Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
1.
Biosystems ; 234: 105066, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37898397

ABSTRACT

Trypanosoma cruzi is the causal agent of American Trypanosomiasis or Chagas Disease in humans. The current drugs for its treatment benznidazole and nifurtimox have inconveniences of toxicity and efficacy; therefore, the search for new therapies continues. Validation through genetic strategies of new drug targets against the parasite metabolism have identified numerous essential genes. Target validation can be further narrowed by applying Metabolic Control Analysis (MCA) to determine the flux control coefficients of the pathway enzymes. That coefficient is a quantitative value that represents the degree in which an enzyme/transporter determines the flux of a metabolic pathway; those with the highest coefficients can be promising drug targets. Previous studies have demonstrated that cysteine (Cys) is a key precursor for the synthesis of trypanothione, the main antioxidant metabolite in the parasite. In this research, MCA was applied in an ex vivo system to the enzymes of the reverse transsulfuration pathway (RTP) for Cys synthesis composed by cystathionine beta synthase (CBS) and cystathionine gamma lyase (CGL). The results indicated that CGL has 90% of the control of the pathway flux. Inhibition of CGL with propargylglycine (PAG) decreased the levels of Cys and trypanothione and depleted those of glutathione in epimastigotes (proliferative stage in the insect vector); these metabolite changes were prevented by supplementing with Cys, suggesting a compensatory role of the Cys transport (CysT). Indeed, Cys supplementation (but not PAG treatment) increased the activity of the CysT in epimastigotes whereas in trypomastigotes (infective stage in mammals) CysT was increased when they were incubated with PAG. Our results suggested that CGL could be a potential drug target given its high control on the RTP flux and its effects on the parasite antioxidant defense. However, the redundant Cys supply pathways in the parasite may require inhibition of the CysT as well. Our findings also suggest differential responses of the Cys supply pathways in different parasite stages.


Subject(s)
Cysts , Trypanosoma cruzi , Humans , Animals , Antioxidants/metabolism , Cysteine/metabolism , Cystathionine gamma-Lyase/genetics , Cystathionine gamma-Lyase/metabolism , Mammals
2.
Nitric Oxide ; 136-137: 12-23, 2023 07 01.
Article in English | MEDLINE | ID: mdl-37182786

ABSTRACT

AIM: Increasing evidence has proposed that mitochondrial abnormalities may be an important factor contributing to the development of heart failure with preserved ejection fraction (HFpEF). Hydrogen sulfide (H2S) has been suggested to play a pivotal role in regulating mitochondrial function. Therefore, the present study was designed to explore the protective effect of H2S on mitochondrial dysfunction in a multifactorial mouse model of HFpEF. METHODS: Wild type, 8-week-old, male C57BL/6J mice or cardiomyocyte specific-Cse (Cystathionine γ-lyase, a major H2S-producing enzyme) knockout mice (CSEcko) were given high-fat diet (HFD) and l-NAME (an inhibitor of constitutive nitric oxide synthases) or standardized chow. After 4 weeks, mice were randomly administered with NaHS (a conventional H2S donor), ZLN005 (a potent transcriptional activator of PGC-1α) or vehicle. After additional 4 weeks, echocardiogram and mitochondrial function were evaluated. Expression of PGC-1α, NRF1 and TFAM in cardiomyocytes was assayed by Western blot. RESULTS: Challenging with HFD and l-NAME in mice not only caused HFpEF but also inhibited the production of endogenous H2S in a time-dependent manner. Meanwhile the expression of PGC-1α and mitochondrial function in cardiomyocytes were impaired. Supplementation with NaHS not only upregulated the expression of PGC-1α, NRF1 and TFAM in cardiomyocytes but also restored mitochondrial function and ultrastructure, conferring an obvious improvement in cardiac diastolic function. In contrast, cardiac deletion of CSE gene aggravated the inhibition of PGC-1α-NRF1-TFAM pathway, mitochondrial abnormalities and diastolic dysfunction. The deleterious effect observed in CSEcko HFpEF mice was partially counteracted by pre-treatment with ZLN005 or supplementation with NaHS. CONCLUSION: Our findings have demonstrated that H2S ameliorates left ventricular diastolic dysfunction by restoring mitochondrial abnormalities via upregulating PGC-1α and its downstream targets NRF1 and TFAM, suggesting the therapeutic potential of H2S supplementation in multifactorial HFpEF.


Subject(s)
Heart Failure , Hydrogen Sulfide , Mice , Male , Animals , Hydrogen Sulfide/pharmacology , Hydrogen Sulfide/therapeutic use , Hydrogen Sulfide/metabolism , Heart Failure/drug therapy , NG-Nitroarginine Methyl Ester/pharmacology , Stroke Volume , Mice, Inbred C57BL , Myocytes, Cardiac/metabolism , Mice, Knockout , Cystathionine gamma-Lyase/metabolism
3.
Int J Mol Sci ; 23(20)2022 Oct 19.
Article in English | MEDLINE | ID: mdl-36293383

ABSTRACT

Deodorized garlic (DG) may favor the activity of the antioxidant enzymes and promote the synthesis of hydrogen sulfide (H2S). The objective was to test if DG favors an increase in H2S and if it decreases the oxidative stress caused by lipopolysaccharide (LPS) in rat hearts. A total of 24 rats were divided into 4 groups: Group 1 control (C), Group 2 LPS, Group 3 DG, and Group 4 LPS plus DG. The cardiac mechanical performance (CMP), coronary vascular resistance (CVR), and oxidative stress markers, such as total antioxidant capacity (TAC), glutathione (GSH), selenium (Se), lipid peroxidation (LPO), thiols, hydrogen sulfide (H2S), and the activities and expressions of thioredoxin reductase (TrxR), glutathione peroxidase (GPx), and glutathione-S-transferase (GST), cystathionine synthetase (CBS), cystathionine γ-lyase (CTH), iNOS, and eNOS-p, were analyzed in the heart. Infarct zones in the cardiac tissue were present (p = 0.01). The CMP and CVR decreased and increased (p ≤ 0.05), TAC, GSH, H2S, NO, thiols, and GST activity (p ≤ 0.01) decreased, and LPO and iNOS increased (p ≤ 0.05). The activities and expressions of TrxR, GPx, eNOS-p, CTH, and CBS (p ≤ 0.05) decreased with the LPS treatment; however, DG normalized this effect. DG treatment decreases heart damage caused by LPS through the cross-talk between the H2S and NO systems.


