Transcription Factors/metabolism , Animals , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , Carbazoles/administration & dosage , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Cystathionine gamma-Lyase/genetics , Hepatocytes/metabolism , Hydrogen Sulfide/metabolism , Mice , Mice, Knockout , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Pyrroles/administration & dosage , Receptors, Glucocorticoid/antagonists & inhibitors , Transcription Factors/genetics
11.
Biochim Biophys Acta ; 1850(11): 2293-303, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26272431

ABSTRACT

BACKGROUND: Cystathionine gamma-lyase (CSE)-derived hydrogen sulfide (H(2)S) possesses diverse roles in the liver, affecting lipoprotein synthesis, insulin sensitivity, and mitochondrial biogenesis. H(2)S S-sulfhydration is now proposed as a major mechanism for H(2)S-mediated signaling. Pyruvate carboxylase (PC) is an important enzyme for gluconeogenesis. S-sulfhydration regulation of PC by H(2)S and its implication in gluconeogenesis in the liver have been unknown. METHODS: Gene expressions were analyzed by real-time PCR and western blotting, and protein S-sulfhydration was assessed by both modified biotin switch assay and tag switch assay. Glucose production and PC activity was measured with coupled enzyme assays, respectively. RESULTS: Exogenously applied H(2)S stimulates PC activity and gluconeogenesis in both HepG2 cells and mouse primary liver cells. CSE overexpression enhanced but CSE knockout reduced PC activity and gluconeogenesis in liver cells, and blockage of PC activity abolished H(2)S-induced gluconeogenesis. H(2)S had no effect on the expressions of PC mRNA and protein, while H(2)S S-sulfhydrated PC in a dithiothreitol-sensitive way. PC S-sulfhydration was significantly strengthened by CSE overexpression but attenuated by CSE knockout, suggesting that H(2)S enhances glucose production through S-sulfhydrating PC. Mutation of cysteine 265 in human PC diminished H(2)S-induced PC S-sulfhydration and activity. In addition, high-fat diet feeding of mice decreased both CSE expression and PC S-sulfhydration in the liver, while glucose deprivation of HepG2 cells stimulated CSE expression. CONCLUSIONS: CSE/H(2)S pathway plays an important role in the regulation of glucose production through S-sulfhydrating PC in the liver. GENERAL SIGNIFICANCE: Tissue-specific regulation of CSE/H(2)S pathway might be a promising therapeutic target of diabetes and other metabolic syndromes.


Subject(s)
Gluconeogenesis , Hydrogen Sulfide/pharmacology , Liver/metabolism , Pyruvate Carboxylase/metabolism , Animals , Cysteine/metabolism , Diet, High-Fat , Glucose/biosynthesis , HEK293 Cells , Hep G2 Cells , Humans , Male , Mice
12.
Antioxid Redox Signal ; 20(5): 805-17, 2014 Feb 10.
Article in English | MEDLINE | ID: mdl-23582095

ABSTRACT

SIGNIFICANCE: Stigmatized as a toxic environmental pollutant for centuries, hydrogen sulfide (H2S) has gained recognition over the last decade as an important gasotransmitter that functions in physiological and pathophysiological conditions, such as atherosclerosis. RECENT ADVANCES: Atherosclerosis is a common disease that stems from the buildup of fatty/cholesterol plaques on the endothelial cells of arteries. The deposits mitigate thickening and stiffening of arterial tissue, which contributes to concomitant systemic or localized vascular disorders. Recently, it has been recognized that H2S plays an anti-atherosclerotic role, and its deficiency leads to early development and progression of atherosclerosis. This review article presents multiple lines of evidence for the protective effects of H2S against the development of atherosclerosis. Also highlighted are the characterization of altered metabolism of H2S in the development of atherosclerosis, underlying molecular and cellular mechanisms, and potential therapeutic intervention based on H2S supplementation for atherosclerosis management. CRITICAL ISSUES: Although a protective role of H2S against atherosclerosis has emerged, controversy remains regarding the mechanisms underlying H2S-induced endothelial cell proliferation and angiogenesis as well as its anti-inflammatory properties. The therapeutic value of H2S to this pathophysiological condition has not been tested clinically but, nonetheless, it shows tremendous promise. FUTURE DIRECTIONS: The efficiency and safety profile of H2S-based therapeutic approaches should be refined, and the mechanisms by which H2S exerts its beneficial effects should be elucidated to develop more specific and potent therapeutic strategies to treat atherosclerosis. Whether the therapeutic effects of H2S in animal studies are transferable to clinical studies merits future investigation.


