Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 45
1.
Am J Physiol Regul Integr Comp Physiol ; 326(5): R427-R437, 2024 May 01.
Article En | MEDLINE | ID: mdl-38497128

Methylglyoxal (MG), a reactive metabolic byproduct of glycolysis, is a causative of painful diabetic neuropathy. Patients with diabetes are associated with more frequent severe asthma exacerbation. Stimulation of capsaicin-sensitive lung vagal (CSLV) afferents may contribute to the pathogenesis of hyperreactive airway diseases such as asthma. However, the possibility of the stimulatory effect of MG on CSLV afferents and the underlying mechanisms remain unknown. Our results showed that intravenous injection of MG (25 mg/kg, MG25) in anesthetized, spontaneously breathing rats elicited pulmonary chemoreflexes characterized by apnea, bradycardia, and hypotension. The MG-induced apneic response was reproducible and dose dependent. MG25 no longer evoked these reflex responses after perineural capsaicin treatment of both cervical vagi to block C-fibers' conduction, suggesting that the reflexes were mediated through the stimulation of CSLV afferents. Pretreatment with HC030031 [an antagonist of transient receptor potential ankyrin subtype 1 protein (TRPA1)] or AP18 (another TRPA1 antagonist), but not their vehicle, markedly attenuated the apneic response induced by MG25. Consistently, electrophysiological results showed that pretreatment with HC030031 largely attenuated the intense discharge in CSLV afferents induced by injection of MG25 in open-chest and artificially ventilated rats. In isolated CSLV neurons, the perfusion of MG evoked an abrupt and pronounced increase in calcium transients in a concentration-dependent manner. This stimulatory effect on CSLV neurons was also abolished by HC030031 treatment but not by its vehicle. In conclusion, these results suggest that MG exerts a stimulatory effect on CSLV afferents, inducing pulmonary chemoreflexes, and such stimulation is mediated through the TRPA1 activation.NEW & NOTEWORTHY Methylglyoxal (MG) is implicated in the development of painful diabetic neuropathy. A retrospective cohort study revealed an increased incidence of asthma exacerbations in patients with diabetes. This study demonstrated that elevated circulating MG levels stimulate capsaicin-sensitive lung vagal afferents via activation of TRPA1, which in turn triggers respiratory reflexes. These findings provide new information for understanding the pathogenic mechanism of diabetes-associated hyperreactive airway diseases and potential therapy.


Acetanilides , Asthma , Diabetic Neuropathies , Purines , Humans , Rats , Animals , Capsaicin/pharmacology , Rats, Sprague-Dawley , Pyruvaldehyde/adverse effects , Pyruvaldehyde/metabolism , Diabetic Neuropathies/metabolism , Retrospective Studies , Lung , Vagus Nerve/physiology , Apnea , Asthma/metabolism , TRPA1 Cation Channel/metabolism
2.
Front Endocrinol (Lausanne) ; 13: 910868, 2022.
Article En | MEDLINE | ID: mdl-35872977

Methylglyoxal, a major precursor of advanced glycation end products, is elevated in the plasma of patients with type 2 diabetes mellitus. Islet ß-cell function was recently shown to be regulated by N6-methyladenosine (m6A), an RNA modification consisting of methylation at the N6 position of adenosine. However, the role of m6A methylation modification in methylglyoxal-induced impairment of insulin secretion in pancreatic ß cells has not been clarified. In this study, we showed that treatment of two ß-cell lines, NIT-1 and ß-TC-6, with methylglyoxal reduced m6A RNA content and methyltransferase-like 3 (METTL3) expression levels. We also showed that silencing of METTL3 inhibited glucose-stimulated insulin secretion (GSIS) from NIT-1 cells, whereas upregulation of METTL3 significantly reversed the methylglyoxal-induced decrease in GSIS. The methylglyoxal-induced decreases in m6A RNA levels and METTL3 expression were not altered by knockdown of the receptor for the advanced glycation end product but were further decreased by silencing of glyoxalase 1. Mechanistic investigations revealed that silencing of METTL3 reduced m6A levels, mRNA stability, and the mRNA and protein expression levels of musculoaponeurotic fibrosarcoma oncogene family A (MafA). Overexpression of MafA greatly improved the decrease in GSIS induced by METTL3 silencing; silencing of MafA blocked the reversal of the MG-induced decrease in GSIS caused by METTL3 overexpression. The current study demonstrated that METTL3 ameliorates MG-induced impairment of insulin secretion in pancreatic ß cells by regulating MafA.


Diabetes Mellitus, Type 2 , Insulin Secretion , Insulin-Secreting Cells , Maf Transcription Factors, Large , Methyltransferases , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Glucose/metabolism , Humans , Insulin Secretion/drug effects , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Maf Transcription Factors, Large/genetics , Methyltransferases/genetics , Methyltransferases/metabolism , Pyruvaldehyde/adverse effects , RNA, Messenger/genetics
3.
Mol Nutr Food Res ; 65(13): e2000799, 2021 07.
Article En | MEDLINE | ID: mdl-33890707