Subject(s)
Garlic , Hydrogen Sulfide , Selenium , Animals , Rats , Antioxidants/pharmacology , Antioxidants/metabolism , Cystathionine beta-Synthase/metabolism , Cystathionine gamma-Lyase/metabolism , Garlic/metabolism , Glutathione/metabolism , Glutathione Peroxidase/metabolism , Hydrogen Sulfide/pharmacology , Hydrogen Sulfide/metabolism , Lipopolysaccharides/pharmacology , Oxidative Stress , Selenium/pharmacology , Sulfhydryl Compounds/pharmacology , Thioredoxin-Disulfide Reductase/metabolism , Transferases/metabolism
4.
Oxid Med Cell Longev ; 2022: 1392896, 2022.
Article in English | MEDLINE | ID: mdl-35910846

ABSTRACT

Although it is an essential nutrient, high choline intake directly or indirectly via its metabolite is associated with increased risk of cardiovascular disease, the mechanism of which remains to be elucidated. The present study was performed to investigate whether hydrogen sulfide (H2S) was involved in high choline-induced cardiac dysfunction and explore the potential mechanisms. We found that ejection fraction (EF) and fractional shortening (FS), the indicators of cardiac function measured by echocardiography, were significantly decreased in mice fed a diet containing 1.3% choline for 4 months as compared to the control, while applying 3,3-dimethyl-1-butanol (DMB) to suppress trimethylamine N-oxide (TMAO, a metabolite of choline) generation ameliorated the cardiac function. Subsequently, we found that feeding choline or TMAO significantly increased the protein levels of cyclic GMP-AMP (cGAMP) synthase (cGAS), stimulator of interferon genes (STING), NOD-like receptor protein 3 (NLRP3), caspase-1, and interleukin-1ß (IL-1ß) as compared to the control, which indicated the activation of cGAS-STING-NLRP3 inflammasome axis. Moreover, the protein expression of cystathionine γ-lyase (CSE), the main enzyme for H2S production in the cardiovascular system, was significantly increased after dietary supplementation with choline, but the plasma H2S levels were significantly decreased. To observe the effect of endogenous H2S, CSE knockout (KO) mice were used, and we found that the EF, FS, and plasma H2S levels in WT mice were significantly decreased after dietary supplementation with choline, while there was no difference between CSE KO + control and CSE KO + choline group. To observe the effect of exogenous H2S, mice were intraperitoneally injected with sodium hydrosulfide (NaHS, a H2S donor) for 4 months, and we found that NaHS improved the cardiac function and reduced the protein levels of cGAS, STING, NLRP3, caspase-1, and IL-1ß in mice receiving dietary choline. In conclusion, our studies revealed that high choline diet decreased plasma H2S levels and induced cardiac dysfunction via cGAS-STING-NLRP3 inflammasome axis while H2S treatment could restore the cardiac function by inhibiting cGAS-STING-NLRP3 inflammasome axis.


Subject(s)
Heart Diseases , Hydrogen Sulfide , Animals , Caspase 1/metabolism , Choline/toxicity , Cystathionine gamma-Lyase/metabolism , Heart Diseases/chemically induced , Hydrogen Sulfide/metabolism , Hydrogen Sulfide/pharmacology , Hydrogen Sulfide/therapeutic use , Inflammasomes/metabolism , Membrane Proteins/metabolism , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Proteins , Nucleotidyltransferases
5.
Int J Mol Sci ; 23(11)2022 May 25.
Article in English | MEDLINE | ID: mdl-35682634

ABSTRACT

Sarcopenia is a gradual and generalized skeletal muscle (SKM) syndrome, characterized by the impairment of muscle components and functionality. Hydrogen sulfide (H2S), endogenously formed within the body from the activity of cystathionine-γ-lyase (CSE), cystathionine- ß-synthase (CBS), and mercaptopyruvate sulfurtransferase, is involved in SKM function. Here, in an in vitro model of sarcopenia based on damage induced by dexamethasone (DEX, 1 µM, 48 h treatment) in C2C12-derived myotubes, we investigated the protective potential of exogenous and endogenous sources of H2S, i.e., glucoraphanin (30 µM), L-cysteine (150 µM), and 3-mercaptopyruvate (150 µM). DEX impaired the H2S signalling in terms of a reduction in CBS and CSE expression and H2S biosynthesis. Glucoraphanin and 3-mercaptopyruvate but not L-cysteine prevented the apoptotic process induced by DEX. In parallel, the H2S-releasing molecules reduced the oxidative unbalance evoked by DEX, reducing catalase activity, O2- levels, and protein carbonylation. Glucoraphanin, 3-mercaptopyruvate, and L-cysteine avoided the changes in myotubes morphology and morphometrics after DEX treatment. In conclusion, in an in vitro model of sarcopenia, an impairment in CBS/CSE/H2S signalling occurs, whereas glucoraphanin, a natural H2S-releasing molecule, appears more effective for preventing the SKM damage. Therefore, glucoraphanin supplementation could be an innovative therapeutic approach in the management of sarcopenia.