Subject(s)
Atherosclerosis/metabolism , Hydrogen Sulfide/metabolism , Animals , Antioxidants/metabolism , Atherosclerosis/immunology , Atherosclerosis/pathology , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Humans , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/physiology , Signal Transduction , Vascular Calcification/metabolism
13.
Circulation ; 127(25): 2523-34, 2013 Jun 25.
Article in English | MEDLINE | ID: mdl-23704252

ABSTRACT

BACKGROUND: Cystathionine γ-lyase (CSE) produces hydrogen sulfide (H2S) in the cardiovascular system. The deficiency of CSE in mice leads to a decreased endogenous H2S level, an age-dependent increase in blood pressure, and impaired endothelium-dependent vasorelaxation. To date, there is no direct evidence for a causative role of altered metabolism of endogenous H2S in atherosclerosis development. METHODS AND RESULTS: Six-week-old CSE gene knockout and wild-type mice were fed with either a control chow or atherogenic paigen-type diet for 12 weeks. Plasma lipid profile and homocysteine levels, blood pressure, oxidative stress, atherosclerotic lesion size in the aortic roots, cell proliferation, and adhesion molecule expression were then analyzed. CSE-knockout mice fed with atherogenic diet developed early fatty streak lesions in the aortic root, elevated plasma levels of cholesterol and low-density lipoprotein cholesterol, hyperhomocysteinemia, increased lesional oxidative stress and adhesion molecule expression, and enhanced aortic intimal proliferation. Treatment of CSE-knockout mice with NaHS, but not N-acetylcysteine or ezetimibe, inhibited the accelerated atherosclerosis development. Double knockout of CSE and apolipoprotein E gene expression in mice exacerbated atherosclerosis development more than that in the mice with only apolipoprotein E or CSE knockout. CONCLUSIONS: Endogenously synthesized H2S protects vascular tissues from atherogenic damage by reducing vessel intimal proliferation and inhibiting adhesion molecule expression. Decreased endogenous H2S production predisposes the animals to vascular remodeling and early development of atherosclerosis. The CSE/H2S pathway is an important therapeutic target for protection against atherosclerosis.


Subject(s)
Atherosclerosis/etiology , Atherosclerosis/metabolism , Cystathionine gamma-Lyase/deficiency , Hydrogen Sulfide/antagonists & inhibitors , Hydrogen Sulfide/metabolism , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Atherosclerosis/drug therapy , Cell Proliferation/drug effects , Cystathionine gamma-Lyase/genetics , Cystathionine gamma-Lyase/metabolism , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidative Stress/physiology , Signal Transduction/physiology , Sulfides/pharmacology , Sulfides/therapeutic use , Tunica Intima/drug effects , Tunica Intima/pathology
14.
Endocrinology ; 154(1): 114-26, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23183179

ABSTRACT

Mounting evidence has established hydrogen sulfide (H(2)S) as an important gasotransmitter with multifaceted physiological functions. The aim of the present study was to investigate the role of H(2)S on glucose utilization, glycogen synthesis, as well as gluconeogenesis in both HepG(2) cells and primary mouse hepatocytes. Incubation with NaHS (a H(2)S donor) impaired glucose uptake and glycogen storage in HepG(2) cells via decreasing glucokinase activity. Adenovirus-mediated cystathionine γ-lyase (CSE) overexpression increased endogenous H(2)S production and lowered glycogen content in HepG(2) cells. Glycogen content was significantly higher in liver tissues from CSE knockout (KO) mice compared to that from wild type (WT) mice in fed condition. Glucose consumption was less in primarily cultured hepatocytes isolated from WT mice than those from CSE KO mice, but more glucose was produced by hepatocytes via gluconeogenesis and glycogenolysis pathways in WT mice than in CSE KO mice. NaHS treatment reduced the phosphorylation of AMP-activated protein kinase, whereas stimulation of AMP-activated protein kinase by 5-aminoimidazole-4-carboxamide-1-ß-d-ribofuranoside reversed H(2)S-impaired glucose uptake. H(2)S-increased glucose production was likely through increased phosphoenolpyruvate carboxykinase activity. In addition, insulin at the physiological range inhibited CSE expression, and H(2)S decreased insulin-stimulated phosphorylation of Akt in HepG(2) cells. CSE expression was increased, however, in insulin-resistant state induced by exposing cells to high levels of insulin (500 nm) and glucose (33 mm) for 24 h. Taken together, these data suggest that the interaction of H(2)S and insulin in liver plays a pivotal role in regulating insulin sensitivity and glucose metabolism.


