Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 265
Filtrar
1.
Neurotox Res ; 42(2): 19, 2024 Feb 29.
Artigo em Inglês | MEDLINE | ID: mdl-38421481

RESUMO

Maternal hyperhomocysteinemia (HCY) induced by genetic defects in methionine cycle enzymes or vitamin imbalance is known to be a pathologic factor that can impair embryonal brain development and cause long-term consequences in the postnatal brain development as well as changes in the expression of neuronal genes. Studies of the gene expression on this model requires the selection of optimal housekeeping genes. This work aimed to analyze the expression stability of housekeeping genes in offspring brain. Pregnant female Wistar rats were treated daily with a 0.15% L-methionine solution in the period starting on the 4th day of pregnancy until delivery, to cause the increase in the homocysteine level in fetus blood and brain. Housekeeping gene expression was assessed by RT-qPCR on whole embryonic brain and selected rat brain areas at P20 and P90. The amplification curves were analyzed, and raw means Cq data were imported to the RefFinder online tool to assess the reference genes stability. Most of the analyzed genes showed high stability of mRNA expression in the fetal brain at both periods of analysis (E14 and E20). However, the most stably expressed genes at different age points differed. Actb, Ppia, Rpl13a are the most stably expressed on E14, Ywhaz, Pgk1, Hprt1 - on E20 and P20, Hprt1, Actb, and Pgk1 - on P90. Gapdh gene used as a reference in various studies demonstrates high stability only in the hippocampus and cannot be recommended as the optimal reference gene on HCY model. Hprt1 and Pgk1 genes were found to be the most stably expressed in the brain of rat subjected to HCY. These two genes showed high stability in the brain on E20 and in various areas of the brain on the P20 and P90. On E14, the preferred genes for normalization are Actb, Ppia, Rpl13a.


Assuntos
Hiper-Homocisteinemia , Feminino , Gravidez , Ratos , Animais , Hiper-Homocisteinemia/induzido quimicamente , Hiper-Homocisteinemia/genética , Ratos Wistar , Encéfalo , Metionina , Racemetionina , Hipoxantina Fosforribosiltransferase
2.
Int J Mol Sci ; 25(4)2024 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-38396625

RESUMO

The aim of this study was to investigate the effects of aerobic treadmill training regimen of four weeks duration on oxidative stress parameters, metabolic enzymes, and histomorphometric changes in the colon of hyperhomocysteinemic rats. Male Wistar albino rats were divided into four groups (n = 10, per group): C, 0.9% NaCl 0.2 mL/day subcutaneous injection (s.c.) 2x/day; H, homocysteine 0.45 µmol/g b.w./day s.c. 2x/day; CPA, saline (0.9% NaCl 0.2 mL/day s.c. 2x/day) and an aerobic treadmill training program; and HPA, homocysteine (0.45 µmol/g b.w./day s.c. 2x/day) and an aerobic treadmill training program. The HPA group had an increased level of malondialdehyde (5.568 ± 0.872 µmol/mg protein, p = 0.0128 vs. CPA (3.080 ± 0.887 µmol/mg protein)), catalase activity (3.195 ± 0.533 U/mg protein, p < 0.0001 vs. C (1.467 ± 0.501 U/mg protein), p = 0.0012 vs. H (1.955 ± 0.293 U/mg protein), and p = 0.0003 vs. CPA (1.789 ± 0.256 U/mg protein)), and total superoxide dismutase activity (9.857 ± 1.566 U/mg protein, p < 0.0001 vs. C (6.738 ± 0.339 U/mg protein), p < 0.0001 vs. H (6.015 ± 0.424 U/mg protein), and p < 0.0001 vs. CPA (5.172 ± 0.284 U/mg protein)) were detected in the rat colon. In the HPA group, higher activities of lactate dehydrogenase (2.675 ± 1.364 mU/mg protein) were detected in comparison to the CPA group (1.198 ± 0.217 mU/mg protein, p = 0.0234) and higher activities of malate dehydrogenase (9.962 (5.752-10.220) mU/mg protein) were detected in comparison to the CPA group (4.727 (4.562-5.299) mU/mg protein, p = 0.0385). Subchronic treadmill training in the rats with hyperhomocysteinemia triggers the colon tissue antioxidant response (by increasing the activities of superoxide dismutase and catalase) and elicits an increase in metabolic enzyme activities (lactate dehydrogenase and malate dehydrogenase). This study offers a comprehensive assessment of the effects of aerobic exercise on colonic tissues in a rat model of hyperhomocysteinemia, evaluating a range of biological indicators including antioxidant enzyme activity, metabolic enzyme activity, and morphometric parameters, which suggested that exercise may confer protective effects at both the physiological and morphological levels.