Subject(s)
Hydrogen Sulfide , Sarcopenia , Cystathionine , Cystathionine beta-Synthase/metabolism , Cystathionine gamma-Lyase/metabolism , Cysteine/metabolism , Glucosinolates , Humans , Hydrogen Sulfide/metabolism , Hydrogen Sulfide/pharmacology , Oximes , Sarcopenia/drug therapy , Sulfoxides , Sulfurtransferases/metabolism
6.
Oxid Med Cell Longev ; 2022: 6153772, 2022.
Article in English | MEDLINE | ID: mdl-35571249

ABSTRACT

Doxorubicin (DOX) is an efficient antitumor anthracycline drug, but its cardiotoxicity adversely affects the prognosis of the patients. In this study, we explored whether endogenous gasotransmitter hydrogen sulfide (H2S) could protect against DOX-induced cardiomyocyte apoptosis and its mechanisms. The results indicated that DOX significantly downregulated endogenous H2S production and endogenous synthetase cystathionine γ-lyase (CSE) expression and obviously stimulated the apoptosis in H9C2 cells. The supplement of H2S donor sodium hydrosulfide (NaHS) or overexpression of CSE inhibited DOX-induced H9C2 cell apoptosis. DOX enhanced the activities of caspase family members in cardiomyocytes, while NaHS attenuated DOX-enhanced caspase-3, caspase-2, and caspase-9 activities by 223.1%, 73.94%, and 52.29%, respectively. Therefore, taking caspase-3 as a main target, we demonstrated that NaHS or CSE overexpression alleviated the cleavage of caspase-3, suppressed caspase-3 activity, and inhibited the cleavage of poly ADP-ribose polymerase (PARP). Mechanistically, we found that H2S persulfidated caspase-3 in H9C2 cells and human recombinant caspase-3 protein, while the thiol-reducing agent dithiothreitol (DTT) abolished H2S-induced persulfidation of caspase-3 and thereby prevented the antiapoptotic effect of H2S on caspase-3 in H9C2 cells. The mutation of caspase-3 C148S and C170S failed to block caspase-3 persulfidation by H2S in H9C2 cells. However, caspase-3 C163S mutation successfully abolished the effect of H2S on caspase-3 persulfidation and the corresponding protection of H9C2 cells. Collectively, these findings indicate that endogenous H2S persulfidates caspase-3 at cysteine 163, inhibiting its activity and cardiomyocyte apoptosis. Sufficient endogenous H2S might be necessary for the protection against myocardial cell apoptosis induced by DOX. The results of the study might open new avenues with respect to the therapy of DOX-stimulated cardiomyopathy.


Subject(s)
Antineoplastic Agents , Hydrogen Sulfide , Antineoplastic Agents/pharmacology , Apoptosis , Caspase 3/genetics , Caspase 3/metabolism , Cystathionine gamma-Lyase/genetics , Cystathionine gamma-Lyase/metabolism , Cysteine/metabolism , Cysteine/pharmacology , Doxorubicin/pharmacology , Humans , Hydrogen Sulfide/metabolism , Hydrogen Sulfide/pharmacology , Myocytes, Cardiac/metabolism
7.
Biomolecules ; 11(12)2021 12 10.
Article in English | MEDLINE | ID: mdl-34944503

ABSTRACT

This paper provides information concerning the activity and expression levels of three sulfurtransferases (STRs): rhodanese (TST, EC: 2.8.1.1), 3-mercaptopyruvate sulfurtransferase (MPST, EC: 2.8.1.2) and cystathionine γ-lyase (CTH, EC: 4.4.1.1) in various cell lines. Since very limited data are available in the scientific literature on this subject, the available data are included in this paper. These shortages often force the researchers to carry out their own screening tests that allow them to choose an appropriate model for their further studies. This work supplements the existing deficiencies in this area and presents the activity and expression of STRs in the eight most frequently chosen cell lines: the mouse mammary gland cell line (NMuNG, ATCC: CRL-1636), mouse mammary gland tumor (4T1, ATCC: CRL-2539), mouse fibroblast (MEF, ATCC: SCRC-1008), mouse melanoma (B16-F1, ATCC: CRL-6323), human colorectal adenocarcinoma (Caco-2, ATCC: HTB-37), human embryonic kidney (HEK-293, ATCC: CRL-1573), human osteosarcoma (MG-63, ATCC: CRL-1427) and rat myocardium (H9c2, ATCC: CRL-1446). Changes in STRs activity are directly related to the bioavailability of cysteine and the sulfane sulfur level, and thus the present authors also measured these parameters, as well as the level of glutathione (its reduced (GSH) and oxidized (GSSG) form) and the [GSH]/[GSSG] ratio that determines the antioxidant capacity of the cells. STRs demonstrate diverse functionality and clinical relevance; therefore, we also performed an analysis of genetic variation of STRs genes that revealed a large number of polymorphisms. Although STRs still provide challenges in several fields, responding to them could not only improve the understanding of various diseases, but may also provide a way to treat them.