Subject(s)
Gluconeogenesis/drug effects , Glucose/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Hydrogen Sulfide/pharmacology , AMP-Activated Protein Kinases/metabolism , Animals , Cells, Cultured , Cystathionine gamma-Lyase/genetics , Cystathionine gamma-Lyase/metabolism , Glucokinase/metabolism , Glycogen/metabolism , Hep G2 Cells , Humans , Male , Mice , Mice, Knockout , Phosphorylation/drug effects
15.
Expert Rev Clin Pharmacol ; 4(1): 63-73, 2011 Jan.
Article in English | MEDLINE | ID: mdl-22115349

ABSTRACT

The metabolic syndrome is a group of abnormalities including obesity, high blood pressure, hyperinsulinemia, high blood glucose levels and hyperlipidemia that together greatly increase the risk of developing cardiovascular disease and Type 2 diabetes. Hydrogen sulfide (H(2)S) is a vasodilatory gasotransmitter mediator in the cardiovascular system, proposed as an endothelium-derived relaxing factor. A lack of H(2)S and its synthesizing enzyme, cystathionine γ-lyase, in the vasculature causes hypertension, whereas an increase in the pancreas reduces insulin secretion. Thus, research is making inroads to determine whether H(2)S is involved in the pathogenesis of the metabolic syndrome. Several laboratories are synthesizing and testing clinically used drugs that release H(2)S. Some of these compounds are being tested for effectiveness in the metabolic syndrome.


Subject(s)
Hydrogen Sulfide/metabolism , Metabolic Syndrome/metabolism , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Humans , Hydrogen Sulfide/therapeutic use , Hypertension/drug therapy , Hypertension/metabolism , Metabolic Syndrome/drug therapy , Obesity/drug therapy , Obesity/metabolism
16.
PLoS One ; 6(12): e29592, 2011.
Article in English | MEDLINE | ID: mdl-22216325

ABSTRACT

We have previously reported that hydrogen sulfide (H(2)S), a gasotransmitter and vasodilator has cytoprotective properties against methylglyoxal (MG), a reactive glucose metabolite associated with diabetes and hypertension. Recently, H(2)S was shown to up-regulate peroxisome proliferator-activated receptor-γ coactivator (PGC)-1α, a key gluconeogenic regulator that enhances the gene expression of the rate-limiting gluconeogenic enzyme, fructose-1,6-bisphosphatase (FBPase). Thus, we sought to determine whether MG levels and gluconeogenic enzymes are altered in kidneys of 6-22 week-old cystathionine γ-lyase knockout (CSE(-/-); H(2)S-producing enzyme) male mice. MG levels were determined by HPLC. Plasma glucose levels were measured by an assay kit. Q-PCR was used to measure mRNA levels of PGC-1α and FBPase-1 and -2. Coupled-enzymatic assays were used to determine FBPase activity, or triosephosphate levels. Experimental controls were either age-matched wild type mice or untreated rat A-10 cells. Interestingly, we observed a significant decrease in plasma glucose levels along with a significant increase in plasma MG levels in all three age groups (6-8, 14-16, and 20-22 week-old) of the CSE(-/-) mice. Indeed, renal MG and triosephosphates were increased, whereas renal FBPase activity, along with its mRNA levels, were decreased in the CSE(-/-) mice. The decreased FBPase activity was accompanied by lower levels of its product, fructose-6-phosphate, and higher levels of its substrate, fructose-1,6-bisphosphate in renal extracts from the CSE(-/-) mice. In agreement, PGC-1α mRNA levels were also significantly down-regulated in 6-22 week-old CSE(-/-) mice. Furthermore, FBPase-1 and -2 mRNA levels were reduced in aorta tissues from CSE(-/-) mice. Administration of NaHS, a H(2)S donor, increased the gene expression of PGC-1α and FBPase-1 and -2 in cultured rat A-10 cells. In conclusion, overproduction of MG in CSE(-/-) mice is due to a H(2)S-mediated down-regulation of the PGC-1α-FBPase pathway, further suggesting the important role of H(2)S in the regulation of glucose metabolism and MG generation.


Subject(s)
Cystathionine gamma-Lyase/metabolism , Down-Regulation , Fructose-Bisphosphatase/metabolism , Kidney/metabolism , Pyruvaldehyde/metabolism , Transcription Factors/metabolism , Animals , Blood Glucose/analysis , Cell Line , Chromatography, High Pressure Liquid , Cystathionine gamma-Lyase/genetics , DNA Primers , Fructose-Bisphosphatase/genetics , Kidney/enzymology , Male , Mice , Mice, Knockout , RNA, Messenger/genetics , Rats , Real-Time Polymerase Chain Reaction , Transcription Factors/genetics
17.
Can J Physiol Pharmacol ; 88(3): 273-84, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20393592

ABSTRACT

Aging is a multifactorial process that involves changes at the cellular, tissue, organ and the whole body levels resulting in decreased functioning, development of diseases, and ultimately death. Oxidative stress is believed to be a very important factor in causing aging and age-related diseases. Oxidative stress is caused by an imbalance between oxidants such as reactive oxygen species (ROS) and antioxidants. ROS are produced from the mitochondrial electron transport chain and many oxidative reactions. Methylglyoxal (MG) is a highly reactive dicarbonyl metabolite formed during glucose, protein and fatty acid metabolism. MG levels are elevated in hyperglycemia and other conditions. An excess of MG formation can increase ROS production and cause oxidative stress. MG reacts with proteins, DNA and other biomolecules, and is a major precursor of advanced glycation end products (AGEs). AGEs are also associated with the aging process and age-related diseases such as cardiovascular complications of diabetes, neurodegenerative diseases and connective tissue disorders. AGEs also increase oxidative stress. In this review we discuss the potential role of MG in the aging process through increasing oxidative stress besides causing AGEs formation. Specific and effective scavengers and crosslink breakers of MG and AGEs are being developed and can become potential treatments to slow the aging process and prevent many diseases.


Subject(s)
Aging/metabolism , Oxidative Stress/physiology , Pyruvaldehyde/metabolism , Aging/drug effects , Animals , Glycation End Products, Advanced/adverse effects , Glycation End Products, Advanced/biosynthesis , Humans , Oxidative Stress/drug effects , Pyruvaldehyde/adverse effects , Reactive Oxygen Species/adverse effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology
18.
Antioxid Redox Signal ; 12(9): 1093-100, 2010 May 01.
Article in English | MEDLINE | ID: mdl-19803740

ABSTRACT

Hydrogen sulfide (H(2)S) is a gasotransmitter with multifaceted physiological functions, including the regulation of glucose metabolism. Methylglyoxal (MG) is an intermediate of glucose metabolism and plays an important role in the pathogenesis of insulin resistance syndromes. In the present study, we investigated the effect of MG on H(2)S synthesis and the interaction between these two endogenous substances. In cultured vascular smooth muscle cells (VSMCs), MG (10, 30, and 50 microM) significantly decreased cellular H(2)S levels in a concentration-dependent manner, while H(2)S donor, NaHS (30, 60, and 90 microM), significantly decreased cellular MG levels. The expression level and activity of H(2)S-producing enzyme, cystathionine gamma-lyase (CSE), were significantly decreased by MG treatment. NaHS (30-90 microM) significantly inhibited MG (10 or 30 microM)-induced ROS production. Cellular levels of GSH, cysteine, and homocysteine were also increased by MG or NaHS treatment. Furthermore, direct reaction of H(2)S with MG in both concentration- and time-dependent manners were observed in in vitro incubations. In conclusion, MG regulates H(2)S level in VSMCs by downregulating CSE protein expression and directly reacting with H(2)S molecule. Interaction of MG with H(2)S may be one of future directions for the studies on glucose metabolism and the development of insulin resistance syndromes.


Subject(s)
Hydrogen Sulfide/chemistry , Muscle, Smooth, Vascular/cytology , Pyruvaldehyde/chemistry , Animals , Cell Line , Cysteine/chemistry , Dose-Response Relationship, Drug , Glutathione/metabolism , Homocysteine/chemistry , Hydrogen Sulfide/metabolism , Lyases/metabolism , Myocytes, Smooth Muscle/cytology , Pyruvaldehyde/metabolism , Rats , Reactive Oxygen Species , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...