Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Drug Deliv Transl Res ; 13(9): 2286-2296, 2023 09.
Article in English | MEDLINE | ID: mdl-36749479

ABSTRACT

Diabetic wounds are a serious complication of diabetes mellitus (DM) that can lead to persistent infection, amputation, and even death. Prolonged oxidative stress has been widely recognized as a major instigator in the development of diabetic wounds; therefore, oxidative stress is considered a promising therapeutic target. In the present study, Keap1/Nrf2 signaling was confirmed to be activated in streptozotocin (STZ)-induced diabetic mice and methylglyoxal (MGO)-treated human umbilical vein endothelial cells (HUVECs). Knockdown of Keap1 by siRNA reversed the increase in Keap1 levels, promoted the nuclear translocation of Nrf2, and increased the expression of HO-1, an antioxidant protein. To explore therapeutic delivery strategies, milk-derived exosomes (mEXOs) were developed as a novel, efficient, and non-toxic siRNA carrier. SiRNA-Keap1 (siKeap1) was loaded into mEXOs by sonication, and the obtained mEXOs-siKeap1 were found to promote HUVEC proliferation and migration while relieving oxidative stress in MGO-treated HUVECs. Meanwhile, in a mouse model of diabetic wounds, injection of mEXOs-siKeap1 significantly accelerated diabetic wound healing with enhanced collagen formation and neovascularization. Taken together, these data support the development of Keap1 knockdown as a potential therapeutic strategy for diabetic wounds and demonstrated the feasibility of mEXOs as a scalable, biocompatible, and cost-effective siRNA delivery system. The therapeutic effect of siKeap1-loaded mEXOs on diabetic wound healing was assessed. First, we found that the expression of Keap1 was upregulated in the wounds of diabetic mice and in human umbilical vein endothelial cells (HUVECs) pretreated with methylglyoxal (MGO). Next, we extracted exosomes from raw milk by differential centrifugation and loaded siKeap1 into milk-derived exosomes by sonication. The in vitro application of the synthetic complex (mEXOs-siKeap1) was found to increase the nuclear localization of Nrf2 and the expression of the antioxidant protein HO-1, thus reversing oxidative stress. Furthermore, in vivo mEXOs-siKeap1 administration significantly accelerated the healing rate of diabetic wounds (Scheme 1). Scheme 1 Schematic diagram. A Synthesis of mEXOs-siKeap1 complex. B Mechanism of mEXOs-siKeap1 in vitro. C The treatment effect of mEXOs-siKeap1 on an in vivo mouse model of diabetic wounds.


Subject(s)
Diabetes Mellitus, Experimental , Exosomes , Mice , Humans , Animals , Antioxidants/pharmacology , Kelch-Like ECH-Associated Protein 1/metabolism , Diabetes Mellitus, Experimental/drug therapy , RNA, Small Interfering/pharmacology , Milk/metabolism , Exosomes/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/pharmacology , Magnesium Oxide/metabolism , Magnesium Oxide/pharmacology , Magnesium Oxide/therapeutic use , Pyruvaldehyde/metabolism , Pyruvaldehyde/pharmacology , Pyruvaldehyde/therapeutic use , Wound Healing , Oxidative Stress , Human Umbilical Vein Endothelial Cells/metabolism
2.
Oxid Med Cell Longev ; 2022: 8854457, 2022.
Article in English | MEDLINE | ID: mdl-35035668

ABSTRACT

Cerebral endothelial cells play an essential role in brain angiogenesis, and their function has been found to be impaired in diabetes. Methylglyoxal (MG) is a highly reactive dicarbonyl metabolite of glucose formed mainly during glycolysis, and its levels can be elevated in hyperglycemic conditions. MG is a potent precursor of AGEs (advanced glycation end-products). In this study, we investigated if MG can induce angiogenesis dysfunction and whether MG scavengers can ameliorate angiogenesis dysfunction induced by MG. Here, we used cultured human brain microvascular endothelial cells (HBMECs) treated with MG and oxygen-glucose deprivation (OGD) to mimic diabetic stroke in vitro. We also used the MG challenged chicken embryo chorioallantoic membrane (CAM) to study angiogenesis in vivo. Interestingly, administration of MG significantly impaired cell proliferation, cell migration, and tube formation and decreased protein expression of angiogenesis-related factors, which was rescued by three different MG scavengers, glyoxalase 1 (GLO1), aminoguanidine (AG), and N-acetyl cysteine (NAC). In cultured CAM, MG exposure significantly reduced angiogenesis and the angiogenesis-related dysfunction could be attenuated by pretreatment with AG or NAC. Treatment of cultured HBMECs with MG plus OGD increased cellular apoptosis significantly, which could be prevented by exposure to GLO1, AG, or NAC. We also noted that administration of MG increased cellular oxidative stress as measured by reactive oxygen species (ROS) generation, enhanced AGE accumulation, and receptor for advanced glycation end-product (RAGE) expression in the cultured HBMECs, which were partially reversed by GLO1, AG, or NAC. Taken together, our findings demonstrated that GLO1, AG, or NAC administration can ameliorate MG-induced angiogenesis dysfunction, and this can be mainly attributed to attenuated ROS production, reduced cellular apoptosis, and increased levels of angiogenic factors. Overall, this study suggested that GLO1, AG, or NAC may be promising candidate compounds for the treatment of angiogenesis dysfunction caused by hyperglycemia in diabetic ischemic stroke.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Glucose/metabolism , Oxidative Stress/drug effects , Oxygen/metabolism , Pyruvaldehyde/therapeutic use , Angiogenesis Inhibitors/pharmacology , Animals , Chick Embryo , Humans , Pyruvaldehyde/pharmacology
3.
Planta Med ; 88(12): 1026-1035, 2022 Oct.
Article in English | MEDLINE | ID: mdl-34861700

ABSTRACT

Accumulation of advanced glycation end products (AGEs) of the Maillard reaction has been implicated in the pathogenesis of diabetes and its complications. Connarus ruber has been used as a folk remedy for several diseases, including diabetes; however, its underlying mechanism has not yet been investigated. This study investigated the effects of C. ruber extract against glycation on collagen-linked AGEs in vitro and streptozotocin-induced diabetic rats (STZ-DM rats) in vivo. The antiglycation activities of C. ruber extract and aminoguanidine (AG) were examined using a collagen glycation assay kit. Nonfluorescent AGE, Nε-carboxymethyl lysine (CML), Nω-carboxymethyl arginine, and Nε-carboxyethyl lysine levels were measured via electrospray ionization-liquid chromatography-tandem mass spectrometry. The effect of the extract on the cytotoxicity of methylglyoxal (MG), a precursor of AGEs, was examined in HL60 cells. STZ-DM rats were treated with the extract for 4 wk, and the effect was assessed using biochemical markers in the serum and CML-positive cells in renal tissues. C. ruber extract dose-dependently inhibited the glycation of collagen and formation of nonfluorescent AGEs, which was comparable to AG, and it significantly attenuated MG-induced cytotoxicity in HL60 cells. Furthermore, the glycated albumin levels in STZ-DM rats decreased, the increase in serum lipid levels was reversed, and immunohistochemistry demonstrated that CML deposition in the glomerulus of STZ-DM rats significantly decreased. Although further studies are needed, C. ruber could be a potential therapeutic for preventing and progressing many pathological conditions, including diabetes.


Subject(s)
Connaraceae , Diabetes Mellitus, Experimental , Animals , Arginine/analysis , Arginine/therapeutic use , Collagen , Diabetes Mellitus, Experimental/drug therapy , Glycation End Products, Advanced , Guanidines , Lipids , Lysine/analysis , Lysine/therapeutic use , Pyruvaldehyde/therapeutic use , Rats , Streptozocin
4.
Platelets ; 32(4): 507-515, 2021 May 19.
Article in English | MEDLINE | ID: mdl-32449466

ABSTRACT

Advanced glycation end products (AGEs) arising from dietary intake have been associated with numerous chronic diseases including cardiovascular diseases. The interaction between platelets and AGEs has been proposed to play a role in the etiology of cardiovascular diseases. However, the effects of the interaction between platelets and Maillard reaction products generated from glyoxal (Gly) or methylglyoxal (MG) are poorly understood. In this work, the effects of AGEs generated by the reaction between Gly or MG with Lys or bovine serum albumin (BSA) on platelet activation and aggregation were assessed. AGEs were generated incubating Gly or MG with Lys or BSA during 5 hours or 14 days, respectively. AGEs generation were characterized by kinetic studies and by amino acid analysis. Human platelet-rich plasma (PRP) was incubated with different concentrations of AGEs from Lys-MG or Lys-Gly and BSA-MG or BSA-Gly. Platelet activation was determined quantifying the expression of CD62 (P-selectin) in PRP exposed to different AGEs concentrations. It was found that Lys-MG and Lys-Gly induced an increase in P-selectin expression (p < .05), being 33.9% higher for Lys-MG when compared to Lys-Gly. Platelets incubated in the presence of BSA-MG and BSA-Gly did not show an increase in the P-selectin expression. Platelet aggregation was significantly higher for the mixture Lys-MG (in all the range of concentrations evaluated), whereas for Lys-Gly it was only significant the highest concentration (Lys 168 µM/Gly 168 µM). It was observed a significant increase in platelet aggregation induced by ADP for samples BSA-Gly. AGEs formed with MG-Lys induce a higher activation and aggregation of platelets when compared to those formed from Gly-Lys.


Subject(s)
Glycation End Products, Advanced/adverse effects , Glyoxal/therapeutic use , Platelet Activation/genetics , Platelet Aggregation/genetics , Pyruvaldehyde/therapeutic use , Glyoxal/pharmacology , Humans , Pyruvaldehyde/pharmacology
5.
Int J Nanomedicine ; 15: 3681-3693, 2020.
Article in English | MEDLINE | ID: mdl-32547022

ABSTRACT

BACKGROUND: Fungal infections are becoming more prevalent and threatening because of the continuous emergence of azole-resistant fungal infections. The present study was aimed to assess the activity of free Methylglyoxal (MG) or MG-conjugated chitosan nanoparticles (MGCN) against fluconazole-resistant Candida albicans. MATERIALS AND METHODS: A novel formulation of MGCN was prepared and characterized to determine their size, shape and polydispersity index. Moreover, the efficacy of fluconazole or MG or MGCN was determined against intracellular C. albicans in macrophages and the systematic candidiasis in a murine model. The safety of MG or MGCN was tested in mice by analyzing the levels of hepatic and renal toxicity parameters. RESULTS: Candida albicans did not respond to fluconazole, even at the highest dose of 20 mg/kg, whereas MG and MGCN effectively eliminated C. albicans from the macrophages and infected mice. Mice in the group treated with MGCN at a dose of 10 mg/kg exhibited a 90% survival rate and showed the lowest fungal load in the kidney, whereas the mice treated with free MG at the same dose exhibited 50% survival rate. Moreover, the administration of MG or MGCN did not induce any liver and kidney toxicity in the treated mice. CONCLUSION: The findings of the present work suggest that MGCN may be proved a promising therapeutic formulation to treat azole-resistant C. albicans infections.


Subject(s)
Candidiasis/drug therapy , Chitosan/chemistry , Drug Resistance, Fungal , Fluconazole/therapeutic use , Nanoparticles/chemistry , Pyruvaldehyde/therapeutic use , Animals , Antifungal Agents/pharmacology , Antifungal Agents/therapeutic use , Candida albicans/drug effects , Candida albicans/growth & development , Candidiasis/microbiology , Disease Models, Animal , Drug Resistance, Fungal/drug effects , Female , Fluconazole/pharmacology , Hyphae/drug effects , Hyphae/growth & development , Mice, Inbred BALB C , Microbial Sensitivity Tests , Nanoparticles/ultrastructure , Particle Size , Pyruvaldehyde/pharmacology
6.
Biochem Pharmacol ; 166: 231-241, 2019 08.
Article in English | MEDLINE | ID: mdl-31158339

ABSTRACT

Protein glycation in the body can lead to malfunction of intracellular and extracellular proteins. Reactive carbonyl species (RCS) have been identified to be key intermediates in the reactions. The reaction products, generally termed as advanced glycation end products (AGEs), have been implicated in the development of diabetic complications. In this study, the activity of apigenin (API), a natural flavone in scavenging RCS and the molecular mechanism involved in its protective effect against AGEs-induced oxidative stress and inflammation were examined in vitro. Results showed that API could directly trap methylglyoxal (MGO) to form API-MGO adducts, thus inhibiting AGEs formation. API and di-apigenin adduct (DMA) were found to inhibit AGEs-induced oxidative stress and inflammation in human umbilical vein endothelial cells (HUVECs) by significantly suppressing reactive oxygen species (ROS) production (30% relative to control) and decreasing the protein expression of pro-inflammatory cytokines and adhesion molecules by 30-70%. Further mechanistic investigation revealed that the protective effect was likely mediated via suppression of the extracellular-signal-regulated kinase 1/2 (ERK)/nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling pathway initiated by AGEs-RAGE (receptor for AGEs) interaction and induction of ERK/nuclear factor (erythroid-derived 2)-like 2 (Nrf2) pathway with subsequent up-regulation of antioxidant defense molecules. In summary, our results suggest that API possesses great potential to protect against AGEs-associated health disorders by modulating cellular inflammatory and antioxidant defense signaling pathways.


Subject(s)
Apigenin/pharmacology , Glycation End Products, Advanced/antagonists & inhibitors , Human Umbilical Vein Endothelial Cells/drug effects , Oxidative Stress/drug effects , Pyruvaldehyde/pharmacology , Animals , Apigenin/therapeutic use , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Glycation End Products, Advanced/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Inflammation/drug therapy , Inflammation/metabolism , Oxidative Stress/physiology , Pyruvaldehyde/therapeutic use , Swine
7.
Neurotox Res ; 34(1): 164-172, 2018 07.
Article in English | MEDLINE | ID: mdl-29417441

ABSTRACT

There is a strong association between neurodegeneration and protein glycation; possible origins of neurotoxic glycated protein, also called glycotoxins, include (i) diet (i.e., proteins cooked at high temperatures), (ii) protein glycation in the gut, and (iii) intracellular reaction of proteins with deleterious aldehydes, especially methylglyoxal (MG). It is likely that excessive glycolysis provokes increased generation of dihydroxyacetone phosphate which decomposes into MG due to activity-induced deamidation of certain asparagine residues in the glycolytic enzyme triose-phosphate isomerase (TPI). It is suggested that, following hyperglycemia, erythrocytes (i) possibly participate in MG distribution throughout the body and (ii) could provide a source of glycated alpha-synuclein which also accumulates in PD brains as Lewy bodies. The dipeptide carnosine, recently shown to be present in erythrocytes, could help to protect against MG reactivity by scavenging the reactive bicarbonyl, especially if glyoxalase activity is insufficient, as often occurs during aging. By reacting with MG, carnosine may also prevent generation of the neurotoxin 1-acetyl-6,7-dihydroxy-1,2,3,4-tetrahydroisoquinoline (ADTIQ), which accumulates in PD and diabetic brains. It is suggested that carnosine's therapeutic potential could be explored via nasal administration in order to avoid the effects of serum carnosinase. The possibility that some glycated proteins (e.g., alpha-synuclein) could possess prion-like properties is also considered.


Subject(s)
Carnosine/toxicity , Diet/methods , Neurotoxicity Syndromes/therapy , Pyruvaldehyde/therapeutic use , Animals , Humans , Neurotoxins/toxicity , Tetrahydroisoquinolines/toxicity
8.
Exp Neurol ; 296: 74-82, 2017 10.
Article in English | MEDLINE | ID: mdl-28729113

ABSTRACT

Bortezomib is a first-line chemotherapeutic drug widely used for multiple myeloma and other nonsolid malignancies. Although bortezomib-induced persistent pain is easily diagnosed in clinic, the pathogenic mechanism remains unclear. Here, we studied this issue with use of a rat model of systemic intraperitoneal administration of bortezomib for consecutive 5days. Consisted with our previous study, we found that bortezomib treatment markedly induced mechanical allodynia in rats. Furthermore, we first found that bortezomib treatment significantly induced the upregulation of methylglyoxal in spinal dorsal horn of rats. Spinal local application of methylglyoxal also induced mechanical allodynia and central sensitization in normal rats. Moreover, administration of bortezomib upregulated the expression of receptors for advanced glycation end products (RAGE) and phosphorylated STAT3 (p-STAT3) in dorsal horn. Importantly, intrathecal injection of metformin, a known scavenger of methylglyoxal, significantly attenuated the upregulation of methylglyoxal and RAGE in dorsal horn, central sensitization and mechanical allodynia induced by bortezomib treatment, and blockage of RAGE also prevented the upregulation of p-STAT3, central sensitization and mechanical allodynia induced by bortezomib treatment. In addition, inhibition of STAT3 activity by S3I-201 attenuated bortezomib-induced mechanical allodynia and central sensitization. Local knockdown of STAT3 also ameliorated the mechanical allodynia induced by bortezomib administration. Our results suggest that accumulation of methylglyoxal may activate the RAGE/STAT3 signaling pathway in dorsal horn, and contributes to the spinal central sensitization and persistent pain induced by bortezomib treatment.


Subject(s)
Bortezomib/toxicity , Central Nervous System Sensitization/drug effects , Pain/chemically induced , Pain/drug therapy , Pyruvaldehyde/pharmacology , Pyruvaldehyde/therapeutic use , Spinal Cord/physiopathology , Animals , Antineoplastic Agents/toxicity , Disease Models, Animal , Male , Nerve Fibers, Unmyelinated/drug effects , Nerve Fibers, Unmyelinated/physiology , Pain/pathology , Pain Measurement/drug effects , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Receptor for Advanced Glycation End Products/genetics , Receptor for Advanced Glycation End Products/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Spinal Cord/cytology , Spinal Cord/drug effects , Synaptic Potentials/drug effects , Synaptic Potentials/genetics , Transduction, Genetic , Up-Regulation/drug effects , Up-Regulation/genetics
9.
Mol Carcinog ; 56(9): 2086-2103, 2017 09.
Article in English | MEDLINE | ID: mdl-28418078

ABSTRACT

Triple negative breast cancer (TNBC) tends to form aggressive tumors associated with high mortality and morbidity which urge the need for development of new therapeutic strategies. Recently, the normal metabolite Methylglyoxal (MG) has been documented for its anti-proliferative activity against human breast cancer. However, the mode of action of MG against TNBC remains open to question. In our study, we investigated the anticancer activity of MG in MDA MB 231 and 4T1 TNBC cell lines and elucidated the underlying mechanisms. MG dose-dependently caused cell death, induced apoptosis, and generated ROS in both the TNBC cell lines. Furthermore, such effects were attenuated in presence of ROS scavenger N-Acetyl cysteine. MG triggered mitochondrial cytochrome c release in the cytosol and up-regulated Bax while down-regulated anti-apoptotic protein Bcl-2. Additionally, MG treatment down-regulated phospho-akt and inhibited the nuclear translocation of the p65 subunit of NF-κB. MG exhibited a tumor suppressive effect in BALB/c mouse 4T1 breast tumor model as well. The cytotoxic effect was studied using MTT assay. Apoptosis, ROS generation, and mitochondrial dysfunction was evaluated by flow cytometry as well as fluorescence microscopy. Western blot assay was performed to analyze proteins responsible for apoptosis. This study demonstrated MG as a potent anticancer agent against TNBC both in vitro and in vivo. The findings will furnish fresh insights into the treatment of this subgroup of breast cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Mitochondria/drug effects , Pyruvaldehyde/therapeutic use , Reactive Oxygen Species/metabolism , Triple Negative Breast Neoplasms/drug therapy , Animals , Cell Line, Tumor , Cell Survival/drug effects , Cytochromes c/metabolism , Drug Screening Assays, Antitumor , Humans , Mice , Mice, Inbred BALB C , Proto-Oncogene Proteins c-akt/metabolism , Transcription Factor RelA/metabolism , Triple Negative Breast Neoplasms/metabolism , Xenograft Model Antitumor Assays
10.
Dig Liver Dis ; 45(11): 938-46, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24071451

ABSTRACT

BACKGROUND: Hepatocellular carcinoma accounts for more than 600,000 deaths per year due to it being a highly invasive tumor. The α-dicarbonyl, methylglyoxal demonstrates efficacy at reducing tumor burden, however the anti-cancerous activities of 3-deoxyglucosone, have never been studied. AIMS: To determine the anti-cancerous potential of methylglyoxal and 3-deoxyglucosone on liver tumor cells. METHODS: The in vitro effects of methylglyoxal and 3-deoxyglucosone were studied by investigating migration, invasion, and adhesion of Huh-7, HepG2, and Hep3B cells. RESULTS: 3-Deoxyglucosone inhibited migration of Huh-7 and HepG2 cells. Methylglyoxal decreased migration of HepG2 cells. Additionally, 3-deoxyglucosone and methylglyoxal impaired invasion, and adhesion of Huh-7 and HepG2 cells. In Hep3B cells, a p53 null cell line, 3-deoxyglucosone and methylglyoxal had no effect on migration, invasion, or adhesion. However, both compounds inhibited invasion of wild-type p53 transfected Hep3B cells. Silencing of p53 in Huh-7 and HepG2 cells abrogated the effects of the α-dicarbonyls on cell invasion. 3DG and MG did not alter p53 total protein but promoted nuclear translocation of p53. CONCLUSIONS: These studies suggest that 3-deoxyglucosone and methylglyoxal impair invasion, migration, and adhesion of hepatocellular carcinoma. The effects of both compounds on cell invasion are dependent on p53 and imply that α-dicarbonyls could be efficacious in the treatment of p53-expressing invasive liver tumors.


Subject(s)
Cell Adhesion/drug effects , Deoxyglucose/analogs & derivatives , Down-Regulation/drug effects , Genes, p53/genetics , Liver Neoplasms/drug therapy , Neoplasm Invasiveness/genetics , Pyruvaldehyde/therapeutic use , Apoptosis , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Deoxyglucose/therapeutic use , Drug Therapy, Combination , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology
11.
Arch Physiol Biochem ; 118(2): 58-68, 2012 May.
Article in English | MEDLINE | ID: mdl-22364223

ABSTRACT

CONTEXT: Adipose tissue is one of the first organs to develop insulin resistance even with moderate BMI. However, the contribution of developing hyperglycaemia and concomitant methylglyoxal increment to tissue dysfunction during type 2 diabetes progression was not addressed before. METHODS: Young and aged Wistar and Goto-Kakizaki rats (non-obese model of type 2 diabetes) and a group of MG-treated W rats were used to investigate the chronic effects of hyperglycaemia and ageing and specifically MG-induced mechanisms. RESULTS: Diabetic and aged rats showed decreased adipose tissue irrigation and interstitial hypoxia. Hyperglycaemia of diabetic rats leaded to fibrosis and accumulation of PAS-positive components, exacerbated in aged animals, which also showed decreased hipoadiponectinemia, increased MCP-1 expression and macrophage infiltration to glycated fibrotic regions. MG leaded to increased free fatty acids, hipoadiponectinemia, decreased irrigation, hypoxia and macrophage recruitment for glycated fibrotic regions. CONCLUSIONS: MG contributes to dysfunction of adipose tissue during type 2 diabetes progression.


Subject(s)
Adipose Tissue/drug effects , Pyruvaldehyde/pharmacology , Adipokines/metabolism , Adipose Tissue/cytology , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , Apoptosis/drug effects , Biomarkers/metabolism , Cell Hypoxia/drug effects , Fibrosis , Hyperglycemia/metabolism , Hyperglycemia/pathology , Inflammation/drug therapy , Neovascularization, Physiologic/drug effects , Obesity/pathology , Oxidative Stress/drug effects , Pyruvaldehyde/metabolism , Pyruvaldehyde/therapeutic use , Rats , Rats, Wistar
12.
Biochemistry (Mosc) ; 74(10): 1059-69, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19916918

ABSTRACT

In various organisms, an array of enzymes is involved in the synthesis and breakdown of methylglyoxal. Through these enzymes, it is intimately linked to several other physiologically important metabolites, suggesting that methylglyoxal has some important role to play in the host organism. Several in vitro and in vivo studies showed that methylglyoxal acts specifically against different types of malignant cells. These studies culminated in a recent investigation to evaluate a methylglyoxal-based formulation in treating a small group of cancer patients, and the results were promising. Methylglyoxal acts against a number of pathogenic microorganisms. However, recent literature abounds with the toxic effects of methylglyoxal, which are supposed to be mediated through methylglyoxal-derived advanced glycation end products (AGE). Many diseases such as diabetes, cataract formation, hypertension, and uremia are proposed to be intimately linked with methylglyoxal-derived AGE. However methylglyoxal-derived AGE formation and subsequent pathogenesis might be a very minor event because AGE are nonspecific reaction products that are derived through the reactions of carbonyl groups of reducing sugars with amino groups present in the side chains of lysine and arginine and in terminal amino groups of proteins. Moreover, the results of some in vitro experiments with methylglyoxal under non-physiological conditions were extrapolated to the in vivo situation. Some experiments even showed contradictory results and were differently interpreted. For this reason conclusions about the potential beneficial effects of methylglyoxal have often been neglected, thus hindering the advancement of medical science and causing some confusion in fundamental understanding. Overall, the potential beneficial effects of methylglyoxal far outweigh its possible toxic role in vivo, and it should be utilized for the benefit of suffering humanity.


Subject(s)
Cataract/drug therapy , Maillard Reaction/drug effects , Oxidative Stress/physiology , Pyruvaldehyde/toxicity , Saccharomyces cerevisiae Proteins/chemistry , Animals , Arginine/chemistry , Cataract/complications , Cataract/etiology , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2/metabolism , Drug Delivery Systems , Glycation End Products, Advanced/pharmacology , Glycation End Products, Advanced/toxicity , Humans , Kidney Failure, Chronic/chemically induced , Lysine/chemistry , Oxidative Stress/drug effects , Protein Conformation , Protein Folding , Pyruvaldehyde/administration & dosage , Pyruvaldehyde/therapeutic use , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins/poisoning , Serum Albumin
13.
Drug Metabol Drug Interact ; 23(1-2): 69-91, 2008.
Article in English | MEDLINE | ID: mdl-18533365

ABSTRACT

Methylglyoxal, an alpha-oxoaldehyde discovered in the 1880s, has had a hectic scientific career, at times being considered of fundamental importance and at other times viewed as playing a very subordinate role. Much has been learned about methylglyoxal, but the function of its production in the metabolic machinery is still unknown. This paper gives an overview of the changing role of methylglyoxal from a historical aspect and arrives at the conclusion that methylglyoxal is tightly bound to glycolysis from an evolutionary perspective, its production therefore being inevitable. It is not situated in the main stream of the glycolytic sequence, but a role can be assigned to its production in the phosphate supply of operating glycolysis in some prokaryotes and yeast under conditions of phosphate deficiency. This function is presumed to be performed by the enzyme methylglyoxal synthase, which is specialized for the conversion of dihydroxyacetone-phosphate to methylglyoxal. However, it is still unknown whether this enzyme and this kind of regulation also exist in animals.


Subject(s)
Biochemistry/history , Glycolysis/physiology , Pyruvaldehyde/history , Animals , Antineoplastic Agents/therapeutic use , Evolution, Molecular , History, 19th Century , History, 20th Century , Neoplasms/drug therapy , Pyruvaldehyde/metabolism , Pyruvaldehyde/therapeutic use , Signal Transduction/physiology
14.
Drug Metabol Drug Interact ; 23(1-2): 175-210, 2008.
Article in English | MEDLINE | ID: mdl-18533369

ABSTRACT

A historical perspective on methylglyoxal research is briefly presented, mentioning the documented anticancer and antiviral effects of methylglyoxal. The idea and the supporting experimental evidence of Albert Szent-Györgyi et al. that methylglyoxal is a natural growth regulator and can act as an anticancer agent are mentioned. Previously a few in vivo studies suggested safe administration of methylglyoxal. However, recent literature abounds with the toxic effects of methylglyoxal. The authors present a brief critical overview of studies indicating both toxic and beneficial effects of methylglyoxal and suggest that the beneficial effects of methylglyoxal outweigh its toxic effects. Encouraged by the studies of Szent-Györgyi et al., the present authors undertook systematic investigations to understand the mechanism of the anticancer effect of methylglyoxal. The results of these investigations led to the proposal that the fundamental changes in malignant cells are critical alterations of glyceraldehyde-3-phosphate dehydrogenase and mitochondrial complex I, and methylglyoxal's anticancer effect might be mediated by acting on these altered sites. Moreover, a new hypothesis on cancer has been proposed, suggesting that excessive ATP formation in cells may lead to malignancy. Toxicity and pharmacokinetic studies were performed on animals and it was observed that methylglyoxal is potentially safe for humans. A methylglyoxal-based anticancer formulation was developed and a three-phase study of treating a total number of 86 cancer patients was carried out. The results appear to be promising. Most of the cancer patients benefited greatly and a significant number of patients became free of the disease. Contrary to the effect of existing anticancer drugs, this methylglyoxal-based formulation is devoid of any toxic effect and reasonably effective against a wide variety of cancers. The symptomatic improvements of the many patients who died of progressive disease suggest that the formulation could also be used for palliation. The authors urge the scientific community to test the formulation and if found effective then to improve it further.


Subject(s)
Antineoplastic Agents/pharmacology , Pyruvaldehyde/pharmacology , Aged , Aged, 80 and over , Animals , Antineoplastic Agents/therapeutic use , Clinical Trials as Topic , Diabetes Complications/metabolism , Female , Glycation End Products, Advanced/biosynthesis , Humans , Male , Middle Aged , Neoplasms/drug therapy , Pyruvaldehyde/metabolism , Pyruvaldehyde/therapeutic use
16.
Toxicol Appl Pharmacol ; 212(1): 45-58, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16112157

ABSTRACT

Previous in vivo studies from several laboratories had shown remarkable curative effect of methylglyoxal on cancer-bearing animals. In contrast, most of the recent in vitro studies have assigned a toxic role for methylglyoxal. The present study was initiated with the objective to resolve whether methylglyoxal is truly toxic in vivo and to reassess its therapeutic potential. Four species of animals, both rodent and non-rodent, were treated with different doses of methylglyoxal through oral, subcutaneous and intravenous routes. Acute (treatment for only 1 day) toxicity tests had been done with mouse and rat. These animals received 2, 1 and 0.3 g of methylglyoxal/kg of body weight in a day through oral, subcutaneous and intravenous routes respectively. Chronic (treatment for around a month) toxicity test had been done with mouse, rat, rabbit and dog. Mouse, rat and dog received 1, 0.3 and 0.1 g of methylglyoxal/kg of body weight in a day through oral, subcutaneous and intravenous routes respectively. Rabbit received 0.55, 0.3 and 0.1 g of methylglyoxal/kg of body weight in a day through oral, subcutaneous and intravenous routes respectively. It had been observed that methylglyoxal had no deleterious effect on the physical and behavioral pattern of the treated animals. Fertility and teratogenecity studies were done with rats that were subjected to chronic toxicity tests. It had been observed that these animals produced healthy litters indicating no damage of the reproductive systems as well as no deleterious effect on the offspring. Studies on several biochemical and hematological parameters of methylglyoxal-treated rats and dogs and histological studies of several organs of methylglyoxal-treated mouse were performed. These studies indicated that methylglyoxal had no apparent deleterious effect on some vital organs of these animals. A detailed pharmacokinetic study was done with mouse after oral administration of methylglyoxal. The effect of methylglyoxal alone and in combination with creatine and ascorbic acid on cancer-bearing animals had been investigated by measuring the increase in life span and tumor cell growth inhibition. The results indicated that anticancer effect of methylglyoxal was significantly augmented by ascorbic acid and further augmented by ascorbic acid and creatine. Nearly 80% of the animals treated with methylglyoxal plus ascorbic acid plus creatine were completely cured and devoid of any malignant cells within the peritoneal cavity.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/toxicity , Ascorbic Acid/therapeutic use , Carcinoma, Ehrlich Tumor/drug therapy , Creatine/therapeutic use , Pyruvaldehyde/pharmacokinetics , Pyruvaldehyde/toxicity , Vitamins/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Body Weight/drug effects , Carcinoma, Ehrlich Tumor/pathology , Dogs , Drug Synergism , Enzymes/blood , Female , Fertility/drug effects , Longevity/drug effects , Male , Mice , Neoplasm Transplantation , Pyruvaldehyde/therapeutic use , Rabbits , Rats , Reproduction/drug effects , Species Specificity , Survival Analysis , Teratogens/toxicity
17.
Gen Pharmacol ; 27(4): 565-73, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8853285

ABSTRACT

1. Methylglyoxal is a reactive alpha-oxoaldehyde and physiological metabolite formed by the fragmentation of triose-phosphates, and by the metabolism of acetone and aminoacetone. 2. Methylglyoxal modifies guanylate residues to form 6,7-dihydro-6,7-dihydroxy-6-methyl-imidazo[2,3-b]purine-9(8)one and N2-(1-carboxyethyl)guanylate residues and induces apoptosis. 3. Methylglyoxal modifies arginine residues in proteins to form N(delta)-(4,5-dihydroxy-4-methylimidazolidin-2-yl) ornithine, N(delta)-(5-hydro-5-methylimidazol-4-on-2-yl)ornithine and N(delta)-(5)methylimidazol-4-on-2-yl)ornithine residues. 4. Methylglyoxal-modified proteins undergo receptor-mediated endocytosis and lysosomal degradation in monocytes and macrophages, and induce cytokine synthesis and secretion. 5. Methylglyoxal is detoxified by the glyoxalase system. Decreased detoxification of methylglyoxal may be induced pharmacologically by glyoxalase I inhibitors which have anti-tumor and anti-malarial activities. 6. The modification of nucleic acids and protein by methylglyoxal is a signal for their degradation and may have a role in the development of diabetic complications, atherosclerosis, the immune response in starvation, aging and oxidative stress.


Subject(s)
Antineoplastic Agents/pharmacology , Nucleic Acids/drug effects , Proteins/drug effects , Pyruvaldehyde/pharmacology , Animals , Biotransformation , Humans , Neoplasms/drug therapy , Neoplasms/pathology , Pyruvaldehyde/metabolism , Pyruvaldehyde/therapeutic use , Pyruvaldehyde/toxicity
19.
Invest Urol ; 15(6): 493-5, 1978 May.
Article in English | MEDLINE | ID: mdl-649300

ABSTRACT

Male Wistar rats were fed a basal diet, Purina Laboratory Chow, and an oxalate calculi-producing diet (CPD). The CPD was the basal diet containing 3 per cent glycolic acid. Sodium pyruvate, DL-alanine, alpha-keto glutaric acid, thiamine pyrophosphate, and L-glutamic acid were added to the CPD to determine their effectiveness in preventing calculi formation. The effectiveness of methyl glyoxal was determined by adding it to the drinking water. Rats fed CPD for 4 weeks developed calculi in the ureters, bladder, renal tubules, and/or renal pelvis and papilla. Rats in groups fed alanine and/or pyruvate had no calculi in their renal tubules or ureters; additionally, these rats had a significant reduction in incidence and amount of deposits in the renal pelvis and bladder. Rats in groups fed alpha-keto glutaric acid, thiamine pyrophosphate, L-glutamic acid, and methyl glyoxal developed equally or more severe oxalate urolithiasis than those on CPD alone. Results of this study show that either pyruvate or alanine at appropriate levels may be beneficial in preventing oxalate urolith formation.


Subject(s)
Alanine/therapeutic use , Oxalates/metabolism , Pyruvates/therapeutic use , Urinary Calculi/prevention & control , Animals , Drug Evaluation, Preclinical , Glutamates/therapeutic use , Glutarates/therapeutic use , Male , Pyruvaldehyde/therapeutic use , Rats , Thiamine Pyrophosphate/therapeutic use
20.
Proc Natl Acad Sci U S A ; 74(7): 2844-7, 1977 Jul.
Article in English | MEDLINE | ID: mdl-268635

ABSTRACT

Complex living structures developed on our globe after the appearance of light and oxygen. In functions of these structures, solid state phenomena play a major role. The structural proteins were made into radicals by doping, the covalent incorporation of electron acceptors. This lent mobility to their electrons and a subtle reactivity to their molecules. Cancer is unable to go into the radical state.


Subject(s)
Neoplasms/metabolism , Aldehydes , Caseins , Cell Division/drug effects , Chemical Phenomena , Chemistry , Chemistry, Physical , Free Radicals , Ketones , Lactoylglutathione Lyase/metabolism , Light , Methylamines , Neoplasms/drug therapy , Oxygen , Proteins , Pyruvaldehyde/metabolism , Pyruvaldehyde/pharmacology , Pyruvaldehyde/therapeutic use , Schiff Bases , Sulfhydryl Compounds
SELECTION OF CITATIONS
SEARCH DETAIL
...