Subject(s)
Cystathionine gamma-Lyase/metabolism , Polymorphism, Single Nucleotide , Sulfurtransferases/metabolism , Thiosulfate Sulfurtransferase/metabolism , Animals , Caco-2 Cells , Cell Line , Cystathionine gamma-Lyase/genetics , Cysteine/metabolism , Gene Expression Profiling , Gene Expression Regulation, Enzymologic , HEK293 Cells , Humans , Mice , Rats , Sulfur/metabolism , Sulfurtransferases/genetics , Thiosulfate Sulfurtransferase/genetics
8.
Circulation ; 143(9): 935-948, 2021 03 02.
Article in English | MEDLINE | ID: mdl-33307764

ABSTRACT

BACKGROUND: In vascular endothelial cells, cysteine metabolism by the cystathionine γ lyase (CSE), generates hydrogen sulfide-related sulfane sulfur compounds (H2Sn), that exert their biological actions via cysteine S-sulfhydration of target proteins. This study set out to map the "S-sulfhydrome" (ie, the spectrum of proteins targeted by H2Sn) in human endothelial cells. METHODS: Liquid chromatography with tandem mass spectrometry was used to identify S-sulfhydrated cysteines in endothelial cell proteins and ß3 integrin intraprotein disulfide bond rearrangement. Functional studies included endothelial cell adhesion, shear stress-induced cell alignment, blood pressure measurements, and flow-induced vasodilatation in endothelial cell-specific CSE knockout mice and in a small collective of patients with endothelial dysfunction. RESULTS: Three paired sample sets were compared: (1) native human endothelial cells isolated from plaque-free mesenteric arteries (CSE activity high) and plaque-containing carotid arteries (CSE activity low); (2) cultured human endothelial cells kept under static conditions or exposed to fluid shear stress to decrease CSE expression; and (3) cultured endothelial cells exposed to shear stress to decrease CSE expression and treated with solvent or the slow-releasing H2Sn donor, SG1002. The endothelial cell "S-sulfhydrome" consisted of 3446 individual cysteine residues in 1591 proteins. The most altered family of proteins were the integrins and focusing on ß3 integrin in detail we found that S-sulfhydration affected intraprotein disulfide bond formation and was required for the maintenance of an extended-open conformation of the ß leg. ß3 integrin S-sulfhydration was required for endothelial cell mechanotransduction in vitro as well as flow-induced dilatation in murine mesenteric arteries. In cultured cells, the loss of S-sulfhydration impaired interactions between ß3 integrin and Gα13 (guanine nucleotide-binding protein subunit α 13), resulting in the constitutive activation of RhoA (ras homolog family member A) and impaired flow-induced endothelial cell realignment. In humans with atherosclerosis, endothelial function correlated with low H2Sn generation, impaired flow-induced dilatation, and failure to detect ß3 integrin S-sulfhydration, all of which were rescued after the administration of an H2Sn supplement. CONCLUSIONS: Vascular disease is associated with marked changes in the S-sulfhydration of endothelial cell proteins involved in mediating responses to flow. Short-term H2Sn supplementation improved vascular reactivity in humans highlighting the potential of interfering with this pathway to treat vascular disease.


Subject(s)
Integrin beta Chains/chemistry , Sulfhydryl Compounds/chemistry , Animals , Chromatography, High Pressure Liquid , Cystathionine gamma-Lyase/genetics , Cystathionine gamma-Lyase/metabolism , Cysteine/chemistry , Disulfides/analysis , Disulfides/chemistry , Endothelial Cells/cytology , Endothelial Cells/metabolism , Humans , Hydrogen Sulfide/pharmacology , Integrin beta Chains/metabolism , Mechanotransduction, Cellular , Mice , Shear Strength , Tandem Mass Spectrometry , Vasodilation/drug effects , rhoA GTP-Binding Protein/metabolism
9.
Sci Rep ; 10(1): 15810, 2020 09 25.
Article in English | MEDLINE | ID: mdl-32978411

ABSTRACT

Endothelial dysfunction is a hallmark of preeclampsia, a life-threatening complication of pregnancy characterised by hypertension and elevated soluble Fms-Like Tyrosine Kinase-1 (sFlt-1). Dysregulation of hydrogen sulfide (H2S) by inhibition of cystathionine γ-lyase (CSE) increases sFlt-1 and soluble endoglin (sEng) release. We explored whether compromise in CSE/H2S pathway is linked to dysregulation of the mitochondrial bioenergetics and oxidative status. We investigated whether these effects were linked to CSE-induced sFlt-1 and sEng production in endothelial cells. Here, we demonstrate that CSE/H2S pathway sustain endothelial mitochondrial bioenergetics and loss of CSE increases the production of mitochondrial-specific superoxide. As a compensatory effect, low CSE environment enhances the reliance on glycolysis. The mitochondrial-targeted H2S donor, AP39, suppressed the antiangiogenic response and restored the mitochondrial bioenergetics in endothelial cells. AP39 revealed that upregulation of sFlt-1, but not sEng, is independent of the mitochondrial H2S metabolising enzyme, SQR. These data provide new insights into the molecular mechanisms for antiangiogenic upregulation in a mitochondrial-driven environment. Targeting H2S to the mitochondria may be of therapeutic benefit in the prevention of endothelial dysfunction associated with preeclampsia.


Subject(s)
Cystathionine gamma-Lyase/metabolism , Endoglin/antagonists & inhibitors , Endothelium, Vascular/metabolism , Energy Metabolism , Hydrogen Sulfide/pharmacology , Mitochondria/metabolism , Vascular Endothelial Growth Factor Receptor-1/antagonists & inhibitors , Air Pollutants/pharmacology , Cystathionine gamma-Lyase/genetics , Endoglin/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Mitochondria/drug effects , Reactive Oxygen Species/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism
10.
Sci Rep ; 10(1): 14657, 2020 09 04.
Article in English | MEDLINE | ID: mdl-32887901

ABSTRACT

Cystathionine ß-synthase (CBS) catalyzes the condensation of serine and homocysteine to water and cystathionine, which is then hydrolyzed to cysteine, α-ketobutyrate and ammonia by cystathionine γ-lyase (CGL) in the reverse transsulfuration pathway. The protozoan parasite Toxoplasma gondii, the causative agent of toxoplasmosis, includes both CBS and CGL enzymes. We have recently reported that the putative T. gondii CGL gene encodes a functional enzyme. Herein, we cloned and biochemically characterized cDNA encoding CBS from T. gondii (TgCBS), which represents a first example of protozoan CBS that does not bind heme but possesses two C-terminal CBS domains. We demonstrated that TgCBS can use both serine and O-acetylserine to produce cystathionine, converting these substrates to an aminoacrylate intermediate as part of a PLP-catalyzed ß-replacement reaction. Besides a role in cysteine biosynthesis, TgCBS can also efficiently produce hydrogen sulfide, preferentially via condensation of cysteine and homocysteine. Unlike the human counterpart and similar to CBS enzymes from lower organisms, the TgCBS activity is not stimulated by S-adenosylmethionine. This study establishes the presence of an intact functional reverse transsulfuration pathway in T. gondii and demonstrates the crucial role of TgCBS in biogenesis of H2S.


Subject(s)
Cystathionine beta-Synthase/metabolism , Cysteine/biosynthesis , Hydrogen Sulfide/metabolism , Toxoplasma/enzymology , Toxoplasma/genetics , Biocatalysis , Cystathionine/biosynthesis , Cystathionine beta-Synthase/genetics , Cystathionine gamma-Lyase/metabolism , DNA, Complementary , Genes, Protozoan , Heme/metabolism , Homocysteine/metabolism , Kinetics , Serine/analogs & derivatives , Serine/metabolism
11.
Med Sci Monit ; 25: 6313-6321, 2019 Aug 22.
Article in English | MEDLINE | ID: mdl-31437131

ABSTRACT

BACKGROUND To explore the protective effects of Shexiang Tongxin Dropping Pill (STP) in improving peripheral microvascular dysfunction in mice and to explore the involved mechanism. MATERIAL AND METHODS A peripheral microvascular dysfunction model was established by combined myocardial infarction (MI) and lipopolysaccharide (LPS) injection in mice. Then, the mice were randomized into a model group (n=10) or an STP group (n=10), which were treated with normal saline and STP, respectively. The cremaster muscle microvascular blood flow velocity and numbers of leukocytes adherent to the venular wall were evaluated before and after drug intervention. We assessed the expression of adhesion molecule CD11b and related transcript factor FOXO1 in leukocytes, cystathionine-γ-lyase (CSE) mRNA expression in the cremaster muscle, and mitochondrial DNA copy numbers. RESULTS Compared with those of control mice, the cremaster microvascular blood flow velocity, cremaster CSE expression, and mitochondrial DNA copy number in mice from the model group were significantly lower and leukocyte adhesion and CD11b and FOXO1 expression were significantly higher. Intervention with STP could significantly increase the cremaster microvascular flow velocity (0.480±0.010 mm/s vs. 0.075±0.005 mm/s), mRNA expression of cremaster CSE, and mitochondrial DNA copy number, but it inhibited leukocyte adhesion and decreased leukocyte CD11b and FOXO1 expression. CONCLUSIONS STP significantly improved peripheral microcirculation, in which increased CSE expression might be the underlying mechanism.


Subject(s)
Cystathionine gamma-Lyase/metabolism , Drugs, Chinese Herbal/pharmacology , Microvessels/drug effects , Animals , Blood Flow Velocity/drug effects , CD11b Antigen/analysis , Cell Adhesion/drug effects , Cystathionine gamma-Lyase/analysis , Drugs, Chinese Herbal/metabolism , Forkhead Box Protein O1/analysis , Hydrogen Sulfide/pharmacology , Leukocytes/metabolism , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , Microcirculation/drug effects , Muscles/blood supply , Random Allocation , Regional Blood Flow/drug effects
12.
Br J Pharmacol ; 176(17): 3180-3192, 2019 09.
Article in English | MEDLINE | ID: mdl-31140595

ABSTRACT

BACKGROUND AND PURPOSE: Hydrogen sulfide donors can block the cardiovascular injury of hyperhomocysteinemia. H2 S also lowers serum homocysteine in rats with mild hyperhomocysteinemia, but the pharmacological mechanism is unknown. The present study investigated the mechanism(s) involved. EXPERIMENTAL APPROACH: ApoE-knockout mice were fed a Paigen diet and L-methionine in drinking water for 16 weeks to create a mouse model of atherosclerosis with hyperhomocysteinemia. H2 S donors (NaHS and GYY4137) were administered by intraperitoneal injection. We also assayed the H2 S produced (by methylene blue assay and mito-HS [H2 S fluorescence probe]), cystathionine γ lyase (CSE) mRNA and protein expression, and CSE sulfhydration and nitrosylation and its activity. KEY RESULTS: H2 S donor treatment significantly lowered atherosclerotic plaque area, macrophage infiltration, and serum homocysteine level in the mouse model of atherosclerosis with co-existing hyperhomocysteinemia. mRNA and protein levels of CSE, a key enzyme catalyzing homocysteine trans-sulfuration, were down-regulated with hyperhomocysteinemia, and CSE catalytic activity was inhibited. All these effects were reversed with H2 S donor treatment. Hyperhomocysteinemia induced CSE nitrosylation, whereas H2 S sulfhydrated CSE at the same cysteine residues. Nitrosylated CSE decreased and sulfhydrated CSE increased its catalytic and binding activities towards L-homocysteine. Mutation of C252, C255, C307, and C310 residues in CSE abolished CSE nitrosylation or sulfhydration and prevented its binding to L-homocysteine. CONCLUSIONS AND IMPLICATIONS: Sulfhydration or nitrosylation of CSE represents a yin/yang regulation of catalysis or binding to L-homocysteine. H2 S donor treatment enhanced CSE sulfhydration, thus lowering serum L-homocysteine, which contributed in part to the anti-atherosclerosis effects in ApoE-knockout mice with hyperhomocysteinemia.


Subject(s)
Atherosclerosis/drug therapy , Cystathionine gamma-Lyase/metabolism , Hydrogen Sulfide/pharmacology , Hyperhomocysteinemia/drug therapy , Animals , Atherosclerosis/metabolism , Cells, Cultured , Disease Models, Animal , Dose-Response Relationship, Drug , HEK293 Cells , Hep G2 Cells , Humans , Hydrogen Sulfide/analysis , Hydrogen Sulfide/metabolism , Hyperhomocysteinemia/metabolism , Male , Mice , Mice, Knockout, ApoE , Structure-Activity Relationship
13.
Biol Reprod ; 101(1): 208-222, 2019 07 01.
Article in English | MEDLINE | ID: mdl-30998234

ABSTRACT

Oxidative stress is considered a major mechanism causing sperm damage during cryopreservation and storage, and underlies male factor infertility. Currently, oxidative stress is no longer believed to be caused only by the overproduction of reactive oxygen species, but rather by the deregulation of redox signaling and control mechanisms. With this concept in mind, here, we describe for the first time the presence of the soluble carrier family 7 member 11 (SLC7A11) antiporter, which exchanges extracellular cystine (Cyss) for intracellular glutamate, in stallion spermatozoa, as well as its impact on sperm function using the specific inhibitor sulfasalazine. Spermatozoa incubated with Cyss exhibited an increased intracellular GSH content compared with controls (P < 0.01): 50% in fresh extended stallion spermatozoa and 30% in frozen-thawed spermatozoa. This effect was prevented by the addition of sulfasalazine to the media. Cystine supplementation also reduced the oxidation-reduction potential of spermatozoa, with sulfasalazine only preventing this effect on fresh spermatozoa that were incubated for 3 h at 37°C, but not in frozen-thawed spermatozoa. While sulfasalazine reduced the motility of frozen-thawed spermatozoa, it increased motility in fresh samples. The present findings provide new and relevant data on the mechanism regulating the redox status of spermatozoa and suggest that a different redox regulatory mechanism exists in cryopreserved spermatozoa, thus providing new clues to improve current cryopreservation technologies and treat male factor infertility.


Subject(s)
Amino Acid Transport System y+/metabolism , Cystine/metabolism , Horses/metabolism , Spermatozoa/metabolism , Animals , Cystathionine gamma-Lyase/metabolism , Cystine/pharmacology , Glutathione/metabolism , Male , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Oxidative Stress/physiology , Reactive Oxygen Species/metabolism , Semen Analysis/methods , Semen Analysis/veterinary , Semen Preservation/methods , Semen Preservation/veterinary , Spermatozoa/drug effects
14.
Article in English | MEDLINE | ID: mdl-30658157

ABSTRACT

We recently reported that celecoxib, a selective cyclooxygenase-2 (COX2) inhibitor, counteracts the adverse circulatory and renal actions of cyclosporine (CSA). Despite the seemingly advantageous nature of this interaction particularly in clinical settings that necessitate the combined use of the two drugs such as immune-related arthritis, the underlying mechanism remains elusive. This prompted us to test the hypothesis that the facilitation of the cystathionine-γ-lyase (CSE)/hydrogen sulfide (H2S) signaling accounts for such favorable effects of celecoxib on CSA nephrotoxicity. The data showed that the 10-day co-treatment of rats with celecoxib (10 mg/kg/day) ameliorated the hypertensive and biochemical and renal structural damages caused by CSA (20 mg/kg/day). Celecoxib also reversed the CSA-evoked (i) reductions in the tubular and glomerular protein expression of CSE and levels of H2S, prostaglandin E2 (PGE2), and total antioxidant capacity (TAC), and (ii) increases in inflammatory (tumor necrosis factor-α, TNF-α), fibrotic (transforming growth factor-ß1, TGF-ß1) and apoptotic (caspase-3) cytokines. These celecoxib effects disappeared when rats were treated concomitantly with the CSE inhibitor DL-propargylglycine (DL-PAG), indicating the importance of the CSE-derived H2S in mediating the renoprotective action of celecoxib. This view is bolstered by the observation that the beneficial hemodynamic and renal actions of celecoxib were replicated after supplementation of rats with sodium sulfide (Na2S, H2S donor). Together, the increased abundance of renal CSE and H2S and subsequent dampening down of inflammatory, fibrotic, oxidant, and apoptotic pathways play pivotal roles in the capacity of celecoxib to compromise the troublesome hypertensive and nephrotoxic insults caused by CSA in rats.


Subject(s)
Celecoxib/pharmacology , Cyclosporine/toxicity , Cystathionine gamma-Lyase/metabolism , Hydrogen Sulfide/metabolism , Hypertension/drug therapy , Kidney/drug effects , Up-Regulation/drug effects , Alkynes/pharmacology , Animals , Apoptosis/drug effects , Celecoxib/therapeutic use , Cytokines/metabolism , Drug Interactions , Glycine/analogs & derivatives , Glycine/pharmacology , Hemodynamics/drug effects , Hypertension/chemically induced , Hypertension/pathology , Hypertension/physiopathology , Kidney/metabolism , Kidney/pathology , Kidney/physiopathology , Male , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Signal Transduction
15.
Sci Rep ; 9(1): 684, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30679627

ABSTRACT

Biosynthesis of hydrogen sulfide (H2S), a key signalling molecule in human (patho)physiology, is mostly accomplished by the human enzymes cystathionine ß-synthase (CBS), cystathionine γ-lyase (CSE) and 3-mercaptopyruvate sulfurtransferase (MST). Several lines of evidence have shown a close correlation between increased H2S production and human diseases, such as several cancer types and amyotrophic lateral sclerosis. Identifying compounds selectively and potently inhibiting the human H2S-synthesizing enzymes may therefore prove beneficial for pharmacological applications. Here, the human enzymes CBS, CSE and MST were expressed and purified from Escherichia coli, and thirty-one pyridine derivatives were synthesized and screened for their ability to bind and inhibit these enzymes. Using differential scanning fluorimetry (DSF), surface plasmon resonance (SPR), circular dichroism spectropolarimetry (CD), and activity assays based on fluorimetric and colorimetric H2S detection, two compounds (C30 and C31) sharing structural similarities were found to weakly inhibit both CBS and CSE: 1 mM C30 inhibited these enzymes by approx. 50% and 40%, respectively, while 0.5 mM C31 accounted for CBS and CSE inhibition by approx. 40% and 60%, respectively. This work, while presenting a robust methodological platform for screening putative inhibitors of the human H2S-synthesizing enzymes, highlights the importance of employing complementary methodologies in compound screenings.


Subject(s)
Cystathionine beta-Synthase/antagonists & inhibitors , Cystathionine gamma-Lyase/antagonists & inhibitors , Hydrogen Sulfide/metabolism , Pyridines/pharmacology , Sulfurtransferases/antagonists & inhibitors , Circular Dichroism , Cystathionine beta-Synthase/metabolism , Cystathionine gamma-Lyase/metabolism , Drug Evaluation, Preclinical/methods , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Fluorometry/methods , Humans , Methylene Blue , Pyridines/chemistry , Sulfurtransferases/metabolism , Surface Plasmon Resonance
16.
Physiol Behav ; 188: 128-133, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29425970

ABSTRACT

Hydrogen sulfide (H2S) is an endogenous neuromodulator produced mainly by the enzyme cystathionine gamma-lyase (CSE) in peripheral tissues. A pronociceptive role of endogenously produced H2S has been previously reported by our group in a model of orofacial inflammatory pain. Using the established persistent orofacial pain rat model induced by complete Freund's adjuvant (CFA) injection into temporomandibular joint (TMJ), we have now investigated the putative role of endogenous H2S modulating hypernociceptive responses. Additionally, plasmatic extravasation on TMJ was measured following different treatments by Evans blue dye quantification. Thus, rats were submitted to Von Frey and Formalin tests in orofacial region before and after pharmacological inhibition of the CSE-H2S system combined or not with CFA-induced TMJ inflammation. Pretreatment with CSE inhibitor, propargylglycine (PAG; 88.4 µmol/kg) reduced temporomandibular inflammatory pain when injected locally as well as systemically. In particular, local PAG injection seems to be more effective for hypernociceptive responses in orofacial persistent inflammation since its action is evidenced in the majority analyzed periods of the inflammatory process compared to its systemic use. Moreover, local injection seems to act on temporomandibular vascular permeability, evidenced by decreased plasmatic extravasation induced by local PAG administration. Our data are consistent with the notion that the endogenous synthetized gas H2S modulates persistent orofacial pain responses revealing the pharmacological importance of the CSE inhibitor as a possible therapeutic target for their control.


Subject(s)
Cystathionine gamma-Lyase/metabolism , Facial Pain/enzymology , Facial Pain/etiology , Inflammation/complications , Inflammation/pathology , Temporomandibular Joint/pathology , Alkynes/therapeutic use , Analysis of Variance , Animals , Enzyme Inhibitors/therapeutic use , Facial Pain/complications , Facial Pain/drug therapy , Freund's Adjuvant/toxicity , Glycine/analogs & derivatives , Glycine/therapeutic use , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Inflammation/chemically induced , Male , Pain Measurement , Rats , Rats, Wistar , Time Factors , Treatment Outcome
17.
Toxicology ; 393: 102-112, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29129814

ABSTRACT

Cav3.2 T-type Ca2+ channel activity is suppressed by zinc that binds to the extracellular histidine-191 of Cav3.2, and enhanced by H2S that interacts with zinc. Cav3.2 in nociceptors is upregulated in an activity-dependent manner. The enhanced Cav3.2 activity by H2S formed by the upregulated cystathionine-γ-lyase (CSE) is involved in the cyclophosphamide (CPA)-induced cystitis-related bladder pain in mice. We thus asked if zinc deficiency affects the cystitis-related bladder pain in mice by altering Cav3.2 function and/or expression. Dietary zinc deficiency for 2 weeks greatly decreased zinc concentrations in the plasma but not bladder tissue, and enhanced the bladder pain/referred hyperalgesia (BP/RH) following CPA at 200mg/kg, a subeffective dose, but not 400mg/kg, a maximal dose, an effect abolished by pharmacological blockade or gene silencing of Cav3.2. Acute zinc deficiency caused by systemic N,N,N',N'-tetrakis-(2-pyridylmethyl)-ethylendiamine (TPEN), a zinc chelator, mimicked the dietary zinc deficiency-induced Cav3.2-dependent promotion of BP/RH following CPA at 200mg/kg. CPA at 400mg/kg alone or TPEN plus CPA at 200mg/kg caused Cav3.2 overexpression accompanied by upregulation of Egr-1 and USP5, known to promote transcriptional expression and reduce proteasomal degradation of Cav3.2, respectively, in the dorsal root ganglia (DRG). The CSE inhibitor, ß-cyano-l-alanine, prevented the BP/RH and upregulation of Cav3.2, Egr-1 and USP5 in DRG following TPEN plus CPA at 200mg/kg. Together, zinc deficiency promotes bladder pain accompanying CPA-induced cystitis by enhancing function and expression of Cav3.2 in nociceptors, suggesting a novel therapeutic avenue for treatment of bladder pain, such as zinc supplementation.


Subject(s)
Calcium Channels, T-Type/metabolism , Cystitis/metabolism , Pain/metabolism , Zinc/deficiency , Animals , Chelating Agents/pharmacology , Cyclophosphamide , Cystathionine gamma-Lyase/metabolism , Cystitis/chemically induced , Diet , Disease Models, Animal , Ethylamines , Ganglia, Spinal/metabolism , Mice , Oligodeoxyribonucleotides/pharmacology , Pyridines , Urinary Bladder/metabolism , Zinc/blood , Zinc/metabolism
18.
Arch Toxicol ; 92(3): 1249-1265, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29222744

ABSTRACT

Nonsteroidal anti-inflammatory drugs (NSAIDs)-induced small intestinal injury (enteropathy) occurs in about two-thirds of regular NSAID users. To date, there is no proven-effective treatment for NSAID enteropathy, and its underlying mechanism remains obscure. The present study showed that glucocorticoids are an important determinant of NSAID enteropathy. High dose dexamethasone (DEX, 75 mg/kg) markedly exacerbated the acute toxicity of ibuprofen (IBU, 200 mg/kg) in the small intestine of mice, which was not due to the pregnane-X-receptor pathway. Instead, glucocorticoid receptor (GR) mediated the effect of DEX (5 mg/kg) on both the acute (200 mg/kg) and 7-day repeated-dose (50 mg/kg) toxicity of IBU in the small intestine. Combined treatment of DEX (5 mg/kg) and IBU (50 mg/kg) synergistically repressed the intestinal farnesoid X receptor (FXR)-cystathionine-γ-lyase signaling, which was accompanied with an elevation in the biliary excretion of bile acids, especially the FXR antagonist tauro-ß-muricholic acid. DEX (5 mg/kg) also activated intestinal peroxisome proliferator-activated receptor α (PPARα)-UDP-glucuronosyltransferase (UGT) pathway, which increased the formation and enterohepatic circulation of IBU-acyl glucuronide. Furthermore, DEX (5 mg/kg) and IBU (50 mg/kg) altered the intestinal microbial composition, characterized with a marked decrease in Actinobacteria. To conclude, the present study for the first time suggests that glucocorticoids play vital roles in control of IBU enteropathy via intestinal GR-FXR and PPARα-UGT signaling.


Subject(s)
Glucuronosyltransferase/metabolism , Ibuprofen/adverse effects , Intestinal Diseases/chemically induced , Intestinal Diseases/metabolism , Receptors, Glucocorticoid/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Bile Acids and Salts/metabolism , Cystathionine gamma-Lyase/metabolism , Dexamethasone/adverse effects , Intestine, Small/drug effects , Intestine, Small/metabolism , Intestine, Small/pathology , Male , Mice, Inbred C57BL , PPAR alpha/metabolism , Rats, Wistar , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction/drug effects , Taurocholic Acid/analogs & derivatives , Taurocholic Acid/metabolism , Taurocholic Acid/pharmacology
19.
Biochem Pharmacol ; 149: 143-152, 2018 03.
Article in English | MEDLINE | ID: mdl-29248598

ABSTRACT

Hydrogen sulfide (H2S) is mostly produced by cystathionine-gamma-lyase (CSE) in vascular system and it inhibits the proliferation of vascular smooth muscle cells (SMCs). Insulin-like growth factor-1 (IGF-1), via its receptor (IGF-1R), exerts multiple physiological and pathophysiological effects on the vasculature, including stimulating SMC proliferation and migration, and inhibiting SMC apoptosis. Since H2S and IGF-1/IGF-1R have opposite effects on SMC proliferation, it becomes imperative to better understand the interaction of these two signaling mechanisms on SMC proliferation. SMCs isolated from small mesenteric arteries of CSE knockout (KO) and wild-type (WT) mice were used in the present study. The effects of IGF-1 and H2S on SMC proliferation were evaluated with 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and bromodeoxyuridine (BrdU) assays. Protein expression was determined by western blot, and H2S-induced protein S-sulfhydration was assessed with a modified biotin switch assay. We found that IGF-1 dose-dependently increased the proliferation of both WT-SMCs and KO-SMCs, and this effect was more significant in KO-SMCs. Supplement of sodium hydrosulfide (NaHS) inhibited IGF-1-induced cell proliferation, while this effect was abolished by blocking IGF-1/IGF-1R signaling with picropodophyllin (PPP) or knocking out of the expression of IGF-1R. H2S significantly down-regulates the expression of IGF-1R, stimulates IGF-1R S-sulfhydration, and attenuates the binding of IGF-1 with IGF-1R. This study provides novel insight on the involvement of IGF-1/IGF-1R in H2S-inhibited SMC proliferation and suggests H2S-based innovative treatment strategies for proliferative cardiovascular diseases such as atherosclerosis.


Subject(s)
Cell Proliferation/drug effects , Hydrogen Sulfide/pharmacology , Insulin-Like Growth Factor I/pharmacology , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Receptor, IGF Type 1/metabolism , Animals , Cells, Cultured , Cystathionine gamma-Lyase/genetics , Cystathionine gamma-Lyase/metabolism , Gene Expression Regulation/drug effects , Hydrogen Sulfide/metabolism , Insulin-Like Growth Factor I/metabolism , Mice , Mice, Knockout , Podophyllotoxin/analogs & derivatives , RNA/genetics , RNA/metabolism , Receptor, IGF Type 1/genetics
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL