Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 763
Filter
1.
Geroscience ; 46(5): 4275-4314, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38558215

ABSTRACT

Down syndrome (DS) is a genetic condition where the person is born with an extra chromosome 21. DS is associated with accelerated aging; people with DS are prone to age-related neurological conditions including an early-onset Alzheimer's disease. Using the Dp(17)3Yey/ + mice, which overexpresses a portion of mouse chromosome 17, which encodes for the transsulfuration enzyme cystathionine ß-synthase (CBS), we investigated the functional role of the CBS/hydrogen sulfide (H2S) pathway in the pathogenesis of neurobehavioral dysfunction in DS. The data demonstrate that CBS is higher in the brain of the DS mice than in the brain of wild-type mice, with primary localization in astrocytes. DS mice exhibited impaired recognition memory and spatial learning, loss of synaptosomal function, endoplasmic reticulum stress, and autophagy. Treatment of mice with aminooxyacetate, a prototypical CBS inhibitor, improved neurobehavioral function, reduced the degree of reactive gliosis in the DS brain, increased the ability of the synaptosomes to generate ATP, and reduced endoplasmic reticulum stress. H2S levels in the brain of DS mice were higher than in wild-type mice, but, unexpectedly, protein persulfidation was decreased. Many of the above alterations were more pronounced in the female DS mice. There was a significant dysregulation of metabolism in the brain of DS mice, which affected amino acid, carbohydrate, lipid, endocannabinoid, and nucleotide metabolites; some of these alterations were reversed by treatment of the mice with the CBS inhibitor. Thus, the CBS/H2S pathway contributes to the pathogenesis of neurological dysfunction in DS in the current animal model.


Subject(s)
Autophagy , Cystathionine beta-Synthase , Disease Models, Animal , Down Syndrome , Endoplasmic Reticulum Stress , Hydrogen Sulfide , Up-Regulation , Animals , Cystathionine beta-Synthase/metabolism , Cystathionine beta-Synthase/genetics , Down Syndrome/metabolism , Down Syndrome/physiopathology , Down Syndrome/genetics , Hydrogen Sulfide/metabolism , Mice , Endoplasmic Reticulum Stress/physiology , Brain/metabolism , Aminooxyacetic Acid/pharmacology , Behavior, Animal , Male , Female , Synapses/metabolism
2.
Hypertens Res ; 47(5): 1323-1337, 2024 May.
Article in English | MEDLINE | ID: mdl-38491106

ABSTRACT

Paroxysmal sympathetic hyperactivity (PSH) is a common clinical feature secondary to ischemic stroke (IS), but its mechanism is poorly understood. We aimed to investigate the role of H2S in the pathogenesis of PSH. IS patients were divided into malignant (MCI) and non-malignant cerebral infarction (NMCI) group. IS in rats was induced by the right middle cerebral artery occlusion (MCAO). H2S donor (NaHS) or inhibitor (aminooxy-acetic acid, AOAA) were microinjected into the hypothalamic paraventricular nucleus (PVN). Compared with the NMCI group, patients in the MCI group showed PSH, including tachycardia, hypertension, and more plasma norepinephrine (NE) that was positively correlated with levels of creatine kinase, glutamate transaminase, and creatinine respectively. The 1-year survival rate of patients with high plasma NE levels was lower. The hypothalamus of rats with MCAO showed increased activity, especially in the PVN region. The levels of H2S in PVN of the rats with MCAO were reduced, while the blood pressure and renal sympathetic discharge were increased, which could be ameliorated by NaHS and exacerbated by AOAA. NaHS completely reduced the disulfide bond of NMDAR1 in PC12 cells. The inhibition of NMDAR by MK-801 microinjected in PVN of rats with MCAO also could lower blood pressure and renal sympathetic discharge. In conclusion, PSH may be associated with disease progression and survival in patients with IS. Decreased levels of H2S in PVN were involved in regulating sympathetic efferent activity after cerebral infarction. Our results might provide a new strategy and target for the prevention and treatment of PSH.


Subject(s)
Hydrogen Sulfide , Paraventricular Hypothalamic Nucleus , Animals , Paraventricular Hypothalamic Nucleus/metabolism , Paraventricular Hypothalamic Nucleus/drug effects , Hydrogen Sulfide/metabolism , Hydrogen Sulfide/blood , Male , Rats , Humans , Aged , Cerebral Infarction , Middle Aged , Rats, Sprague-Dawley , Female , Norepinephrine/blood , Autonomic Nervous System Diseases , Aminooxyacetic Acid/pharmacology , Sympathetic Nervous System/physiopathology , Sympathetic Nervous System/drug effects , Infarction, Middle Cerebral Artery/complications , Blood Pressure/drug effects
3.
J Neuroimmunol ; 389: 578324, 2024 04 15.
Article in English | MEDLINE | ID: mdl-38422691

ABSTRACT

Sickness behavior reflects a state of altered physiology and central nervous system function that occurs during systemic infection or inflammation, serving as an adaptive response to illness. This study aims to elucidate the role of hydrogen sulfide (H2S) in regulating sickness behavior and neuroinflammatory responses in a rat model of systemic inflammation. Adult male Wistar rats were treated with lipopolysaccharide (LPS) to induce sickness behavior. Intracerebroventricular (i.c.v.) pretreatments included aminooxyacetic acid (AOAA), an inhibitor of H2S synthesis, and sodium sulfide (NaHS), an H2S donor. Behavioral assays were conducted, along with the assessment of astrocyte activation, as indicated by GFAP expression in the hypothalamus. Pretreatment with NaHS mitigated LPS-induced behavioral changes, including hypophagia, social and exploratory deficits, without affecting peripheral cytokine levels, indicating a central modulatory effect. AOAA, conversely, accentuated certain behavioral responses, suggesting a complex role of endogenous H2S in sickness behavior. These findings were reinforced by a lack of effect on plasma interleukin levels but significant reduction in GFAP expression. Our findings support the central role of H2S in modulating neuroinflammation and sickness behavior, highlighting the therapeutic potential of targeting H2S signaling in neuroinflammatory conditions.


Subject(s)
Hydrogen Sulfide , Sulfides , Rats , Male , Animals , Hydrogen Sulfide/pharmacology , Hydrogen Sulfide/therapeutic use , Lipopolysaccharides/toxicity , Illness Behavior , Rats, Wistar , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/metabolism , Aminooxyacetic Acid/pharmacology , Neurotransmitter Agents
4.
Biomed Pharmacother ; 167: 115605, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37801901

ABSTRACT

The second most common cancer among men is prostate cancer, which is also the fifth leading reason for male cancer deaths worldwide. Bone metastases are the main factor affecting the prognosis of prostate cancer. Consequently, antitumor and anti-prostate cancer-induced bone destruction medicines are urgently needed. We previously discovered that aminooxyacetic acid hemihydrochloride (AOAA) suppressed bone resorption and osteoclast growth by decreasing adenosine triphosphate (ATP) production and limiting oxidative phosphorylation (OXPHOS). Here, we evaluated the impacts of AOAA on prostate cancer RM-1 cells in vitro. It's found that AOAA significantly inhibited cell proliferation, migration, and invasiveness, decreased ATP levels, increased ROS, halted the cell cycle phase, and triggered apoptosis. AOAA also decreased mitochondrial membrane potential and the ability to uptake glucose, suggesting that the antitumor effects of AOAA were expressed through the inhibition of OXPHOS and glycolysis. Furthermore, we assessed the effects of AOAA in vivo using a prostate cancer-induced bone osteolysis mice model. AOAA also delayed tumor growth and bone destruction in vivo. On the whole, our findings imply that AOAA may potentially have therapeutic effects on prostate cancer and prostate cancer-induced osteolysis.


Subject(s)
Osteolysis , Prostatic Neoplasms , Mice , Animals , Male , Humans , Aminooxyacetic Acid/pharmacology , Adenosine Triphosphate/metabolism , Energy Metabolism , Prostatic Neoplasms/drug therapy , Cell Cycle , Cell Line, Tumor
5.
Toxicology ; 495: 153611, 2023 08 15.
Article in English | MEDLINE | ID: mdl-37544576

ABSTRACT

Trichloroethylene (TCE) is a known human carcinogen with toxicity attributed to its metabolism. S-(1,2-Dichlorovinyl)-L-cysteine (DCVC) is a metabolite of TCE formed downstream in TCE glutathione (GSH) conjugation and is upstream of several toxic metabolites. Despite knowledge that DCVC stimulates reactive oxygen species (ROS) generation and apoptosis in placental cells, the extent to which these outcomes are attributable to DCVC metabolism is unknown. The current study used N-acetyl-L-cysteine (NAC) at 5 mM and aminooxyacetic acid (AOAA) at 1 mM as pharmacological modifiers of DCVC metabolism to investigate DCVC toxicity at concentrations of 5-50 µM in the human placental trophoblast BeWo cell model capable of forskolin-stimulated syncytialization. Exposures of unsyncytialized BeWo cells, BeWo cells undergoing syncytialization, and syncytialized BeWo cells were studied. NAC pre/co-treatment with DCVC either failed to inhibit or exacerbated DCVC-induced H2O2 abundance, PRDX2 mRNA expression, and BCL2 mRNA expression. Although NAC increased mRNA expression of CYP3A4, which would be consistent with increased generation of the toxic metabolite N-acetyl-DCVC sulfoxide (NAcDCVCS), a CYP3A4 inhibitor ketoconazole did not significantly alter BeWo cell responses. Moreover, AOAA failed to inhibit cysteine conjugate ß-lyase (CCBL), which bioactivates DCVC, and did not affect the percentage of nuclei condensed or fragmented, a measure of apoptosis, in all BeWo cell models. However, syncytialized cells had higher CCBL activity compared to unsyncytialized cells, suggesting that the former may be more sensitive to DCVC toxicity. Together, although neither NAC nor AOAA mitigated DCVC toxicity, differences in CCBL activity and potentially CYP3A4 expression dictated the differential toxicity derived from DCVC.


Subject(s)
Acetylcysteine , Trichloroethylene , Humans , Female , Pregnancy , Acetylcysteine/pharmacology , Acetylcysteine/metabolism , Cysteine , Trichloroethylene/toxicity , Trichloroethylene/metabolism , Placenta/metabolism , Aminooxyacetic Acid/metabolism , Aminooxyacetic Acid/pharmacology , Trophoblasts/metabolism , Cytochrome P-450 CYP3A/metabolism , Hydrogen Peroxide/metabolism , RNA, Messenger/metabolism
6.
Cell Mol Life Sci ; 79(8): 438, 2022 Jul 21.
Article in English | MEDLINE | ID: mdl-35864237

ABSTRACT

Cystathionine beta-synthase (CBS) is a pivotal enzyme of the transsulfuration pathway responsible for diverting homocysteine to the biosynthesis of cysteine and production of hydrogen sulfide (H2S). Aberrant upregulation of CBS and overproduction of H2S contribute to pathophysiology of several diseases including cancer and Down syndrome. Therefore, pharmacological CBS inhibition has emerged as a prospective therapeutic approach. Here, we characterized binding and inhibitory mechanism of aminooxyacetic acid (AOAA), the most commonly used CBS inhibitor. We found that AOAA binds CBS tighter than its respective substrates and forms a dead-end PLP-bound intermediate featuring an oxime bond. Surprisingly, serine, but not cysteine, replaced AOAA from CBS and formed an aminoacrylate reaction intermediate, which allowed for the continuation of the catalytic cycle. Indeed, serine rescued and essentially normalized the enzymatic activity of AOAA-inhibited CBS. Cellular studies confirmed that AOAA decreased H2S production and bioenergetics, while additional serine rescued CBS activity, H2S production and mitochondrial function. The crystal structure of AOAA-bound human CBS showed a lack of hydrogen bonding with residues G305 and Y308, found in the serine-bound model. Thus, AOAA-inhibited CBS could be reactivated by serine. This difference may be important in a cellular environment in multiple pathophysiological conditions and may modulate the CBS-inhibitory activity of AOAA. In addition, our results demonstrate additional complexities of using AOAA as a CBS-specific inhibitor of H2S biogenesis and point to the urgent need to develop a potent, selective and specific pharmacological CBS inhibitor.


Subject(s)
Cystathionine beta-Synthase , Hydrogen Sulfide , Aminooxyacetic Acid/pharmacology , Cystathionine beta-Synthase/metabolism , Cysteine , Humans , Hydrogen Sulfide/metabolism , Hydrogen Sulfide/pharmacology , Serine
7.
Front Immunol ; 13: 837669, 2022.
Article in English | MEDLINE | ID: mdl-35251031

ABSTRACT

Targeting T cell metabolism is an established method of immunomodulation. Following activation, T cells engage distinct metabolic programs leading to the uptake and processing of nutrients that determine cell proliferation and differentiation. Redirection of T cell fate by modulation of these metabolic programs has been shown to boost or suppress immune responses in vitro and in vivo. Using publicly available T cell transcriptomic and proteomic datasets we identified vitamin B6-dependent transaminases as key metabolic enzymes driving T cell activation and differentiation. Inhibition of vitamin B6 metabolism using the pyridoxal 5'-phosphate (PLP) inhibitor, aminoxyacetic acid (AOA), suppresses CD8+ T cell proliferation and effector differentiation in a dose-dependent manner. We show that pyridoxal phosphate phosphatase (PDXP), a negative regulator of intracellular vitamin B6 levels, is under the control of the hypoxia-inducible transcription factor (HIF1), a central driver of T cell metabolism. Furthermore, by adoptive transfer of CD8 T cells into a C57BL/6 mouse melanoma model, we demonstrate the requirement for vitamin B6-dependent enzyme activity in mediating effective anti-tumor responses. Our findings show that vitamin B6 metabolism is required for CD8+ T cell proliferation and effector differentiation in vitro and in vivo. Targeting vitamin B6 metabolism may therefore serve as an immunodulatory strategy to improve anti-tumor immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes , Neoplasms , Vitamin B 6 , Aminooxyacetic Acid/pharmacology , Animals , CD8-Positive T-Lymphocytes/immunology , Hypoxia-Inducible Factor 1, alpha Subunit , Melanoma/immunology , Mice , Mice, Inbred C57BL , Neoplasms/immunology , Phosphoprotein Phosphatases , Proteomics , Pyridoxal Phosphate/antagonists & inhibitors , Vitamin B 6/metabolism
8.
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
9.
Arch Toxicol ; 95(4): 1303-1321, 2021 04.
Article in English | MEDLINE | ID: mdl-33599830