Assuntos
Antioxidantes , Hiper-Homocisteinemia , Ratos , Masculino , Animais , Catalase/metabolismo , Antioxidantes/farmacologia , Ratos Wistar , Malato Desidrogenase/metabolismo , Hiper-Homocisteinemia/induzido quimicamente , Hiper-Homocisteinemia/metabolismo , Solução Salina , Estresse Oxidativo , Superóxido Dismutase/metabolismo , Homocisteína/metabolismo , Colo/metabolismo
3.
Anal Chem ; 95(26): 9872-9880, 2023 07 04.
Artigo em Inglês | MEDLINE | ID: mdl-37344960

RESUMO

Elevated plasma level of homocysteine, also termed as hyperhomocysteinemia, is acknowledged as a significant and independent risk factor of Alzheimer's disease. However, the mechanistic insight has not been thoroughly elucidated yet. In this work, 3,5-dihydroxybenzyloxy was explored as the unique reaction trigger and integrated into the naphthalimide fluorophore via a carbamate linker to afford a new probe for •OH imaging. •OH treatment induced aromatic hydroxylation and subsequent elimination reaction to release the caged fluorophore, accompanied with a highly specific and sensitive turn-on fluorescence response. Cell imaging results revealed that excess homocysteine triggered overwhelming •OH production, which was mediated by N-methyl-d-aspartate receptor and NADPH oxidase, and the resultant •OH stress further initiated neuronal ferroptosis, also confirmed by western blot analyses. Additionally, hyperhomocysteinemic mouse models were established, and Alzheimer-like dementia of the mice was observed from behavioral tests. Most importantly, with this probe, cerebral •OH fluctuation was in situ visualized in live mice, which positively correlated with the severity of Alzheimer-like dementia induced by hyperhomocysteinemia. These results reveal that cerebral •OH stress may be the critical nexus linking hyperhomocysteinemia and Alzheimer's disease. This work provides a robust fluorescence probe for in situ visualizing the cerebral •OH fluctuations and illuminating critical insights into •OH contributions in brain disorders.


Assuntos
Doença de Alzheimer , Hiper-Homocisteinemia , Camundongos , Animais , Doença de Alzheimer/diagnóstico por imagem , Doença de Alzheimer/complicações , Hiper-Homocisteinemia/complicações , Hiper-Homocisteinemia/induzido quimicamente , Radical Hidroxila , Fatores de Risco , Imagem Óptica , Homocisteína
4.
Biochim Biophys Acta Mol Basis Dis ; 1869(4): 166643, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36669577

RESUMO

OBJECTIVES: Studies in certain cardiac hypertrophy models suggested the individual role of soluble epoxide hydrolase (sEH) and canonical transient receptor potential 3 (TRPC3) channels, however, whether they jointly mediate hypertrophic process remains unexplored. Hyperhomocysteinemia promotes cardiac hypertrophy while the involvement of sEH and TRPC3 channels remains unknown. This study aimed to explore the role of, and interrelation between sEH and TRPC3 channels in homocysteine-induced cardiac hypertrophy. METHODS: Rats were fed methionine-enriched diet to induce hyperhomocysteinemia. H9c2 cells and neonatal rat cardiomyocytes were incubated with homocysteine. Cardiac hypertrophy was evaluated by echocardiography, histological examination, immunofluorescence imaging, and expressions of hypertrophic markers. Epoxyeicosatrienoic acids (EETs) were determined by ELISA. TRPC3 current was recorded by patch-clamp. Gene promotor activity was measured using dual-luciferase reporter assay. RESULTS: Inhibition of sEH by 1-trifluoromethoxyphenyl-3-(1-propionylpiperidin-4-yl) urea (TPPU) reduced ventricular mass, lowered the expression of hypertrophic markers, decreased interstitial collagen deposition, and improved cardiac function in hyperhomocysteinemic rats, associated with restoration of EETs levels in myocardium. TPPU or knockdown of sEH suppressed TRPC3 transcription and translation as well as TRPC3 current that were enhanced by homocysteine. Exogenous 11,12-EET inhibited homocysteine-induced TRPC3 expression and cellular hypertrophy. Silencing C/EBPß attenuated, while overexpressing C/EBPß promoted homocysteine-induced hypertrophy and expressions of sEH and TRPC3, resulting respectively from inhibition or activation of sEH and TRPC3 gene promoters. CONCLUSIONS: sEH and TRPC3 channels jointly contribute to homocysteine-induced cardiac hypertrophy. Homocysteine transcriptionally activates sEH and TRPC3 genes through a common regulatory element C/EBPß. sEH activation leads to an upregulation of TRPC3 channels via a 11,12-EET-dependent manner.


Assuntos
Cardiomegalia , Epóxido Hidrolases , Hiper-Homocisteinemia , Animais , Ratos , Cardiomegalia/induzido quimicamente , Cardiomegalia/genética , Cardiomegalia/metabolismo , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Eicosanoides , Epóxido Hidrolases/genética , Epóxido Hidrolases/metabolismo , Hiper-Homocisteinemia/induzido quimicamente , Hiper-Homocisteinemia/complicações , Miocárdio/metabolismo , Canais de Cátion TRPC/genética , Canais de Cátion TRPC/metabolismo
5.
Redox Biol ; 58: 102540, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36399957