INTRODUCTION: Advanced glycation end products (AGEs) and receptor of advanced glycation end products (RAGE) mediate renal function during diabetic and non-diabetic nephropathy development. Methylglyoxal-lysine dimer (MOLD), a typical toxic advanced glycation end product (TAGE), contributes to inflammatory responses during renal diseases. This study determines the effect of MOLD on inflammatory responses in mouse mesangial cells. METHODS AND RESULTS: The murine mesangial cell line SV40 MES 13 is used to assess nuclear factor-kappa B (NF-κB) expression, reactive oxygen species (ROS) production, and mitochondria labeling. The interaction model between RAGE and MOLD is also determined. MOLD treatment of mesangial cells markedly increases RAGE expression and the linkage with V-type Ig domain of RAGE. MOLD induces ROS production and mitochondrial dysfunction. MOLD activates phosphatidylinositol 3-kinase-protein kinase B (PI3KB) and NF-κB signaling pathways. It is confirmed that these changes are reversed when ROS is suppressed. These effects may be regulated through mitogen-activated protein kinases and pro-inflammatory cytokines in circulatory inflammation responses. CONCLUSION: MOLD plays a major role in nephropathy via ROS production and mitochondrial dysfunction through direct association with RAGE. Further, the NF-kB and PI3K/AKT signaling pathways triggered by ROS mediate the inflammatory response to exacerbate MOLD-induced damages in inflammation-related diabetic and non-diabetic renal diseases.


Glycation End Products, Advanced/adverse effects , Inflammation/chemically induced , Mesangial Cells , Animals , Cell Line , Lysine/adverse effects , Mesangial Cells/drug effects , Mesangial Cells/metabolism , Mice , Mitochondria/drug effects , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Pyruvaldehyde/adverse effects , Reactive Oxygen Species/metabolism , Signal Transduction
4.
Mol Neurobiol ; 58(2): 735-749, 2021 Feb.
Article En | MEDLINE | ID: mdl-33011857

Methylglyoxal (MGO) is an endogenous toxin, mainly produced as a by-product of glycolysis that has been associated to aging, Alzheimer's disease, and inflammation. Cell culture studies reported that MGO could impair the glyoxalase, thioredoxin, and glutathione systems. Thus, we investigated the effect of in vivo MGO administration on these systems, but no major changes were observed in the glyoxalase, thioredoxin, and glutathione systems, as evaluated in the prefrontal cortex and the hippocampus of mice. A previous study from our group indicated that MGO administration produced learning/memory deficits and depression-like behavior. Confirming these findings, the tail suspension test indicated that MGO treatment for 7 days leads to depression-like behavior in three different mice strains. MGO treatment for 12 days induced working memory impairment, as evaluated in the Y maze spontaneous alternation test, which was paralleled by low dopamine and serotonin levels in the cerebral cortex. Increased DARPP32 Thr75/Thr34 phosphorylation ratio was observed, suggesting a suppression of phosphatase 1 inhibition, which may be involved in behavioral responses to MGO. Co-treatment with a dopamine/noradrenaline reuptake inhibitor (bupropion, 10 mg/kg, p.o.) reversed the depression-like behavior and working memory impairment and restored the serotonin and dopamine levels in the cerebral cortex. Overall, the cerebral cortex monoaminergic system appears to be a preferential target of MGO toxicity, a new potential therapeutic target that remains to be addressed.


Depression/physiopathology , Dopamine Uptake Inhibitors/pharmacology , Dopamine/deficiency , Memory, Short-Term , Norepinephrine/metabolism , Pyruvaldehyde/adverse effects , Animals , Bupropion/pharmacology , Dopamine/metabolism , Female , Glutathione/metabolism , Immobilization , Memory, Short-Term/drug effects , Mice, Inbred BALB C , Mice, Inbred C57BL , Motor Activity/drug effects , Phosphorylation/drug effects , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Pyruvaldehyde/administration & dosage , Serotonin/metabolism , Tyrosine 3-Monooxygenase/metabolism
5.
Food Funct ; 11(6): 5486-5497, 2020 Jun 24.
Article En | MEDLINE | ID: mdl-32500907

Non-enzymatic reactions between proteins and methylglyoxal (MG) result in the formation of advanced glycation end products (AGEs). These AGEs play a vital role in the development of diabetic complications by stimulating oxidative stress and acting upon their receptor RAGE (Receptor for Advanced Glycation End products). This study examined the effect of aqueous methanol extract of Bombax ceiba L. calyxes (BCCE) on MG induced protein glycation and oxidative stress, followed by the identification of phytometabolites present in the calyxes using gas chromatography-mass spectrometry (GC-MS). The study revealed that priming of bovine serum albumin protein with the BCCE inhibited MG induced AGE formation in vitro and restrained AGE-induced RAGE up-regulation in HEK-293 cells. The BCCE significantly (p < 0.001) reduced the MG induced increase in reactive oxygen species (ROS), NADPH oxidase (NOX), and mitochondrial dysfunction. Improvements in the levels of antioxidant enzymes such as Mn and Cu/Zn-superoxide dismutase and glutathione reductase were also observed in HEK-293 cells. Furthermore, the decrease in primary cellular defense against AGEs, the glyoxalase 1 (Glo-1) activity, due to MG treatment was restored in BCCE treated cells. GC-MS analysis revealed the presence of antioxidant and antiglycation compounds such as myo-ionisitol, scopoletin, d-sedoheptulose, succinic acid, and xylitol in B. ceiba calyxes. The observed beneficial effect in our study might be attributed to the presence of these compounds in B. Ceiba calyxes. This is the first report presenting the antioxidant and antiglycation activities of B. ceiba calyxes and GC-MS analysis of active phytometabolites. These observations show that B. ceiba calyxes may become a potent and promising functional food to manage/control the development of diabetic complications.


Bombax/chemistry , Oxidative Stress/drug effects , Plant Extracts/pharmacology , Pyruvaldehyde/adverse effects , Antioxidants/pharmacology , Cell Survival/drug effects , Diabetes Complications , Gas Chromatography-Mass Spectrometry , Glycation End Products, Advanced , Glycosylation , HEK293 Cells , Humans , India , Lactoylglutathione Lyase/metabolism , Membrane Potential, Mitochondrial/drug effects , NADPH Oxidases/metabolism , Reactive Oxygen Species/metabolism , Receptor for Advanced Glycation End Products/metabolism , Serum Albumin, Bovine
6.
J Agric Food Chem ; 68(28): 7416-7424, 2020 Jul 15.
Article En | MEDLINE | ID: mdl-32573222

Our previous study has found that dietary genistein could ameliorate high-fat diet (HFD)-induced obesity and especially lower methylglyoxal (MGO) and advanced glycation end product (AGE) accumulation in healthy mice exposed to genistein and HFD. However, it is still unclear whether dietary genistein intervention has a similar beneficial effect in obese mice. In this study, the mice were induced with obesity after being fed a HFD for nine weeks before being administered with two doses of genistein, 0.1% (G 0.1) and 0.2% (G 0.2), in the HFD for additional 19 weeks. After 19 week treatment, genistein supplementation reduced body and liver weights, plasma and liver MGO levels, and kidney AGE levels in mice. Mechanistically, genistein upregulated the expressions of glyoxalase I and II and aldose reductase to detoxify MGO, and genistein and its microbial metabolites, dihydrogenistein and 6'-hydroxy-O-demethylangolensin, were able to trap endogenous MGO via formation of MGO conjugates. Taken together, our results provide novel insights into the antiobesity and antiglycation roles of dietary genistein in obese subjects.


Genistein/metabolism , Glycation End Products, Advanced/metabolism , Obesity/diet therapy , Pyruvaldehyde/metabolism , Aldehyde Reductase/metabolism , Animals , Diet, High-Fat/adverse effects , Humans , Lactoylglutathione Lyase/metabolism , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/etiology , Obesity/metabolism , Plant Extracts/metabolism , Pyruvaldehyde/adverse effects
7.
Nutrients ; 12(5)2020 May 06.
Article En | MEDLINE | ID: mdl-32384625

Recently, the mechanisms responsible for anti-glycation activity of cyanidin and its derivatives on the inhibition of methylglyoxal (MG)-induced protein glycation and advanced glycation-end products (AGEs) as well as oxidative DNA damage were reported. In this study, we investigated the protective effect of cyanidin against MG-induced oxidative stress and apoptosis in rat INS-1 pancreatic ß-cells. Exposure of cells to cytotoxic levels of MG (500 µM) for 12 h caused a significant reduction in cell viability. However, the pretreatment of cells with cyanidin alone (6.25-100 µM) for 12 h, or cotreatment of cells with cyanidin (3.13-100 µM) and MG, protected against cell cytotoxicity. In the cotreatment condition, cyanidin (33.3 and 100 µM) also decreased MG-induced apoptosis as determined by caspase-3 activity. Furthermore, INS-1 cells treated with MG increased the generation of reactive oxygen species (ROS) during a 6 h exposure. The MG-induced increase in ROS production was inhibited by cyanidin (33.3 and 100 µM) after 3 h stimulation. Furthermore, MG diminished the activity of glyoxalase 1 (Glo-1) and its gene expression as well as the level of total glutathione. In contrast, cyanidin reversed the inhibitory effect of MG on Glo-1 activity and glutathione levels. Interestingly, cyanidin alone was capable of increasing Glo-1 activity and glutathione levels without affecting Glo-1 mRNA expression. These findings suggest that cyanidin exerts a protective effect against MG-induced oxidative stress and apoptosis in pancreatic ß-cells by increasing the activity of Glo-1.


Anthocyanins/pharmacology , Apoptosis/drug effects , Gene Expression/drug effects , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/physiology , Lactoylglutathione Lyase/genetics , Lactoylglutathione Lyase/metabolism , Oxidative Stress/drug effects , Pyruvaldehyde/adverse effects , Animals , Caspase 3/metabolism , Cells, Cultured , DNA Damage/drug effects , Glutathione/metabolism , Glycation End Products, Advanced/antagonists & inhibitors , Glycation End Products, Advanced/metabolism , Glycosylation/drug effects , Rats , Reactive Oxygen Species/metabolism
8.
Biochem Biophys Res Commun ; 525(3): 767-772, 2020 05 07.
Article En | MEDLINE | ID: mdl-32147098

The accumulation of glucose degradation products (GDPs) can lead to tissue damage in patients with diabetes and those undergoing long-term peritoneal dialysis (PD). Angiogenesis is occasionally observed in the peritoneal membrane of patients undergoing PD, where it is associated with failure of ultrafiltration. To investigate the mechanism underlying the influence of angiogenesis on fluid absorption, we evaluated the effects of accumulation of the glucose degradation product methylglyoxal (MGO) on angiogenesis in vitro, and analyzed the association with angiogenesis in the peritoneal membrane. To this end, we measured the levels of vascular endothelial growth factor (VEGF) and platelet-derived growth factor (PDGF)-BB in cultured endothelial and smooth muscle cells after administration of MGO. The expression of PDGF-BB mRNA and protein decreased significantly after exposure to MGO, while the expression of VEGF mRNA increased (both P < 0.01). The expression of PDGF-Rß mRNA in cultured smooth muscle cells did not change after administration of MGO, although the expression of VEGF mRNA increased (P < 0.01). We also evaluated the associations between the number of capillary vessels, peritoneal function, and the degree of MGO deposition using peritoneum samples collected from patients undergoing PD. The number of immature capillary vessels was significantly associated with peritoneal dysfunction and the degree of MGO accumulation (both P < 0.01). In conclusion, MGO enhances the production of VEGF and suppresses the production of PDGF-BB, potentially leading to disturbance of angiogenesis in the peritoneal membrane. Accumulation of MGO in the peritoneum may cause immature angiogenesis and peritoneal dysfunction.


Glucose/metabolism , Neovascularization, Pathologic/chemically induced , Peritoneal Dialysis/adverse effects , Pyruvaldehyde/adverse effects , Angiogenesis Inducing Agents/metabolism , Becaplermin/metabolism , Female , Glycation End Products, Advanced/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Male , Middle Aged , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Neovascularization, Pathologic/pathology , Peritoneum/pathology , Vascular Endothelial Growth Factor A/metabolism
9.
Theranostics ; 9(15): 4287-4307, 2019.
Article En | MEDLINE | ID: mdl-31285762

Methylglyoxal (MGO), an endogenous reactive carbonyl compound, plays a key role in the pathogenesis of diabetic neuropathy. The aim of this study is to investigate the role of MGO in diabetic itch and hypoalgesia, two common symptoms associated with diabetic neuropathy. Methods: Scratching behavior, mechanical itch (alloknesis), and thermal hypoalgesia were quantified after intradermal (i.d.) injection of MGO in naïve mice or in diabetic mice induced by intraperitoneal (i.p.) injection of streptozotocin (STZ). Behavioral testing, patch-clamp recording, transgenic mice, and gene expression analysis were used to investigate the mechanisms underlying diabetic itch and hypoalgesia in mice. Results: I.d. injection of MGO evoked dose-dependent scratching in normal mice. Addition of MGO directly activated transient receptor potential ankyrin 1 (TRPA1) to induce inward currents and calcium influx in dorsal root ganglia (DRG) neurons or in TRPA1-expressing HEK293 cells. Mechanical itch, but not spontaneous itch was developed in STZ-induced diabetic mice. Genetic ablation of Trpa1 (Trpa1-/- ), pharmacological blockade of TRPA1 and Nav1.7, antioxidants, and mitogen-activated protein kinase kinase enzyme (MEK) inhibitor U0126 abrogated itch induced by MGO or in STZ-induced diabetic mice. Thermal hypoalgesia was induced by intrathecal (i.t.) injection of MGO or in STZ-induced diabetic mice, which was abolished by MGO scavengers, intrathecal injection of TRPA1 blockers, and in Trpa1-/- mice. Conclusion: This study revealed that Nav1.7 and MGO-mediated activation of TRPA1 play key roles in itch and hypoalgesia in a murine model of type 1 diabetes. Thereby, we provide a novel potential therapeutic strategy for the treatment of itch and hypoalgesia induced by diabetic neuropathy.


Diabetes Mellitus, Type 1/complications , Neuralgia/metabolism , Pruritus/metabolism , TRPA1 Cation Channel/metabolism , Animals , Diabetic Neuropathies/etiology , Diabetic Neuropathies/genetics , Diabetic Neuropathies/metabolism , Disease Models, Animal , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Knockout , NAV1.7 Voltage-Gated Sodium Channel/genetics , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Neuralgia/etiology , Neuralgia/genetics , Pruritus/etiology , Pruritus/genetics , Pyruvaldehyde/adverse effects , TRPA1 Cation Channel/genetics
10.
J Food Biochem ; 43(7): e12907, 2019 07.
Article En | MEDLINE | ID: mdl-31353699

The protective effect and mechanism(s) of action of cinnamaldehyde on the highly reactive secondary sugar derivative, methylglyoxal, induced vascular damage were investigated using isolated rat thoracic aorta. Aorta was incubated with methylglyoxal and cinnamaldehyde where vascular reactivity was assessed through phenylephrine- and acetylcholine-induced contraction and relaxation, respectively. Cinnamaldehyde's antioxidant activity, ability to induce aortic nitric oxide release, and effect on advanced glycation end products formation (AGEs) was also studied. Results showed that cinnamaldehyde significantly alleviated the exaggerated contraction and improved the attenuated dilation of the aorta secondary to incubation with methylglyoxal. Furthermore, cinnamaldehyde stimulated aortic nitric oxide production from isolated aorta giving levels similar to acetylcholine and significantly reduced both methylglyoxal-induced AGEs and protein oxidation products formation. In conclusion, cinnamaldehyde protects from methyglyoxal-induced vascular damage mainly by improving the vasodilation in addition to endothelial nitric oxide production and reducing the detrimental AGE-inflicted vascular damage. PRACTICAL APPLICATIONS: The use of naturally occurring products to alleviate various disease-related complications is highly attractive due to their easy availability, relatively affordable prices compared to pharmaceutical products, and their favorable safety profile. In the case of cinnamaldehyde, its excessive and highly reputable consumption in the food industry facilitates promoting a daily intake of the natural compound with the purpose of counteracting the destructive effect that elevated blood glucose has on vascular function. According to findings obtained from this study, frequent cinnamaldehyde intake will improve vascular reactivity by acting on vasodilatory mechanisms and inhibiting glycation reactions, hence improving the hyperglycemia associated hypertensive state. The study also paves the way for future research to determine the clinical efficacy of cinnamaldehyde having established its competence in protecting vascular function in a lab setting.


Acrolein/analogs & derivatives , Nitric Oxide/metabolism , Protective Agents/pharmacology , Pyruvaldehyde/adverse effects , Acrolein/pharmacology , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/physiology , Male , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Vasoconstriction/drug effects , Vasodilation/drug effects
11.
Chem Biol Interact ; 310: 108728, 2019 Sep 01.
Article En | MEDLINE | ID: mdl-31254498

Disruption of the mitochondrial function has been associated with redox impairment and triggering of cell death in nucleated human cells, as observed in several diseases. The administration of chemicals that would prevent mitochondrial dysfunction is an attractive strategy in cases of neurodegeneration, cardiovascular diseases, and metabolic disorders. Methylglyoxal (MG) is a dicarbonyl compound that exhibits an important role as a mitochondrial toxicant in neurodegenerative diseases (such as Alzheimer's disease and Parkinson's disease) and diabetes mellitus. On the other hand, naringenin (NGN; C15H12O5) is a natural antioxidant that also presents anti-inflammatory effects in mammalian cells. In this context, we have evaluated whether and how NGN would be able to prevent the mitochondria-related bioenergetics and redox dysfunctions induced by MG in the human neuroblastoma SH-SY5Y cells. The cells were pretreated (for 2 h) with NGN (at 10-80 µM) and then challenged with MG at 500 µM for 24 h. NGN significantly attenuated the effects of MG on the mitochondrial function and redox environment in this experimental model. Moreover, NGN prevented the MG-triggered mitochondria-related cell death in SH-SY5Y cells. Nonetheless, the inhibition of the synthesis of glutathione (GSH, a major non-enzymatic antioxidant) suppressed the promotion of mitochondrial protection by NGN in MG-treated cells. We also found that the synthesis of GSH was induced by NGN through a mechanism associated with the transcription factor nuclear factor (erythroid-derived 2)-like 2 (Nrf2). Therefore, NGN caused mitochondrial protection by an Nrf2/GSH-dependent manner in SH-SY5Y cells exposed to MG.


Flavanones/pharmacology , Glutathione/metabolism , Mitochondria/drug effects , NF-E2-Related Factor 2/metabolism , Neuroblastoma/pathology , Neuroprotective Agents/pharmacology , Antioxidants/pharmacology , Antioxidants/therapeutic use , Cell Death/drug effects , Cell Line, Tumor , Flavanones/therapeutic use , Humans , Oxidation-Reduction/drug effects , Pyruvaldehyde/adverse effects
12.
Chem Biol Interact ; 308: 147-154, 2019 Aug 01.
Article En | MEDLINE | ID: mdl-31103701

Diabetes mellitus is characterized by chronic hyperglycemia and its diverse complications. Hyperglycemia is associated with inflammatory responses in different organs, and diabetic patients have a higher risk of bone fracture due to increased bone weakness. Methylglyoxal, a reactive advanced glycation end product precursor, is known to have increased level in diabetic patients. The accumulation of methylglyoxal promotes inflammation and it may play a role in diabetes related osteoporosis. In this study, therefore, the underlying mechanism of methylglyoxal on osteoporosis was studied using both animal and cell models. In the animal model, rats were treated with either methylglyoxal or saline as control. In the cell model, the macrophage RAW264.7 was treated with methylglyoxal or vehicle control. Following the treatment, animal samples were harvested for micro-CT and real-time polymerase chain reaction analyses. Cell samples were harvested for MTT assay, RT-PCR, and Western Blotting analyses. In both animals and cell cultures, methylglyoxal was shown to induce osteoclastogenesis by increased gene expression of osteoclast bone biomarkers CTSK, OSCAR and TRACP5. Furthermore, in methylglyoxal-treated macrophages activation of the c-Jun N-terminal kinases signaling pathway was observed, and inhibition of JNK activities resulted in down-regulation of osteoclast biomarkers gene expressions. Our results therefore suggested that methylglyoxal may contribute to the progression of diabetes-related osteoporosis and imbalanced bone remodeling through JNK pathway in osteoclasts.


MAP Kinase Signaling System/drug effects , Pyruvaldehyde/pharmacology , Animals , Bone and Bones/diagnostic imaging , Cell Differentiation/drug effects , Cell Survival/drug effects , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Mice , Osteoclasts/cytology , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteogenesis/drug effects , Osteoporosis/etiology , Osteoporosis/pathology , Protein-Tyrosine Kinases/metabolism , Pyruvaldehyde/adverse effects , RAW 264.7 Cells , Rats , Rats, Sprague-Dawley , Tartrate-Resistant Acid Phosphatase/metabolism , X-Ray Microtomography
13.
Eur Rev Med Pharmacol Sci ; 23(6): 2669-2680, 2019 Mar.
Article En | MEDLINE | ID: mdl-30964194

OBJECTIVE: Endothelial dysfunction (ED) predisposes to venous thrombosis (VT) and post-thrombotic syndrome (PTS), a long-term VT-related complication. Sulodexide (SDX) is a highly purified glycosaminoglycan with antithrombotic, pro-fibrinolytic and anti-inflammatory activity used in the treatment of chronic venous disease (CVD), including patients with PTS. SDX has recently obtained clinical evidence in the "extension therapy" after initial-standard anticoagulant treatment for the secondary prevention of recurrent deep vein thrombosis (DVT). Herein, we investigated how SDX counteracts ED. MATERIALS AND METHODS: Human umbilical vein endothelial cells (HUVEC) were used. Metabolic and non metabolic-induced ED was induced by treating with methylglyoxal (MGO) or irradiation (IR), respectively. Bafilomycin A1 was used to inhibit autophagy. The production of reactive oxygen species (ROS), tetrazolium bromide (MTT) assay for cell viability, terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) assay for cell apoptosis, Real-time PCR and Western blot analysis for gene and protein expression were used. RESULTS: SDX protected HUVEC from MGO- or IR-induced apoptosis by counteracting the activation of the intrinsic and extrinsic caspase cascades. The cytoprotective effects of SDX resulted from a reduction in a) ROS production, b) neo-synthesis and release of pro-inflammatory cytokines (TNFα, IL1, IL6, IL8), c) DNA damage induced by MGO or IR. These effects were reduced when autophagy was inhibited. CONCLUSIONS: Data herein collected indicate the ability of SDX to counteract ED induced by metabolic or non-metabolic stresses by involving the intracellular autophagy pathway. Our experience significantly increases the knowledge of the mechanisms of action of SDX against ED and supports the use of SDX in the treatment of CVD, PTS and in the secondary prevention of recurrent DVT.


Glycosaminoglycans/pharmacology , Human Umbilical Vein Endothelial Cells/cytology , Pyruvaldehyde/adverse effects , X-Rays/adverse effects , Apoptosis/drug effects , Autophagy/drug effects , Cytokines/genetics , Cytokines/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/radiation effects , Humans , Models, Biological , Reactive Oxygen Species/metabolism
14.
Int J Mol Med ; 40(2): 539-548, 2017 Aug.
Article En | MEDLINE | ID: mdl-28627583

Methylglyoxal (MG) is one of the major precursors of advanced glycation end products (AGEs), which are considered to be one of the causes of diabetes and its complications. The root and rhizomes of black cohosh (Cimicifuga racemosa) have long been used medicinally, and deoxyactein is one of its major constituents. In the present study, the protective effects of deoxyactein against MG-induced oxidative cell damage were investigated in insulin-producing pancreatic ß-cells. We found that deoxyactein protected the pancreatic ß-cells against MG-induced cell death. Pre-treatment with deoxyactein significantly reduced the levels of intracellular reactive oxygen species (ROS), interleukin-1ß (IL-1ß), cardiolipin peroxidation, and protein adduct accumulation induced by MG. Pre-treatment of the cells with deoxyactein restored glyoxalase I activity and insulin secretion which were reduced by MG, and increased the mRNA expression of insulin 2 (INS2) and pancreatic and duodenal homeobox protein-1 (PDX-1). It also increased the levels of endogenous antioxidant enzymes, including superoxide dismutase (SOD) and glutathione peroxidase (GPX). Furthermore, treatment with deoxyactein increased the levels of sirtuin 1 (SIRT1) and peroxisome proliferator-activated receptor-γ co-activator-1α (PGC-1α). These findings indicate that deoxyactein may exert beneficial effects on pancreatic ß-cells via the upregulation of mitochondrial biogenesis. Taken together, these results suggest that deoxyactein may be used for the prevention of pancreatic ß-cell damage.


Insulin-Secreting Cells/drug effects , Mitochondria/drug effects , Organelle Biogenesis , Oxidants/adverse effects , Protective Agents/pharmacology , Pyruvaldehyde/adverse effects , Saponins/pharmacology , Triterpenes/pharmacology , Animals , Cell Line , Cell Survival/drug effects , Cimicifuga/chemistry , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Mitochondria/metabolism , Mitochondria/pathology , Oxidative Stress/drug effects , Protective Agents/chemistry , Protective Agents/isolation & purification , Rats , Reactive Oxygen Species/metabolism , Saponins/chemistry , Saponins/isolation & purification , Triterpenes/chemistry , Triterpenes/isolation & purification
15.
Arch Physiol Biochem ; 123(2): 121-127, 2017 May.
Article En | MEDLINE | ID: mdl-28005432

OBJECTIVE: To investigate whether (+)-catechin, a strong antioxidant, can prevent methylglyoxal (MGO)-induced cytotoxicity and its mechanism. METHODS: Cytotoxicity, apoptosis, reactive oxygen species (ROS) generation, hydrogen peroxide (H2O2) formation, mitochondrial membrane potential (MMP) and mitochondrial morphology were measured in EA.hy926 cells. RESULT: MGO (4 mM)-induced cytotoxicity was markedly inhibited by (+)-catechin (0.1-4 mM) in 24 h. 1 mM MGO-induced apoptotic cell death (44.7%) was significantly inhibited by 4 mM (+)-catechin (to 24.4%), 1 mM aminoguanidine (AG) (to 28.8%) or 4 mM N-acetylcysteine (NAC) (to 24.3%). (+)-Catechin (4 mM) or AG (4 mM) can inhibit the decrease of MMP induced by MGO (2-8 mM) in 3 h. (+)-Catechin (4 mM) or AG (4 mM) can inhibit MGO (4 mM)-induced mitochondrial swelling in 3 h. However, MGO (4 mM)-induced ROS and H2O2 generation was not prevented by (+)-catechin (4 mM). CONCLUSIONS: (+)-Catechin prevents MGO-induced cytotoxicity in EA.Hy926 cells through inhibiting apoptosis and mitochondrial damage.


Apoptosis/drug effects , Catechin/pharmacology , Endothelium, Vascular/drug effects , Mitochondria/drug effects , Mitochondria/pathology , Pyruvaldehyde/adverse effects , Cells, Cultured , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Humans , Hydrogen Peroxide/metabolism , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Oxidants/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
16.
Phytomedicine ; 23(2): 200-13, 2016 Feb 15.
Article En | MEDLINE | ID: mdl-26926182

BACKGROUND: Methylglyoxal (MGO) is known to be a major precursor of advanced glycation end products (AGEs) which are linked to diabetes and its related complications. Naturally occurring bioactive compounds could play an important role in countering AGEs thereby minimizing the risk associated with their formation. METHODS: In this study, eight specific bioactive compounds isolated from apple, tea and ginger were evaluated for their AGEs scavenging activity using Human Retinal Pigment Epithelial (H-RPE) cells treated with MGO. RESULTS: Among the eight specific compounds evaluated, (-)-epigallocatechin 3-gallate (EGCG) from tea, phloretin in apple, and [6]-shogaol and [6]-gingerol from ginger were found to be most effective in preventing MGO-induced cytotoxicity in the epithelial cells. Investigation of possible underlying mechanisms suggests that that these compounds could act by modulating key regulative detoxifying enzymes via modifying nuclear factor-erythroid 2-related factor 2 (Nrf2) function. MGO-induced cytotoxicity led to increased levels of AGEs causing increase in Nε-(Carboxymethyl) lysine (CML) and glutathione (GSH) levels and over expression of receptor for advanced glycation end products (RAGE). Data also showed that translocation of Nrf2 from cytosol to nucleus was inhibited, which decreased the expression of detoxifying enzyme like heme oxygenase-1 (HO-1). The most potent bioactive compounds scavenged dicarbonyl compounds, inhibited AGEs formation and significantly reduced carbonyl stress by Nrf2 related pathway and restoration of HO-1 expression. CONCLUSIONS: These findings demonstrated the protective effect of bioactive compounds derived from food sources against MGO-induced carbonyl stress through activation of the Nrf2 related defense pathway, which is of significant importance for therapeutic interventions in complementary treatment/management of diabetes-related complications.


Epithelial Cells/drug effects , Malus/chemistry , Phytochemicals/pharmacology , Pyruvaldehyde/adverse effects , Tea/chemistry , Zingiber officinale/chemistry , Catechin/analogs & derivatives , Catechin/pharmacology , Catechols/pharmacology , Cell Line , Epithelial Cells/metabolism , Fatty Alcohols/pharmacology , Glutathione/metabolism , Glycation End Products, Advanced/adverse effects , Heme Oxygenase-1/metabolism , Humans , Lysine/analogs & derivatives , Lysine/metabolism , NF-E2-Related Factor 2/metabolism , Phloretin/pharmacology , Protective Agents/pharmacology , Receptor for Advanced Glycation End Products/metabolism , Retinal Pigment Epithelium/cytology
17.
J Periodontal Res ; 51(6): 812-821, 2016 Dec.
Article En | MEDLINE | ID: mdl-26847600

BACKGROUND AND OBJECTIVES: Methylglyoxal is a toxic product derived from glucose metabolism that plays a role in inflammation, diabetes and aging. In addition, the periodontal pathogen Tannerella forsythensis may also generate this compound. However, the effects of methylglyoxal on gingival cells are still poorly understood. In the present study, we have explored whether methylglyoxal or methylglyoxal-treated collagen may modulate cell viability, death and proliferation in gingival connective tissue cells. In addition, we have searched for inflammatory mediators secreted by cells upon exposure to these conditions. MATERIAL AND METHODS: Primary cultures of human gingival fibroblasts were stimulated with soluble methylglyoxal or cultured over a collagen matrix glycated by this agent. Cell viability was evaluated through the MTS assay. Cell death was assessed through DAPI nuclear staining, annexin V and propidium iodide assays. Cell proliferation was evaluated through double immunofluorescence for DAPI and Ki67. Protein levels of matrix metalloproteinases and cytokines were assessed through antibody arrays, enzyme-linked immunosorbent assay, real-time reverse transcription polymerase chain reaction and immunofluorescence. Statistical analysis was performed using the Kruskall-Wallis and Mann-Whitney tests. RESULTS: Soluble methylglyoxal, but not culture of gingival fibroblasts over a methylglyoxal-modified collagen matrix, induced a reduction on cell viability. Moreover, soluble methylglyoxal induced apoptotic cell death as indicated by DAPI nuclear staining, annexin V and propidium iodide assays. Neither soluble methylglyoxal, nor methylglyoxal-modified collagen modified cell proliferation. Using an antibody array, enzyme-linked immunosorbent assay and immunofluorescence assays, we determined that both, soluble methylglyoxal and methylglyoxal-modified collagen stimulated an increase in tissue inhibitor of metalloproteinase (TIMP)-1 protein levels. CONCLUSIONS: Soluble methylglyoxal is a highly cytotoxic compound that induces cell death through apoptosis in gingival fibroblasts. TIMP-1 is induced in these cells upon direct exposure to methylglyoxal or after culture of gingival fibroblasts over methylglyoxal-treated collagen. As TIMP-1 has been implicated in cell survival and matrix remodeling, we propose that increased TIMP-1 protein levels may be part of a protective response of gingival connective tissue cells upon exposure to methylglyoxal or after the interaction with the collagen matrix that has been modified by this agent.


Gingiva/drug effects , Pyruvaldehyde/adverse effects , Adolescent , Adult , Apoptosis/drug effects , Cell Death/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Collagen/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Fibroblasts/drug effects , Fluorescent Antibody Technique , Gingiva/cytology , Humans , Male , Pyruvaldehyde/metabolism , Tissue Inhibitor of Metalloproteinase-1/metabolism , Young Adult
18.
Chem Biol Interact ; 244: 169-77, 2016 Jan 25.
Article En | MEDLINE | ID: mdl-26723916

Honokiol is an active compound isolated from Magnolia officinalis that has been used without notable side effects in traditional medicine. We investigated the effects of honokiol against methylglyoxal (MG)-induced cytotoxicity in MC3T3-E1 osteoblast cells and the possible molecular mechanism(s) involved. The results showed that honokiol alleviated MG-induced cell death and the production of intracellular ROS, mitochondrial superoxide, cardiolipin peroxidation, and inflammatory cytokines. MG induction of the soluble receptor for advanced glycation end product (AGE) was reduced by pretreatment with honokiol. Furthermore, honokiol increased the levels of Nrf2 and increased the levels of glutathione and the activity of glyoxalase I. Pretreatment with honokiol prior to MG exposure reduced MG-induced mitochondrial dysfunction and alleviated MG-induced reduction of nitric oxide and PGC1α levels, suggesting that honokiol may induce mitochondrial biogenesis. It was concluded that honokiol could be useful in the attenuation of MG-induced cell damage.


Biphenyl Compounds/pharmacology , Lignans/pharmacology , Osteoblasts/drug effects , Osteoblasts/pathology , Protective Agents/pharmacology , Pyruvaldehyde/adverse effects , Animals , Cell Death/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Mice , Osteoblasts/metabolism , Structure-Activity Relationship
19.
Chem Res Toxicol ; 28(9): 1666-74, 2015 Sep 21.
Article En | MEDLINE | ID: mdl-26250540

Diabetes mellitus is a metabolic disorder constituting a major health problem whose prevalence has gradually increased worldwide over the past few decades. Type 2 diabetes mellitus (T2DM) remains more complex and heterogeneous and arises as a combination of insulin resistance and inadequate functional ß-cell mass and comprises about 90% of all diabetic cases. Appropriate experimental animal models are essential for understanding the molecular basis, pathogenesis of complications, and the utility of therapeutic agents to abrogate this multifaceted disorder. Currently, animal models for T2DM are obtained as spontaneously developed diabetes or diabetes induced by chemicals or dietary manipulations or through surgical or genetic methods. The currently used diabetogenic agents have certain limitations. Recently, methylglyoxal (MG), a highly reactive compound derived mainly from glucose and fructose metabolism has been implicated in diabetic complications. MG is a major precursor of the advanced glycation end product (AGE) and promotes impaired functions of insulin signaling, GLUT transporters, anion channels, kinases, and endothelial cells and is finally involved in apoptosis. Recent array of literature also cited that higher concentrations of MG causes rapid depolarization, elevated intracellular Ca(2+) concentration, and acidification in pancreatic ß-cells. This review henceforth highlights the mechanism of action of MG and its implications in the pathophysiology of experimental diabetes.


Diabetes Mellitus, Type 2/chemically induced , Disease Models, Animal , Pyruvaldehyde/adverse effects , Animals , Diabetes Mellitus, Type 2/physiopathology , Pyruvaldehyde/chemical synthesis
20.
Lab Invest ; 95(9): 1029-43, 2015 Sep.
Article En | MEDLINE | ID: mdl-26121315

Appropriate fluid balance is important for good clinical outcomes and survival in patients on peritoneal dialysis. We recently reported that lymphangiogenesis associated with fibrosis developed in the peritoneal cavity via the transforming growth factor-ß1-vascular endothelial growth factor-C (VEGF-C) pathway. We investigated whether VEGF receptor-3 (VEGFR-3), the receptor for VEGF-C and -D, might be a new target to improve net ultrafiltration by using adenovirus-expressing soluble VEGFR-3 (Adeno-sVEGFR-3) in rodent models of peritoneal injury induced by methylglyoxal (MGO). We demonstrated that lymphangiogenesis developed in these MGO models, especially in the diaphragm, indicating that lymphangiogenesis is a common feature in the peritoneal cavity with inflammation and fibrosis. In MGO models, VEGF-D was significantly increased in the diaphragm; however, VEGF-C was not significantly upregulated. Adeno-sVEGFR-3, which was detected on day 50 after administration via tail vein injections, successfully suppressed lymphangiogenesis in the diaphragm and parietal peritoneum in mouse MGO models without significant effects on fibrosis, inflammation, or neoangiogenesis. Drained volume in the peritoneal equilibration test using a 7.5% icodextrin peritoneal dialysis solution (the 7.5% icodextrin peritoneal equilibration test) was improved by Adeno-sVEGFR-3 on day 22 (P<0.05) and day 50 after reduction of inflammation (P<0.01), indicating that the 7.5% icodextrin peritoneal equilibration test identifies changes in lymphangiogenesis. The solute transport rate was not affected by suppression of lymphangiogenesis. In human peritoneal dialysis patients, the dialysate to plasma ratio of creatinine positively correlated with the dialysate VEGF-D concentration (P<0.001). VEGF-D mRNA was significantly higher in the peritoneal membranes of patients with ultrafiltration failure, indicating that VEGF-D is involved in the development of lymphangiogenesis in peritoneal dialysis patients. These results indicate that VEGFR-3 is a new target to improve net ultrafiltration by suppressing lymphatic absorption and that the 7.5% icodextrin peritoneal equilibration test is useful for estimation of lymphatic absorption.


Lymphangiogenesis/drug effects , Peritoneal Dialysis/adverse effects , Peritoneum/drug effects , Pyruvaldehyde/adverse effects , Ultrafiltration/methods , Vascular Endothelial Growth Factor Receptor-3/pharmacology , Animals , Creatinine/analysis , Creatinine/blood , Dialysis Solutions/chemistry , Enzyme-Linked Immunosorbent Assay , Glucans , Glucose , Humans , Icodextrin , Immunohistochemistry , Mice , Mice, Inbred C57BL , Peritoneal Dialysis/methods , Peritoneum/injuries , Statistics, Nonparametric , Vascular Endothelial Growth Factor D/analysis , Vascular Endothelial Growth Factor D/blood , Vascular Endothelial Growth Factor Receptor-3/metabolism
...