ABSTRACT

Exposure to the industrial solvent trichloroethylene (TCE) has been associated with adverse pregnancy outcomes in humans and decreased fetal weight in rats. TCE kidney toxicity can occur through formation of reactive metabolites via its glutathione (GSH) conjugation metabolic pathway, largely unstudied in the context of pregnancy. To investigate the contribution of the GSH conjugation pathway and oxidative stress to TCE toxicity during pregnancy, we exposed rats orally to 480 mg TCE/kg/day from gestational day (GD) 6 to GD 16 with and without N-acetyl-L-cysteine (NAC) at 200 mg/kg/day or aminooxyacetic acid (AOAA) at 20 mg/kg/day as pre/co-treatments from GD 5-16. NAC is a reactive oxygen species scavenger that modifies the GSH conjugation pathway, and AOAA is an inhibitor of cysteine conjugate ß-lyase (CCBL) in the GSH conjugation pathway. TCE decreased fetal weight, and this was prevented by AOAA but not NAC pre/co-treatment to TCE. Although AOAA inhibited CCBL activity in maternal kidney, it did not inhibit CCBL activity in maternal liver and placenta, suggesting that AOAA prevention of TCE-induced decreased fetal weight was due to CCBL activity inhibition in the kidneys but not liver or placenta. Unexpectedly, NAC pre/co-treatment with TCE, relative to TCE treatment alone, altered placental morphology consistent with delayed developmental phenotype. Immunohistochemical staining revealed that the decidua basale, relative to basal and labyrinth zones, expressed the highest abundance of CCBL1, flavin-containing monooxygenase 3, and cleaved caspase-3. Together, the findings show the differential effects of NAC and AOAA on TCE-induced pregnancy outcomes are likely attributable to TCE metabolism modulation.


Subject(s)
Acetylcysteine/pharmacology , Aminooxyacetic Acid/pharmacology , Reproduction/drug effects , Trichloroethylene/toxicity , Animals , Enzyme Inhibitors/pharmacology , Female , Free Radical Scavengers/pharmacology , Glutathione/metabolism , Male , Oxidative Stress/drug effects , Placenta/drug effects , Pregnancy , Pregnancy Outcome , Rats , Rats, Wistar , Solvents/metabolism , Solvents/toxicity , Trichloroethylene/metabolism
10.
Nitric Oxide ; 104-105: 44-50, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32891752

ABSTRACT

We recently reported that hydrogen sulfide (H2S) is a possible relaxation factor in the rat bladder. However, there is no available information about the roles of central H2S in the micturition reflex, so we investigated the effects of centrally administered GYY4137 (H2S donor) and AOAA (H2S synthesis inhibitor) on the micturition reflex in urethane-anesthetized (0.8 g/kg, ip) male Wistar rats. Cystometry was performed before and after the administration of GYY4137 (3 or 10 nmol/rat, icv) or AOAA (30 or 100 µg/rat, icv). In some rats, SR95531 (GABAA receptor antagonist, 0.1 nmol/rat, icv) or SCH50911 (GABAB receptor antagonist, 0.1 nmol/rat, icv) was administered 30 min before GYY4137 administration (10 nmol/rat, icv). Centrally administered GYY4137 dose-dependently prolonged the intercontraction intervals (ICI) without altering maximum voiding pressure (MVP). On the other hand, centrally administered AOAA dose-dependently shortened ICI without altering MVP. The AOAA (30 µg/rat, icv)-induced ICI shortening was reversed in the central presence of GYY4137 (10 nmol/rat, icv). Centrally pretreated SR95531 or SCH50911 significantly attenuated the GYY4137 (10 nmol/rat, icv)-induced prolongation of ICI, respectively. These findings suggest that endogenous brain H2S can inhibit the rat micturition reflex via both GABAA and GABAB receptors in the brain.


Subject(s)
Brain/metabolism , Hydrogen Sulfide/metabolism , Receptors, GABA/metabolism , Reflex/drug effects , Urination/drug effects , Aminooxyacetic Acid/pharmacology , Animals , Male , Morpholines/pharmacology , Muscle Contraction/physiology , Organothiophosphorus Compounds/pharmacology , Rats, Wistar , Urinary Bladder/physiology
11.
J Environ Pathol Toxicol Oncol ; 39(3): 281-290, 2020.
Article in English | MEDLINE | ID: mdl-32865918

ABSTRACT

Objective-To investigate cystathionine ß synthase (CBS)/hydrogen sulfide (H2S) signaling in multiple myeloma (MM) patients and to identify its effect on the proliferation of U266 cells. Methods-Bone marrow samples of 19 MM patients and 23 healthy donors were collected. qRT-PCR was performed to measure the mRNA expression levels of H2S synthases, cystathionine ß synthase, and cystathionine γ lyase. ELISA assays quantified the amount of H2S produced by the two enzymes CBS and CSE. CCK-8 experiment was used to investigate the influence of the CBS inhibitor amino oxyacetic acid and the CSE inhibitor propargylglycine on the proliferation of U266 cells. Flow cytometry and western blotting were performed to determine the effects of AOAA, PAG, and NaHS on cell cycle distribution as well as Caspase-3 and Bcl-2 expression. Results-Patients with MM had higher level of CBS compared with healthy donors. AOAA significantly inhibited cell proliferation in both a time and concentration dependent characteristic, whereas PAG does not. After 24 hours of treatment, AOAA significantly elevated the G0/G1 phase proportion of cells, and reduced the cell distribution in both S and G2/M phases, while NaHS accelerated cell cycle progression by reducing the relative number of cells in G0/G1 phase and increasing the proportion of cells in the G2/M phase. Moreover, AOAA abolished the impact of NaHS on cell cycle progression of U266 cells. AOAA treatment also led to a significant decrease in Bcl-2 expression and dramatic increase in Caspase-3 expression, though NaHS reversed these effects. Conclusion-CBS/H2S system might have a certain effect on the proliferation and apoptosis of MM cells.


Subject(s)
Apoptosis , Cell Proliferation , Cystathionine beta-Synthase/metabolism , Hydrogen Sulfide/metabolism , Multiple Myeloma/metabolism , Adult , Aged , Alkynes/pharmacology , Aminooxyacetic Acid/pharmacology , Apoptosis/drug effects , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Case-Control Studies , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cystathionine beta-Synthase/antagonists & inhibitors , Cystathionine gamma-Lyase/antagonists & inhibitors , Cystathionine gamma-Lyase/metabolism , Enzyme Inhibitors/pharmacology , Female , Glycine/analogs & derivatives , Glycine/pharmacology , Humans , Male , Middle Aged , Multiple Myeloma/pathology , Signal Transduction
12.
Life Sci ; 261: 118348, 2020 Nov 15.
Article in English | MEDLINE | ID: mdl-32860803

ABSTRACT

AIMS: 3,3'-Diindolylmethane (DIM) has limited anti-cancer effects in gastric cancer. Hydrogen sulfide (H2S) plays an important role in the tumor development and therapy, cystathionine-ß-synthase (CBS) and cystathionine-γ-lyase (CSE), two key endogenous H2S biosynthesis enzymes, can affect endogenous H2S levels and alter cancer treatment. Our main objective was to investigate whether the aminooxyacetic acid (AOAA) and DL-Propargylglycine (PAG), two specific inhibitors of CBS and CSE, could assist DIM to exert a stronger anti-cancer effects in gastric cancer BGC-823 and SGC-7901 cells. MATERIALS AND METHODS: Cell proliferation was assayed by MTT and cell colony-forming assay. Apoptosis and migration were detected by Hoechst staining and scratch test respectively. Western blot was used to evaluate the expression of proteins related to proliferation, apoptosis and migration. KEY FINDINGS: Combination of AOAA or PAG with DIM synergistically inhibited proliferation and migration, increased apoptosis in gastric cancer cells. The p38-p53 axis was also further activated by the combination of AOAA or PAG with DIM. Exogenous H2S from sodium hydrosulfide, attenuated the efficacy of DIM in cancer cells by reducing the activation level of p38-p53 axis. Taken together, AOAA or PAG inhibited the expression of endogenous H2S biosynthesis enzymes and effectively enhanced susceptibility of gastric cancer to DIM through activating p38-p53 axis. SIGNIFICANCE: The current study highlight more precise requirements for the clinical application of sulfur-containing anti-cancer drugs, and open a new way to enhance the sensitivity of DIM in chemotherapy of gastric cancer.


Subject(s)
Anticarcinogenic Agents/pharmacology , Hydrogen Sulfide/antagonists & inhibitors , Indoles/pharmacology , Stomach Neoplasms/drug therapy , Alkynes/administration & dosage , Alkynes/pharmacology , Aminooxyacetic Acid/administration & dosage , Aminooxyacetic Acid/pharmacology , Anticarcinogenic Agents/administration & dosage , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cystathionine beta-Synthase/metabolism , Cystathionine gamma-Lyase/metabolism , Drug Synergism , Glycine/administration & dosage , Glycine/analogs & derivatives , Glycine/pharmacology , Humans , Hydrogen Sulfide/metabolism , Indoles/administration & dosage , Stomach Neoplasms/pathology , Tumor Suppressor Protein p53/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
13.
Curr Mol Med ; 20(8): 624-632, 2020.
Article in English | MEDLINE | ID: mdl-32072910

ABSTRACT

PURPOSE: A small molecular compound, aminooxy-acetic acid (AOA), has been shown to modulate experimental autoimmune encephalomyelitis (EAE). The current study was designed to investigate whether AOA has a similar effect on the development of experimental autoimmune uveitis (EAU) and to further explore underlying mechanisms of this drug. METHODS: EAU was induced in C57BL/6J mice by immunization with interphotoreceptor retinoid-binding protein peptide 651-670 (IRBP 651-670). AOA (500µg or 750µg) or vehicle was administered by intraperitoneal injection from day 10 to 14 after EAU induction. The severity was assessed by clinical and histological scores. The integrity of the blood retinal barrier was detected with Evans Blue. Frequencies of splenic Th1, Th17 and Foxp3+ Treg cells were examined by flow cytometry. The production of cytokines was tested by ELISA. The mRNA expression of IL-17, IFN-γ and IL-10 was detected by RT-PCR. The expression of p-Stat1 and NF-κB was detected by Western Blotting. RESULTS: AOA was found to markedly inhibit the severity of EAU, as determined by clinical and histopathological examinations. AOA can relieve the leakage of blood retinal barrier (BRB). Functional studies found a decreased frequency of Th1 and Th17 cells and an increased frequency of Treg cells in EAU mice as compared with controls. Further studies showed that AOA not only downregulated the production of the proinflammatory cytokines including IFN-γ and IL-17 but also upregulated the expression of an anti-inflammatory cytokine such as IL-10, which might be caused by inhibiting the expressions of p-Stat1 and NF-κB. CONCLUSION: This study shows that AOA inhibits the severity and development of EAU by modulating the balance between regulatory and pathogenic lymphocyte subsets.


Subject(s)
Aminooxyacetic Acid/pharmacology , Autoimmune Diseases/prevention & control , GABA Agents/pharmacology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/immunology , Uveitis/prevention & control , Animals , Autoimmune Diseases/etiology , Autoimmune Diseases/metabolism , Autoimmune Diseases/pathology , Eye Proteins/immunology , Female , Mice , Mice, Inbred C57BL , NF-kappa B/genetics , NF-kappa B/metabolism , Retinol-Binding Proteins/immunology , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , Uveitis/etiology , Uveitis/metabolism , Uveitis/pathology
14.
Article in English | MEDLINE | ID: mdl-31907178

ABSTRACT

Toxoplasma gondii is an obligate intracellular protozoan parasite and a successful parasitic pathogen in diverse organisms and host cell types. Hydroxylamine (HYD) and carboxymethoxylamine (CAR) have been reported as inhibitors of aspartate aminotransferases (AATs) and interfere with the proliferation in Plasmodium falciparum Therefore, AATs are suggested as drug targets against Plasmodium The T. gondii genome encodes only one predicted AAT in both T. gondii type I strain RH and type II strain PLK. However, the effects of HYD and CAR, as well as their relationship with AAT, on T. gondii remain unclear. In this study, we found that HYD and CAR impaired the lytic cycle of T. gondiiin vitro, including the inhibition of invasion or reinvasion, intracellular replication, and egress. Importantly, HYD and CAR could control acute toxoplasmosis in vivo Further studies showed that HYD and CAR could inhibit the transamination activity of rTgAAT in vitro However, our results confirmed that deficiency of AAT in both RH and PLK did not reduce the virulence in mice, although the growth ability of the parasites was affected in vitro HYD and CAR could still inhibit the growth of AAT-deficient parasites. These findings indicated that HYD and CAR inhibition of T. gondii growth and control of toxoplasmosis can occur in an AAT-independent pathway. Overall, further studies focusing on the elucidation of the mechanism of inhibition are warranted. Our study hints at new substrates of HYD and CAR as potential drug targets to inhibit T. gondii growth.


Subject(s)
Aminooxyacetic Acid/pharmacology , Antiprotozoal Agents/pharmacology , Aspartate Aminotransferases/genetics , Hydroxylamine/pharmacology , Protozoan Proteins/genetics , Toxoplasma/drug effects , Toxoplasmosis/drug therapy , Animals , Aspartate Aminotransferases/deficiency , Cell Line , Chlorocebus aethiops , Female , Fibroblasts/drug effects , Fibroblasts/parasitology , Gene Expression , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Life Cycle Stages/drug effects , Life Cycle Stages/genetics , Mice , Mice, Inbred BALB C , Parasitic Sensitivity Tests , Protozoan Proteins/metabolism , Toxoplasma/genetics , Toxoplasma/growth & development , Toxoplasma/metabolism , Toxoplasmosis/parasitology , Vero Cells
15.
J Cell Mol Med ; 24(4): 2593-2609, 2020 02.
Article in English | MEDLINE | ID: mdl-31930778

ABSTRACT

Excessive activation of pro-inflammatory M1 macrophages following acute myocardial infarction (MI) aggravates adverse cardiac remodelling and heart dysfunction. There are two break points in the tricarboxylic acid cycle of M1 macrophages, and aspartate-arginosuccinate shunt compensates them. Aminooxyacetic acid (AOAA) is an inhibitor of aspartate aminotransferase in the aspartate-arginosuccinate shunt. Previous studies showed that manipulating macrophage metabolism may control macrophage polarization and inflammatory response. In this study, we aimed to clarify the effects of AOAA on macrophage metabolism and polarization and heart function after MI. In vitro, AOAA inhibited lactic acid and glycolysis and enhanced ATP levels in classically activated M1 macrophages. Besides, AOAA restrained pro-inflammatory M1 macrophages and promoted anti-inflammatory M2 phenotype. In vivo, MI mice were treated with AOAA or saline for three consecutive days. Remarkably, AOAA administration effectively inhibited the proportion of M1 macrophages and boosted M2-like phenotype, which subsequently attenuated infarct size as well as improved post-MI cardiac function. Additionally, AOAA attenuated NLRP3-Caspase1/IL-1ß activation and decreased the release of IL-6 and TNF-α pro-inflammatory cytokines and reciprocally increased IL-10 anti-inflammatory cytokine level in both ischaemic myocardium and M1 macrophages. In conclusion, short-term AOAA treatment significantly improves cardiac function in mice with MI by balancing macrophage polarization through modulating macrophage metabolism and inhibiting NLRP3-Caspase1/IL-1ß pathway.


Subject(s)
Aminooxyacetic Acid/pharmacology , Heart Diseases/drug therapy , Heart/drug effects , Macrophages/drug effects , Macrophages/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Caspase 1/metabolism , Heart Diseases/metabolism , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Male , Mice , Mice, Inbred C57BL , Myocardial Infarction/drug therapy , Myocardial Infarction/metabolism , Myocardium/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Tumor Necrosis Factor-alpha/metabolism , Ventricular Remodeling/drug effects
16.
Arch Biochem Biophys ; 680: 108237, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31881188

ABSTRACT

BACKGROUND AND PURPOSE: Several members of the core clock mechanism are equipped with a Per-Arnt-Sim (PAS) domain through which they can bind haem [Fe(II)]. Haem is a ligand for the orphan receptors REV-ERBα/ß (NR1D1/2), which regulate circadian rhythm and metabolism. The ability to bind haem sensitises these clock components to the action of small molecule gases, including NO, CO and H2S. Studies conducted with European hamsters revealed that during winter sleep, key clock genes stop oscillating. At the same time, H2S, when administered at subtoxic concentrations, can induce a hypometabolic state in the cell. We suppose that core clock components, including the nuclear receptors REV-ERBs, neuronal PAS domain protein 2 (nPAS2) and PER2, can be H2S targets. The general objective of this study was to investigate the effect of the H2S system on the expression profile of the core clock genes in cells in vitro. EXPERIMENTAL APPROACH: We analysed the expression of Per1, Per2, Per3, Bmal1, Cry1, Cry2, Nr1d1, Nfil-3 and Dbp messenger RNA (mRNA) in serum-shocked NIH-3T3 cells treated with a slow-releasing H2S donor (GYY4137) or the cystathionine beta-synthase (CBS) inhibitor (AOAA) cultured under constant darkness and collected during 3 days in 3 h interval. KEY RESULTS AND CONCLUSIONS AND IMPLICATIONS: We found that pharmacological CBS inhibition increased the general expression and dynamics of several clock genes. On the other hand, increased H2S decreased Per2 expression. These data suggest that CBS can affect circadian clock and effect on clock-controlled transcription output.


Subject(s)
Aminooxyacetic Acid/pharmacology , Circadian Clocks/drug effects , Circadian Rhythm Signaling Peptides and Proteins/genetics , Morpholines/pharmacology , Organothiophosphorus Compounds/pharmacology , Animals , Cell Cycle/drug effects , Gene Expression Regulation/drug effects , Mice , NIH 3T3 Cells
17.
Carcinogenesis ; 41(5): 656-665, 2020 07 10.
Article in English | MEDLINE | ID: mdl-31276576

ABSTRACT

Metabolic changes accompanying a step-wise malignant transformation was investigated using a syngeneic lineage of human fibroblasts. Cell immortalization was associated with minor alterations in metabolism. Consecutive loss of cell cycle inhibition in immortalized cells resulted in increased levels of oxidative phosphorylation (OXPHOS). Overexpression of the H-Ras oncoprotein produced cells forming sarcomas in athymic mice. These transformed cells exhibited increased glucose consumption, glycolysis and a further increase in OXPHOS. Because of the markedly increased OXPHOS in transformed cells, the impact of a transaminase inhibitor, aminooxyacetic acid (AOA), which decreases glutamine influx to the tricarboxylic acid (TCA) cycle, was tested. Indeed, AOA significantly decreased proliferation of malignantly transformed fibroblasts and fibrosarcoma-derived cells in vitro and in vivo. AOA also decreased proliferation of cells susceptible to malignant transformation. Metabolomic studies in normal and transformed cells indicated that, in addition to the anticipated effect on the TCA cycle, AOA decreased production of nucleotides adenosine triphosphate (ATP) and uridine monophosphate. Exogenous nucleotides partially rescued decreased proliferation of the malignant cells treated with AOA. Our data indicate that AOA blocks several metabolic pathways essential for growth of malignant cells. Therefore, OXPHOS may provide important therapeutic targets for treatment of sarcoma.


Subject(s)
Cell Transformation, Neoplastic/pathology , Fibroblasts/pathology , Fibrosarcoma/pathology , Gene Expression Regulation, Neoplastic , Genomics/methods , Metabolome/drug effects , Skin/pathology , Aminooxyacetic Acid/pharmacology , Animals , Apoptosis , Cell Proliferation , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/metabolism , Enzyme Inhibitors/pharmacology , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibrosarcoma/drug therapy , Fibrosarcoma/genetics , Fibrosarcoma/metabolism , Glycolysis , Humans , Mice , Mice, Nude , Oxidative Phosphorylation , Skin/drug effects , Skin/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
18.
Protoplasma ; 257(1): 213-227, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31410589

ABSTRACT

Self-incompatibility (SI) is genetically determined reproductive barrier preventing inbreeding and thereby providing the maintenance of plant species diversity. At present, active studies of molecular bases of SI mechanisms are underway. S-RNAse-based SI in Petunia hybrida L. is a self-/non-self recognition system that allows the pistil to reject self pollen and to accept non-self pollen for outcrossing. In the present work, using fluorescent methods including the TUNEL method allowed us to reveal the presence of markers of programmed cell death (PCD), such as DNA fragmentation, in growing in vivo petunia pollen tubes during the passage of the SI reaction. The results of statistical analysis reliably proved that PCD is the factor of S-RNAse-based SI. It was found that preliminary treatment before self-pollination of stigmas of petunia self-incompatible line with aminooxyacetic acid (AOA), inhibitor of ACC synthesis, led to stimulation of pollen tubes growth when the latter did not exhibit any hallmarks of PCD. These data argue in favor of assumption that ethylene controls the passage of PCD in incompatible pollen tubes in the course of S-RNAse-based SI functioning. The involvement of the hormonal regulation in SI mechanism in P. hybrida L. is the finding observed by us for the first time.


Subject(s)
Amino Acids, Cyclic/biosynthesis , Aminooxyacetic Acid/pharmacology , Apoptosis/drug effects , Petunia/cytology , Petunia/physiology , Pollen Tube/cytology , Self-Incompatibility in Flowering Plants/drug effects , DNA Fragmentation/drug effects , Petunia/drug effects , Petunia/ultrastructure , Pollen Tube/drug effects , Pollen Tube/ultrastructure , Ribonucleases/metabolism
19.
Proc Natl Acad Sci U S A ; 116(38): 18769-18771, 2019 09 17.
Article in English | MEDLINE | ID: mdl-31481613

ABSTRACT

Down syndrome (DS) is associated with significant perturbances in mitochondrial function. Here we tested the hypothesis that the suppression of mitochondrial electron transport in DS cells is due to high expression of cystathionine-ß-synthase (CBS) and subsequent overproduction of the gaseous transmitter hydrogen sulfide (H2S). Fibroblasts from DS individuals showed higher CBS expression than control cells; CBS localization was both cytosolic and mitochondrial. DS cells produced significantly more H2S and polysulfide and exhibited a profound suppression of mitochondrial electron transport, oxygen consumption, and ATP generation. DS cells also exhibited slower proliferation rates. In DS cells, pharmacological inhibition of CBS activity with aminooxyacetate or siRNA-mediated silencing of CBS normalized cellular H2S levels, restored Complex IV activity, improved mitochondrial electron transport and ATP synthesis, and restored cell proliferation. Thus, CBS-derived H2S is responsible for the suppression of mitochondrial function in DS cells. When H2S overproduction is corrected, the tonic suppression of Complex IV is lifted, and mitochondrial electron transport is restored. CBS inhibition offers a potential approach for the pharmacological correction of DS-associated mitochondrial dysfunction.


Subject(s)
Cystathionine beta-Synthase/metabolism , Down Syndrome/metabolism , Electron Transport Complex IV/metabolism , Hydrogen Sulfide/metabolism , Mitochondria/metabolism , Aminooxyacetic Acid/pharmacology , Cell Proliferation , Cells, Cultured , Cystathionine beta-Synthase/antagonists & inhibitors , Cystathionine beta-Synthase/genetics , Down Syndrome/pathology , Electron Transport Complex IV/genetics , Energy Metabolism , Female , Fibroblasts/metabolism , Gene Expression , Humans , Mitochondria/enzymology , Oxidative Phosphorylation , Oxygen Consumption , RNA, Small Interfering/genetics , Sulfides/metabolism
20.
Int J Mol Sci ; 20(10)2019 May 24.
Article in English | MEDLINE | ID: mdl-31137614

ABSTRACT

A number of factors can trigger amyotrophic lateral sclerosis (ALS), although its precise pathogenesis is still uncertain. In a previous study done by us, poisonous liquoral levels of hydrogen sulphide (H2S) in sporadic ALS patients were reported. In the same study very high concentrations of H2S in the cerebral tissues of the familial ALS (fALS) model of the SOD1G93A mouse, were measured. The objective of this study was to test whether decreasing the levels of H2S in the fALS mouse could be beneficial. Amino-oxyacetic acid (AOA)-a systemic dual inhibitor of cystathionine-ß-synthase and cystathionine-γ lyase (two key enzymes in the production of H2S)-was administered to fALS mice. AOA treatment decreased the content of H2S in the cerebral tissues, and the lifespan of female mice increased by approximately ten days, while disease progression in male mice was not affected. The histological evaluation of the spinal cord of the females revealed a significant increase in GFAP positivity and a significant decrease in IBA1 positivity. In conclusion, the results of the study indicate that, in the animal model, the inhibition of H2S production is more effective in females. The findings reinforce the need to adequately consider sex as a relevant factor in ALS.


Subject(s)
Aminooxyacetic Acid/pharmacology , Amyotrophic Lateral Sclerosis/metabolism , Cystathionine beta-Synthase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Hydrogen Sulfide/metabolism , Aminooxyacetic Acid/therapeutic use , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/genetics , Animals , Brain/drug effects , Brain/metabolism , Cells, Cultured , Enzyme Inhibitors/therapeutic use , Female , Male , Mice , Mice, Inbred C57BL , Neuroglia/drug effects , Sex Factors , Superoxide Dismutase-1/genetics
SELECTION OF CITATIONS
SEARCH DETAIL