RESUMO

BACKGROUND: s: Hyperhomocysteinemia (HHcy) is one of risk factors for vascular cognitive impairment (VCI). GTP cyclohydrolase 1 (GCH1) deficiency is critical to oxidative stress in vascular dysfunction. The aim of this study was designed to examine whether HHcy induces VCI through GCH1 S-nitrosylation, a redox-related post-translational modification of cysteine. METHODS: The VCI model was induced by feeding mice homocysteine thiolactone (HTL) for 16 consecutive weeks. The cognitive functions were evaluated by step-down avoidance test, passive avoidance step-through task test, and Morris water maze (MWM) test. Protein S-nitrosylation was assayed using a biotin-switch method. RESULTS: In cell-free system, nitric oxide (NO) donor induced GCH1 protein S-nitrosylation and decreased GCH1 activity. In endothelial cells, HTL increased GCH1 S-nitrosylation, reduced tetrahydrobiopterin, and induced oxidative stress, which were attenuated by N-acetyl-cysteine, L-N6-1-Iminoethyl-lysine, mutant of GCH1 cysteine 141 to alanine (MT-GCH1) or gene deletion of inducible NO synthase (iNOS). Further, HTL incubation or iNOS overexpression promoted endothelial cellular senescence, but abolished by exogenous expression of MT-GCH1 or pharmacological approaches including N-acetyl-cysteine, L-sepiapterin, and tempol. In wildtype mice, long-term administration of HTL induced GCH1 S-nitrosylation and vascular stiffness, decreased cerebral blood flow, and damaged the cognitive functions. However, these abnormalities induced by HTL administration were rescued by enforced expression of MT-GCH1 or gene knockout of iNOS. In human subjects, GCH1 S-nitrosylation was increased and cognitive functions were impaired in patients with HHcy. CONCLUSION: The iNOS-mediated nitrosative stress induced by HTL drives GCH1 S-nitrosylation to induce cerebral vascular stiffness and cognitive impairments.


Assuntos
Disfunção Cognitiva , Hiper-Homocisteinemia , Animais , Humanos , Camundongos , Disfunção Cognitiva/etiologia , Disfunção Cognitiva/metabolismo , Cisteína/metabolismo , Células Endoteliais/metabolismo , GTP Cicloidrolase , Hiper-Homocisteinemia/induzido quimicamente , Hiper-Homocisteinemia/metabolismo , Óxido Nítrico/metabolismo , Estresse Nitrosativo
6.
Neurotox Res ; 40(6): 2103-2116, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36394770

RESUMO

Brain injury and cognitive impairment are major health issues associated with neurodegenerative diseases in young and aged persons worldwide. Epigallocatechin-3-gallate (EGCG) was studied for its ability to protect against methionine (Met)-induced brain damage and cognitive dysfunction. Male mice were given Met-supplemented in drinking water to produce hyperhomocysteinemia (HHcy)-induced animals. EGCG was administered daily concurrently with Met by gavage. EGCG attenuated the rise in homocysteine levels in the plasma and the formation of amyloid-ß and tau protein in the brain. Cognitive and memory impairment in HHcy-induced mice were significantly improved by EGCG administration. These results were associated with improvement in glutamate and gamma-aminobutyric acid levels in the brain. EGCG maintained the levels of glutathione and the activity of antioxidant enzymes in the brain. As a result of the reduction of oxidative stress, EGCG protected against DNA damage in Met-treated mice. Moreover, maintaining the redox balance significantly ameliorated neuroinflammation evidenced by the normalization of IL-1ß, IL-6, tumor necrosis factor α, C-reactive protein, and IL-13 in the same animals. The decreases in both oxidative stress and inflammatory cytokines were significantly associated with upregulation of the antiapoptotic Bcl-2 protein and downregulation of the proapoptotic protein Bax, caspases 3 and 9, and p53 compared with Met-treated animals, indicating a diminution of neuronal apoptosis. These effects reflect and explain the improvement in histopathological alterations in the hippocampus of Met-treated mice. In conclusion, the beneficial effects of EGCG may be due to interconnecting pathways, including modulation of redox balance, amelioration of inflammation, and regulation of antiapoptotic proteins.


Assuntos
Lesões Encefálicas , Catequina , Hiper-Homocisteinemia , Camundongos , Masculino , Animais , Metionina/farmacologia , Hiper-Homocisteinemia/induzido quimicamente , Hiper-Homocisteinemia/complicações , Hiper-Homocisteinemia/tratamento farmacológico , Estresse Oxidativo , Cognição , Proteínas Reguladoras de Apoptose , Catequina/farmacologia , Catequina/uso terapêutico , Racemetionina/farmacologia
7.
Neurotox Res ; 40(5): 1261-1271, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35920995

RESUMO

Acetazolamide (AZA) is a carbonic anhydrase inhibitor (CAI) with neuroprotective effects. Hyperhomocysteinemia is associated with blood-brain-barrier (BBB) disruption in brain disorders. A previous study indicated that AZA might have a new role in brain disorders. However, its function in hyperhomocysteinemia-related BBB disruption has not been reported. Here, we aim to clarify the role of AZA in homocysteine (Hcy)-mediated BBB dysfunction using both in vivo and in vitro assays. We found that AZA improved memory and cognitive function, and reduced brain edema in Hcy-stimulated hyperhomocysteinemia model rats. This protective effect of AZA on hyperhomocysteinemia rats was accompanied by improved BBB permeability and increased expression levels of the tight junction proteins, occludin, and claudin-5. The in vitro assay results show that AZA prevented Hcy-induced cell injury and attenuated the increased permeability in Hcy-treated bEnd.3 brain endothelial cells. The Hcy-induced decrease in occludin and claudin-5, and increase in MMP-2 and MMP-9 expression levels were attenuated by AZA in bEnd.3 cells. Moreover, the Hcy-induced downregulation of the Wnt/ß-catenin signaling pathway in bEnd.3 cells was abolished by AZA. Inhibition of Wnt/ß-catenin by ICG-001 reversed the protective effects of AZA in Hcy-treated bEnd.3 cells. We also prove that this process is mediated by WTAP. These findings suggest that acetazolamide mitigated the Hcy-induced compromised brain vascular endothelial integrity by regulating the activation of the Wnt/ß-catenin signaling pathway.


Assuntos
Encefalopatias , Hiper-Homocisteinemia , Fármacos Neuroprotetores , Acetazolamida/metabolismo , Acetazolamida/farmacologia , Animais , Barreira Hematoencefálica , Encefalopatias/metabolismo , Inibidores da Anidrase Carbônica/metabolismo , Inibidores da Anidrase Carbônica/farmacologia , Claudina-5/metabolismo , Claudina-5/farmacologia , Células Endoteliais/metabolismo , Homocisteína/metabolismo , Hiper-Homocisteinemia/induzido quimicamente , Hiper-Homocisteinemia/tratamento farmacológico , Hiper-Homocisteinemia/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Fármacos Neuroprotetores/metabolismo , Fármacos Neuroprotetores/farmacologia , Ocludina/metabolismo , Ocludina/farmacologia , Ratos , Via de Sinalização Wnt/fisiologia , beta Catenina/metabolismo , beta Catenina/farmacologia
8.
Am J Med Sci ; 364(6): 746-751, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35896125

RESUMO

BACKGROUND: Previous studies have demonstrated an association between proton pump inhibitors (PPI) use and vitamin B12 deficiency. However, data regarding PPI use and elevated serum homocysteine level, an important marker of vitamin B12 deficiency, are scant. METHODS: Data were collected from medical records of subjects examined at a screening center in Israel. Cross sectional analysis was conducted on 25,953 subjects. Levels of vitamin B12 and homocysteine were compared between subjects who consumed PPI medications and those who did not. RESULTS: The mean age of the study population was 45 years and 33% were females. Subjects who received PPI medications had a minor higher vitamin B12 levels (320 pmol/L vs 300 pmol/L, p=0.024). Levels of vitamin B12 remained higher in females receiving PPI medications after performing a stratified analysis according to subjects' gender. Homocysteine levels were higher in subjects receiving PPI medications as compared to those who did not (12.0 µmol/L vs 11.6 0 µmol/L, p<0.001). Levels remained higher in female subjects after performing a stratified analysis according to subjects' sex. There was no statistically significant difference in the prevalence of vitamin B12 deficiency (according to two cutoffs: vitamin B12≤200 or ≤140 pmol/L) as well as the prevalence of hyperhomocysteinemia (defined as homocysteine >15.0 µmol/L) between the two groups. CONCLUSIONS: According to our study, no association was found between PPI medication use and vitamin B12 deficiency or hyperhomocysteinemia. Patients receiving PPI medications had slightly higher levels of vitamin B12 and homocysteine, however these differences were too small to have any clinical relevance.


Assuntos
Hiper-Homocisteinemia , Deficiência de Vitamina B 12 , Masculino , Humanos , Feminino , Pessoa de Meia-Idade , Vitamina B 12 , Estudos Transversais , Hiper-Homocisteinemia/induzido quimicamente , Hiper-Homocisteinemia/epidemiologia , Inibidores da Bomba de Prótons/efeitos adversos , Deficiência de Vitamina B 12/induzido quimicamente , Deficiência de Vitamina B 12/epidemiologia , Homocisteína , Ácido Fólico
9.
Folia Histochem Cytobiol ; 60(2): 111-124, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35603572

RESUMO

INTRODUCTION: Cardiovascular diseases were defined as coronary artery, cerebrovascular, or peripheral arterial disease. Hyperhomocysteinemia (Hhcy) is an independent risk factor of cardiovascular diseases, including atherosclerosis. Our previous studies demonstrated the involvement of Hhcy in cardiovascular remodeling in the sand rat Psammomys obesus. MATERIAL AND METHODS: An experimental Hhcy was induced, in the sand rat Psammomys obesus, by a daily intraperitoneal injection of 70 mg/kg of methionine for a total duration of 6 months. The impact of Hhcy on the cellular and matrix structures of the heart, aorta and liver was analyzed using histological techniques. Additionally we treatedprimary cultures of aortic smooth muscle cells (SMCs) with high concentration of methionine to investigate the effects of methionine at the cellular level. RESULTS: A moderate Hhcy induced a significant increase in the extracellular matrix components particularly collagens which accumulated in the interstitial and perivascular spaces in the studied organs indicating a developing fibrosis. A liver steatosis was also observed following methionine treatment. Further analysis of the aorta showed that Hhcy also induced vascular alterations including SMCs reorientation and proliferation associated with aneurysm formation. CONCLUSIONS: Our results show for the first time that Hhcy can induce a cardiovascular and liver diseases phenotype in Psammomys obesus, a species previously shown to be a good model for the studies of diabetes and other metabolism-related pathologies.


Assuntos
Doenças Cardiovasculares , Hiper-Homocisteinemia , Animais , Doenças Cardiovasculares/induzido quimicamente , Doenças Cardiovasculares/complicações , Gerbillinae , Hiper-Homocisteinemia/induzido quimicamente , Hiper-Homocisteinemia/complicações , Hiper-Homocisteinemia/patologia , Metionina , Fenótipo
10.
Biomolecules ; 12(5)2022 05 23.
Artigo em Inglês | MEDLINE | ID: mdl-35625662

RESUMO

Homocysteine is a sulfur-containing endogenous amino acid leading to neurotoxic effects at high concentrations. Population studies suggest an association between plasma homocysteine levels and the risk of migraine headaches. The aim of this study was to analyze the sensitivity of rats with prenatal hyperhomocysteinemia (hHCY) in respect of the development of behavioral correlates of headache and spreading cortical depolarization (CSD) in a migraine model induced by the administration of the nitric oxide (NO) donor nitroglycerin. Animals with hHCY were characterized by migraine-related symptoms such as mechanical hyperalgesia, high-level anxiety, photophobia, as well as an enhanced level of neuronal activity in the somatosensory cortex along with a lower threshold of CSD generation. Likewise, acute or chronic intermittent administration of nitroglycerin also induced the development of mechanical allodynia, photophobia and anxiety in control groups. However, these symptoms were more pronounced in rats with hHCY. Unlike hHCY, nitroglycerin administration did not affect the threshold of CSD generation, but like hHCY, increased the background neuronal activity in layers 2/3 and 4 of the cerebral cortex. The latter was more pronounced in animals with hHCY. Thus, the migraine profile associated with hHCY can be further exaggerated in conditions with enhanced levels of migraine triggering the gaseous transmitter NO. Our data are consistent with the view that high levels of plasma homocysteine can act as a risk factor for the development of migraine.


Assuntos
Excitabilidade Cortical , Hiper-Homocisteinemia , Transtornos de Enxaqueca , Animais , Ansiedade , Feminino , Homocisteína , Hiperalgesia/induzido quimicamente , Hiper-Homocisteinemia/induzido quimicamente , Hiper-Homocisteinemia/complicações , Hiper-Homocisteinemia/metabolismo , Transtornos de Enxaqueca/induzido quimicamente , Nitroglicerina/toxicidade , Fotofobia , Gravidez , Ratos
11.
Wiad Lek ; 75(4 pt 1): 831-835, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35633356

RESUMO

OBJECTIVE: The aim: To investigate the distribution of proteolytic activity and cytokine profile in the heart of rats with hyperhomocysteinemia. PATIENTS AND METHODS: Materials and methods: A total of 60 albino non-linear male rats was used in the study. Hyperhomocysteinemia was induced by intragastric administration of DL-homocysteine thiolactone. Total proteolytic activity was measured using casein as a substrate. To determine the activity of metal-dependent and serine proteases, ethylenediaminetetraacetic acid, and phenylmethylsulfonyl fluoride were used. The level of matrix metalloproteinases, tissue inhibitor of metalloproteinases-1, and cytokines was studied by enzyme-linked immunosorbent assay. RESULTS: Results: It was found an increase in the total proteolytic activity in the heart of young, adult, and old animals. In addition, the redistribution of proteolytic activity was revealed - the portion of metal-dependent enzymes increased in all groups while the percentage of serine proteases decreased in the old animals with hyperhomocysteinemia. The state of mild inflammation, evidenced by the increased level of some pro-inflammatory cytokines, was found in the heart of young and old animals with hyperhomocysteinemia. CONCLUSION: Conclusions: The pathogenesis of hyperhomocysteinemia is accompanied by a change in the proteolytic activity in the heart as well as a change in the cytokine profile.


Assuntos
Citocinas , Coração , Hiper-Homocisteinemia , Serina Proteases , Animais , Citocinas/metabolismo , Hiper-Homocisteinemia/induzido quimicamente , Hiper-Homocisteinemia/metabolismo , Masculino , Ratos , Serina Proteases/metabolismo
12.
Mol Neurobiol ; 59(7): 4517-4534, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35578101

RESUMO

Elevated levels of homocysteine (Hcy) in the blood, called hyperhomocysteinemia (HHcy), is a prevalent risk factor for it has been shown that Hcy induces oxidative stress and increases microglial activation and neuroinflammation, as well as causes cognitive impairment, which have been linked to the neurodegenerative process. This study aimed to evaluate the effect of mild hyperhomocysteinemia with or without ibuprofen and rivastigmine treatments on the behavior and neurochemical parameters in male rats. The chronic mild HHcy model was chemically induced in Wistar rats by subcutaneous administration of Hcy (4055 mg/kg body weight) twice daily for 30 days. Ibuprofen (40 mg/kg) and rivastigmine (0.5 mg/kg) were administered intraperitoneally once daily. Motor damage (open field, balance beam, rotarod, and vertical pole test), cognitive deficits (Y-maze), neurochemical parameters (oxidative status/antioxidant enzymatic defenses, presynaptic protein synapsin 1, inflammatory profile parameters, calcium binding adapter molecule 1 (Iba1), iNOS gene expression), and cholinergic anti-inflammatory pathway were investigated. Results showed that mild HHcy caused cognitive deficits in working memory, and impaired motor coordination reduced the amount of synapsin 1 protein, altered the neuroinflammatory picture, and caused changes in the activity of catalase and acetylcholinesterase enzymes. Both rivastigmine and ibuprofen treatments were able to mitigate this damage caused by mild HHcy. Together, these neurochemical changes may be associated with the mechanisms by which Hcy has been linked to a risk factor for AD. Treatments with rivastigmine and ibuprofen can effectively reduce the damage caused by increased Hcy levels.


Assuntos
Hiper-Homocisteinemia , Acetilcolinesterase/metabolismo , Animais , Homocisteína , Hiper-Homocisteinemia/induzido quimicamente , Hiper-Homocisteinemia/complicações , Hiper-Homocisteinemia/tratamento farmacológico , Ibuprofeno , Inflamação/complicações , Inflamação/tratamento farmacológico , Masculino , Estresse Oxidativo/fisiologia , Ratos , Ratos Wistar , Rivastigmina/farmacologia , Rivastigmina/uso terapêutico , Sinapsinas/metabolismo
13.
Dent Med Probl ; 59(2): 255-261, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35313094

RESUMO

BACKGROUND: Periodontal disease is the second most common oral health problem after dental caries. This increasing prevalence makes it not only a health problem, but also a social issue. The pathogenesis of periodontal disease is associated with a number of adverse exogenous and endogenous factors, including hyperhomocysteinemia (HHcy). OBJECTIVES: This study aimed to determine the features of bone metabolism in rats with lipopolysaccharide (LPS)-induced periodontitis combined with chronic thiolactone HHcy. MATERIAL AND METHODS: Forty-eight white, non-linear, mature rats were divided into 4 groups: control (n = 12); LPS­induced periodontitis (n = 12); chronic thiolactone HHcy (n = 12); and periodontitis combined with HHcy (n = 12). The rats were sacrificed the day after the last LPS injection or the day after the last homocysteine (Hcy) thiolactone administration. Bone metabolism was determined based on the activity of alkaline phosphatase (ALP) and acid phosphatase (AP) in blood serum and periodontal homogenate. RESULTS: A decrease in ALP activity (by 40.1%; р = 0.001) and the mineralization index (MI) (3.5 times; р < 0.001) with an increase in AP activity (2.0 times; р < 0.001) was observed in the periodontal homogenate of rats with LPS­induced periodontitis. In the case of LPS­induced periodontitis combined with chronic thiolactone HHcy, more pronounced changes in the activity of phosphatases and in MI were established as compared to rats with LPS­induced periodontitis only. CONCLUSIONS: Chronic thiolactone HHcy enhances disturbances in bone metabolism in LPS­induced periodontitis. The osteotoxic effect of HHcy is associated with the activation of osteoclastogenesis and enhanced bone resorption. However, further research is required on the subject.


Assuntos
Cárie Dentária , Hiper-Homocisteinemia , Periodontite , Animais , Hiper-Homocisteinemia/induzido quimicamente , Hiper-Homocisteinemia/complicações , Hiper-Homocisteinemia/metabolismo , Lipopolissacarídeos/efeitos adversos , Ratos
14.
Mol Cell Biochem ; 477(5): 1621-1628, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35220548

RESUMO

Increased concentration of plasma homocysteine (Hcy) is an independent risk factor of cardiovascular disease, yet the mechanism by which hyperhomocysteinemia (HHcy) causes cardiac dysfunction is largely unknown. The aim of present study was to investigate the contribution of sarcoplasmic reticulum to impaired cardiac contractile function in HHCy. HHcy-induced by subcutaneous injection of Hcy (0.45 µmol/g of body weight) twice a day for a period of 2 weeks resulted in significant decrease in developed left ventricular pressure and maximum rate of ventricular relaxation. Our results show that abundances of SR Ca2+-handling proteins, Ca2+-ATPase (SERCA2), calsequestrin and histidine-rich calcium-binding protein are significantly reduced while the content of phospholamban is unchanged. Moreover, we found that increased PLN:SERCA2 ratio results in the inhibition of SERCA2 activity at low free Ca2+ concentrations. We further discovered that HHcy is not associated with increased oxidative stress in SR. Taken together, these findings suggest that disturbances in SR Ca2+ handling, caused by altered protein contents but not oxidative damage, may contribute to impaired cardiac contractility in HHcy.


Assuntos
Hiper-Homocisteinemia , Retículo Sarcoplasmático , Animais , Cálcio/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Calsequestrina/metabolismo , Coração/fisiologia , Hiper-Homocisteinemia/induzido quimicamente , Contração Miocárdica , Miocárdio/metabolismo , Ratos , Retículo Sarcoplasmático/metabolismo , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático
15.
Int J Neurosci ; 132(4): 384-396, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32859137

RESUMO

AIM: The present study investigates the potential of Tadalafil, a phosphodiesterase-5 inhibitor, in a rat model of hyperhomocysteinemia induced vascular dementia. METHODS: Hyperhomocysteinemia induced vascular dementia in Wistar rats was produced by administering l-Methionine (1.7 g/kg/day; p.o.×8 weeks). Learning and memory was assessed by employing Morris water maze (MWM) test. Endothelial dysfunction was assessed through acetylcholine-induced endothelial-dependent vasorelaxation and serum nitrite levels. Various other biochemical and histopathological estimations were also performed. RESULTS: l-Methionine produced significant impairment in acetylcholine-induced endothelium-dependent vasorelaxation and a decrease in serum nitrite levels indicating endothelial dysfunction. Further, these animals performed poorly on Morris water maze, depicting impairment of learning and memory. There was a significant rise in brain oxidative stress level (indicated by an increase in brain thiobarbituric acid reactive species and a decrease in reduced glutathione levels). Increase in brain acetylcholinesterase activity; brain myeloperoxidase activity and brain neutrophil infiltration (a marker of inflammation) were also observed. Tadalafil (5 and 10 mg/kg, p.o.)/Donepezil (0.5 mg/kg, i.p., serving as standard) treatment ameliorated l-Methionine induced endothelial dysfunction; memory deficits; biochemical and histopathological changes in a significant manner. CONCLUSIONS: It may be concluded that tadalafil has shown efficacy in the rat model of l-Methionine induced vascular dementia and that phosphodiesterase-5 can be considered as an important therapeutic target for the treatment of vascular dementia.


Assuntos
Demência Vascular , Hiper-Homocisteinemia , Tadalafila , Acetilcolina , Acetilcolinesterase/metabolismo , Animais , Encéfalo/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 5 , Demência Vascular/tratamento farmacológico , Demência Vascular/etiologia , Hiper-Homocisteinemia/induzido quimicamente , Hiper-Homocisteinemia/complicações , Hiper-Homocisteinemia/tratamento farmacológico , Aprendizagem em Labirinto , Transtornos da Memória/induzido quimicamente , Transtornos da Memória/tratamento farmacológico , Metionina , Nitritos/sangue , Estresse Oxidativo , Inibidores da Fosfodiesterase 5 , Ratos , Ratos Wistar , Tadalafila/uso terapêutico
16.
Neurochem Res ; 47(3): 762-767, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-34787820

RESUMO

Accumulated evidence demonstrated that an elevated plasma homocysteine level, hyperhomocysteinemia, induced cognitive impairment in animals, elderly and the patients with neurodegenerative diseases. To date, the underlying cellular and molecular mechanisms by which hyperhomocysteinemia induces cognitive impairment has not been clearly defined. The purpose of this study was to investigate the possible cellular and molecular mechanisms behind hyperhomocysteinemia signaling in rat memory impairment. The results from this study demonstrated that hyperhomocysteinemia induced neuronal damage and loss in hippocampal CA3 region and downregulated the cAMP response element-binding protein (CREB) phosphorylation. The findings of this study provide evidence that hyperhomocysteinemia induces rat memory impairment via injuring hippocampal CA3 neurons and downregulating CREB phosphorylation.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico , Hiper-Homocisteinemia , Transtornos da Memória , Animais , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Hipocampo/metabolismo , Hiper-Homocisteinemia/induzido quimicamente , Hiper-Homocisteinemia/complicações , Neurônios/metabolismo , Fosforilação , Ratos
17.
J Clin Neurosci ; 88: 226-231, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33992189

RESUMO

INTRODUCTION: Previous studies have suggested a significant increase in plasma homocysteine (Hcy) levels in levodopa-treated Parkinson's disease (PD) patients, and vitamin B12 and folate supplementation may decrease Hcy levels. However, the effects of catechol-O-methyltransferase inhibitors on levodopa-induced increase in Hcy levels were conflicting. The aim of this study was to evaluate whether Hcy levels are increased in levodopa-treated PD patients and to evaluate the effects of vitamin B12 and folate or entacapone on Hcy levels in levodopa-treated PD patients. METHODS: We analyzed and compared plasma Hcy levels in 20 levodopa-naïve PD patients and 42 levodopa-treated PD patients, followed by randomized assignment of 42 levodopa-treated patients to treatment groups with either vitamin B12 and folate, entacapone, or no medication. RESULTS: Plasma Hcy levels in levodopa-treated PD patients were higher than those in the control group, but the difference was not statistical significant (15.25 ± 6.70 and 13.13 ± 4.68, P = 0.216). Patients treated with vitamin B12 and folate had a significant decrease in plasma Hcy levels (P < 0.001). In the entacapone group, Hcy levels were mildly decreased, but the change did not reach statistical significance. CONCLUSION: Levodopa-treated PD patients had higher plasma Hcy than levodopa-naive PD patients. Unlike entacapone, combination supplementation with vitamin B12 and folate was associated with significantly decreased plasma Hcy. We suggest that plasma Hcy levels should be monitored during levodopa treatment, and supplementation with inexpensive vitamin B12 and folate is beneficial for levodopa-treated patients.


Assuntos
Antiparkinsonianos/uso terapêutico , Catecóis/uso terapêutico , Homocisteína/sangue , Nitrilas/uso terapêutico , Doença de Parkinson/sangue , Doença de Parkinson/tratamento farmacológico , Idoso , Feminino , Ácido Fólico/uso terapêutico , Homocisteína/efeitos dos fármacos , Humanos , Hiper-Homocisteinemia/induzido quimicamente , Hiper-Homocisteinemia/prevenção & controle , Levodopa/uso terapêutico , Masculino , Pessoa de Meia-Idade , Projetos de Pesquisa , Vitamina B 12/uso terapêutico
18.
Behav Brain Res ; 409: 113324, 2021 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-33915239

RESUMO

Epidemiological data suggest that elevated homocysteine is associated with migraine with aura. However, how homocysteine contributes to migraine is still unclear. Here, we tested whether hyperhomocysteinemia (hHCY) promotes cortical spreading depression (CSD), a phenomenon underlying migraine with aura, and whether hHCY contributes to pain behavior. hHCY was induced by dietary methionine in female rats while the testing was performed on their 6-8week-old offspring. CSD and multiple unit activity (MUA) induced by KCl were recorded from the primary somatosensory cortex, S1, using multichannel electrodes. In hHCY rats, compared to control, we found: i) higher probability of CSD occurrence; ii) induction of CSD by lower concentrations of KCl; iii) faster horizontal propagation of CSD; iv) smaller CSD with longer duration; v) higher frequency of MUA at CSD onset along with slower reappearance. Rats with hHCY demonstrated high level of locomotor activity and grooming while spent less time in the central area of the open field, indicating anxiety. These animals showed light sensitivity and facial mechanical allodinia. Thus, hHCY acquired at birth promotes multiple features of migraine such as higher cortical excitability, mechanical allodynia, photophobia, and anxiety. Our results provide the first experimental explanation for the higher occurrence of migraine with aura in patients with hHCY.


Assuntos
Ansiedade/fisiopatologia , Comportamento Animal/fisiologia , Excitabilidade Cortical/fisiologia , Depressão Alastrante da Atividade Elétrica Cortical/fisiologia , Hiperalgesia/fisiopatologia , Hiper-Homocisteinemia/complicações , Fotofobia/fisiopatologia , Córtex Somatossensorial/fisiopatologia , Animais , Ansiedade/etiologia , Depressão Alastrante da Atividade Elétrica Cortical/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Hiperalgesia/etiologia , Hiper-Homocisteinemia/induzido quimicamente , Masculino , Metionina/farmacologia , Enxaqueca com Aura/etiologia , Fotofobia/etiologia , Gravidez , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Ratos , Ratos Wistar
19.
Neurotox Res ; 39(3): 966-974, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33786757

RESUMO

Homocysteine is a sulfur amino acid that does not occur in the diet, but it is an essential intermediate in normal mammalian metabolism of methionine. Hyperhomocysteinemia results from dietary intakes of Met, folate, and vitamin B12 and lifestyle or from the deficiency of specific enzymes, leading to tissue accumulation of this amino acid and/or its metabolites. Severe hyperhomocysteinemic patients can present neurological symptoms and structural brain abnormalities, of which the pathogenesis is poorly understood. Moreover, a possible link between homocysteine (mild hyperhomocysteinemia) and neurodegenerative/neuropsychiatric disorders has been suggested. In recent years, increasing evidence has emerged suggesting that astrocyte dysfunction is involved in the neurotoxicity of homocysteine and possibly associated with the physiopathology of hyperhomocysteinemia. This review addresses some of the findings obtained from in vivo and in vitro experimental models, indicating high homocysteine levels as an important neurotoxin determinant of the neuropathophysiology of brain damage. Recent data show that this amino acid impairs glutamate uptake, redox/mitochondrial homeostasis, inflammatory response, and cell signaling pathways. Therefore, the discussion of this review focuses on homocysteine-induced gliotoxicity, and its impacts in the brain functions. Through understanding the Hcy-induced gliotoxicity, novel preventive/therapeutic strategies might emerge for these diseases.


Assuntos
Homocisteína/metabolismo , Homocisteína/toxicidade , Doenças Neurodegenerativas/induzido quimicamente , Doenças Neurodegenerativas/metabolismo , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Astrócitos/patologia , Gliose/induzido quimicamente , Gliose/metabolismo , Gliose/patologia , Humanos , Hiper-Homocisteinemia/induzido quimicamente , Hiper-Homocisteinemia/metabolismo , Hiper-Homocisteinemia/patologia , Doenças Neurodegenerativas/patologia , Neuroglia/patologia
20.
Georgian Med News ; (321): 119-125, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35000920

RESUMO

The aim of the study was to investigate the phagocytic and metabolic activity of peripheral blood neutrophils in rats with lipopolysaccharide (LPS)-induced periodontitis combined with chronic thiolactone hyperhomocysteinemia (HHcy).The experiment included non-linear mature male rats (n=48), which were divided into 4 groups: control; animals with a periodontitis model; animals with a model of chronic thiolactone HHcy; animals with a model of periodontitis in combination with chronic thiolactone HHcy. Phagocytic activity, phagocytic index and phagocytic number were determined as indicators of phagocytosis of peripheral blood neutrophils. The oxygen-dependent bactericidal activity of peripheral blood neutrophils was studied using nitroblue tetrazolium test (NBT-test).Our research has found that LPS-induced periodontitis in rats is accompanied by dysfunction of phagocytosis process (increased phagocytic activity with a simultaneous decrease of absorption capacity) and activation of oxygen-dependent microbicidal mechanisms of peripheral blood neutrophils, as indicated by an increase of indices of spontaneous and activated NBT-test. Chronic thiolactone HHcy adversely affects the functional and metabolic activity of peripheral blood neutrophils in case of periodontitis, which is confirmed by a violation of the process of phagocytosis, a more pronounced decrease in absorption capacity and depletion of metabolic reserves of these cells in rats with comorbid course of LPS-induced periodontitis vs. animals with only LPS-induced periodontitis. The observed disorders in the process of phagocytosis in rats with comorbid course of periodontitis are an important factor in reducing the non-specific organism resistance which contributes to the progression of periodontitis. The obtained results reveal new aspects of the high Hcys plasma level influence on the course of inflammatory process in periodontal tissues, which opens opportunities for improving pathogenetic therapy in patients with periodontal disease combined with chronic HHcy.


Assuntos
Hiper-Homocisteinemia , Periodontite , Animais , Hiper-Homocisteinemia/induzido quimicamente , Hiper-Homocisteinemia/complicações , Lipopolissacarídeos/toxicidade , Masculino , Neutrófilos , Periodontite/complicações , Fagocitose , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA