Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 319
1.
Sci Rep ; 13(1): 19438, 2023 11 09.
Article En | MEDLINE | ID: mdl-37945738

To provide a theoretical basis for the prevention and treatment of atherosclerosis (As), the current study aimed to investigate the mechanism underlying the effect of homocysteine (Hcy) on inducing the lipid deposition and foam cell formation of the vascular smooth muscle cell (VSMC) via C1q/Tumor necrosis factor-related protein9 (CTRP9) promoter region Hypermethylation negative regulating endoplasmic reticulum stress (ERs). Therefore, apolipoprotein E deficient (ApoE-/-) mice were randomly divided into the control [ApoE-/- + normal diet (NC)] and high methionine [ApoE-/- + (normal diet supplemented with 1.7% methionine (HMD)] groups (n = 6 mice/group). Following feeding for 15 weeks, the serum levels of Homocysteine (Hcy), total cholesterol (TC), and triglyceride (TG) were measured using an automatic biochemical analyzer. HE and oil red O staining were performed on the aorta roots to observe the pathological changes. Additionally, immunofluorescence staining was performed to detect the protein expression levels of CTRP9, glucose-regulated protein 78 kD (GRP78), phosphorylated protein kinase RNA-like ER kinase (p-PERK), activating transcription factor 6a (ATF6a), phosphorylated inositol-requiring enzyme-1α (p-IRE1α), sterol regulatory element binding proteins-1c (SREBP1c) and sterol regulatory element binding proteins-2 (SREBP2) in VSMC derived from murine aortic roots. In vitro, VSMC was stimulated with 100 µmol/l Hcy. After transfection of plasmids with overexpression and interference of CTRP9, ERs agonist (TM) and inhibitor (4-PBA) were given to stimulate VSMC cells. HE staining and oil red O staining were used to observe the effect of Hcy stimulation on lipid deposition in VSMC. Additionally, The mRNA and protein expression levels of CTRP9, GRP78, PERK, ATF6a, IRE1α, SREBP1c, and SREBP2 in VSMC were detected by RT-qPCR and western blot analysis, respectively. Finally, The methylation modification of the CTRP9 promoter region has been studied. The NCBI database was used to search the promoter region of the CTRP9 gene, and CpG Island was used to predict the methylation site. After Hcy stimulation of VSMC, overexpression of DNMT1, and intervention with 5-Azc, assess the methylation level of the CTRP9 promoter through bisulfite sequencing PCR (BSP). The results showed that the serum levels of Hcy, TC, and TG in the ApoE-/- + HMD group were significantly increased compared with the ApoE-/- + NC group. In addition, HE staining and oil red O staining showed obvious AS plaque formation in the vessel wall, and a large amount of fat deposition in VSMC, thus indicating that the hyperhomocysteinemia As an animal model was successfully established. Furthermore, CTRP9 were downregulated, while GRP78, p-PERK, ATF6a, p-IRE1α, SREBP1c, SREBP2 was upregulated in aortic VSMC in the ApoE-/- + HMD group. Consistent with the in vivo results, Hcy can inhibit the expression of CTRP9 in VSMC and induce ERs and lipid deposition in VSMC. Meanwhile, the increased expression of CTRP9 can reduce ERs and protect the lipid deposition in Hcy induced VSMC. Furthermore, ERs can promote Hcy induced VSMC lipid deposition, inhibition of ERs can reduce Hcy induced VSMC lipid deposition, and CTRP9 may play a protective role in Hcy induced VSMC lipid deposition and foam cell transformation through negative regulation of ERs. In addition, The CTRP9 promoter in the Hcy group showed hypermethylation. At the same time as Hcy intervention, overexpression of DNMT1 increases the methylation level of the CTRP9 promoter, while 5-Azc can reduce the methylation level of the CTRP9 promoter. Finally, Hcy can up-regulate the expression of DNMT1 and down-regulate the expression of CTRP9. After overexpression of DNMT1, the expression of CTRP9 is further decreased. After 5-Azc inhibition of DNMT1, the expression of DNMT1 decreases, while the expression of CTRP9 increases. It is suggested that the molecular mechanism of Hcy inhibiting the expression of CTRP9 is related to the hypermethylation of the CTRP9 promoter induced by Hcy and regulated by DNMT1. 5-Azc can inhibit the expression of DNMT1 and reverse the regulatory effect of DNMT1 on CTRP9. Overall, the results of the present study suggested that Hcy induces DNA hypermethylation in the CTRP9 promoter region by up-regulating DNMT1 expression, and negatively regulates ERs mediated VSMC lipid deposition and foam cell formation. CTRP9 may potentially be a therapeutic target in the treatment of hyperhomocysteinemia and As.


Atherosclerosis , Hyperhomocysteinemia , Mice , Animals , Endoribonucleases/metabolism , Endoplasmic Reticulum Chaperone BiP , Muscle, Smooth, Vascular/metabolism , Foam Cells/metabolism , Hyperhomocysteinemia/pathology , Protein Serine-Threonine Kinases/metabolism , Atherosclerosis/metabolism , Promoter Regions, Genetic , Methionine/metabolism , Apolipoproteins E/metabolism , Lipids/pharmacology , Homocysteine/metabolism , Sterol Regulatory Element Binding Proteins/metabolism , Endoplasmic Reticulum Stress
2.
Cell Mol Neurobiol ; 43(6): 2743-2759, 2023 Aug.
Article En | MEDLINE | ID: mdl-37074484

Parkinson's disease (PD) is one of the most common degenerative brain disorders caused by the loss of dopaminergic neurons in the substantia nigra (SN). Lewy bodies and -synuclein accumulation in the SN are hallmarks of the neuropathology of PD. Due to lifestyle changes and prolonged L-dopa administration, patients with PD frequently have vitamin deficiencies, especially folate, vitamin B6, and vitamin B12. These disorders augment circulating levels of Homocysteine with the development of hyperhomocysteinemia, which may contribute to the pathogenesis of PD. Therefore, this review aimed to ascertain if hyperhomocysteinemia may play a part in oxidative and inflammatory signaling pathways that contribute to PD development. Hyperhomocysteinemia is implicated in the pathogenesis of neurodegenerative disorders, including PD. Hyperhomocysteinemia triggers the development and progression of PD by different mechanisms, including oxidative stress, mitochondrial dysfunction, apoptosis, and endothelial dysfunction. Particularly, the progression of PD is linked with high inflammatory changes and systemic inflammatory disorders. Hyperhomocysteinemia induces immune activation and oxidative stress. In turn, activated immune response promotes the development and progression of hyperhomocysteinemia. Therefore, hyperhomocysteinemia-induced immunoinflammatory disorders and abnormal immune response may aggravate abnormal immunoinflammatory in PD, leading to more progression of PD severity. Also, inflammatory signaling pathways like nuclear factor kappa B (NF-κB) and nod-like receptor pyrin 3 (NLRP3) inflammasome and other signaling pathways are intricate in the pathogenesis of PD. In conclusion, hyperhomocysteinemia is involved in the development and progression of PD neuropathology either directly via induction degeneration of dopaminergic neurons or indirectly via activation of inflammatory signaling pathways.


Hyperhomocysteinemia , Neurodegenerative Diseases , Parkinson Disease , Humans , Parkinson Disease/metabolism , Hyperhomocysteinemia/pathology , Levodopa/metabolism , Levodopa/pharmacology , Substantia Nigra/metabolism , Neurodegenerative Diseases/metabolism , Dopaminergic Neurons/metabolism
3.
J Neurosci ; 43(10): 1797-1813, 2023 03 08.
Article En | MEDLINE | ID: mdl-36746627

Despite the indispensable role that astrocytes play in the neurovascular unit, few studies have investigated the functional impact of astrocyte signaling in cognitive decline and dementia related to vascular pathology. Diet-mediated induction of hyperhomocysteinemia (HHcy) recapitulates numerous features of vascular contributions to cognitive impairment and dementia (VCID). Here, we used astrocyte targeting approaches to evaluate astrocyte Ca2+ dysregulation and the impact of aberrant astrocyte signaling on cerebrovascular dysfunction and synapse impairment in male and female HHcy diet mice. Two-photon imaging conducted in fully awake mice revealed activity-dependent Ca2+ dysregulation in barrel cortex astrocytes under HHcy. Stimulation of contralateral whiskers elicited larger Ca2+ transients in individual astrocytes of HHcy diet mice compared with control diet mice. However, evoked Ca2+ signaling across astrocyte networks was impaired in HHcy mice. HHcy also was associated with increased activation of the Ca2+/calcineurin-dependent transcription factor NFAT4, which has been linked previously to the reactive astrocyte phenotype and synapse dysfunction in amyloid and brain injury models. Targeting the NFAT inhibitor VIVIT to astrocytes, using adeno-associated virus vectors, led to reduced GFAP promoter activity in HHcy diet mice and improved functional hyperemia in arterioles and capillaries. VIVIT expression in astrocytes also preserved CA1 synaptic function and improved spontaneous alternation performance on the Y maze. Together, the results demonstrate that aberrant astrocyte signaling can impair the major functional properties of the neurovascular unit (i.e., cerebral vessel regulation and synaptic regulation) and may therefore represent a promising drug target for treating VCID and possibly Alzheimer's disease and other related dementias.SIGNIFICANCE STATEMENT The impact of reactive astrocytes in Alzheimer's disease and related dementias is poorly understood. Here, we evaluated Ca2+ responses and signaling in barrel cortex astrocytes of mice fed with a B-vitamin deficient diet that induces hyperhomocysteinemia (HHcy), cerebral vessel disease, and cognitive decline. Multiphoton imaging in awake mice with HHcy revealed augmented Ca2+ responses in individual astrocytes, but impaired signaling across astrocyte networks. Stimulation-evoked arteriole dilation and elevated red blood cell velocity in capillaries were also impaired in cortex of awake HHcy mice. Astrocyte-specific inhibition of the Ca2+-dependent transcription factor, NFAT, normalized cerebrovascular function in HHcy mice, improved synaptic properties in brain slices, and stabilized cognition. Results suggest that astrocytes are a mechanism and possible therapeutic target for vascular-related dementia.


Alzheimer Disease , Hyperhomocysteinemia , Mice , Male , Female , Animals , Alzheimer Disease/metabolism , Astrocytes/metabolism , Hyperhomocysteinemia/metabolism , Hyperhomocysteinemia/pathology , Diet , Transcription Factors/metabolism
4.
Alzheimers Dement ; 19(2): 671-695, 2023 02.
Article En | MEDLINE | ID: mdl-36401868

This review summarizes recent evidence on how mid-life hypertension, hyperhomocysteinemia (HHcy) and blood pressure variability, as well as late-life hypotension, exacerbate Alzheimer's disease (AD) and dementia risk. Intriguingly, HHcy also increases the risk for hypertension, revealing the importance of understanding the relationship between comorbid cardiovascular risk factors. Hypertension-induced dementia presents more evidently in women, highlighting the relevance of sex differences in the impact of cardiovascular risk. We summarize each major antihypertensive drug class's effects on cognitive impairment and AD pathology, revealing how carbonic anhydrase inhibitors, diuretics modulating cerebral blood flow, have recently gained preclinical evidence as promising treatment against AD. We also report novel vascular biomarkers for AD and dementia risk, highlighting those associated with hypertension and HHcy. Importantly, we propose that future studies should consider hypertension and HHcy as potential contributors to cognitive impairment, and that uncovering the underlying molecular mechanisms and biomarkers would aid in the identification of preventive strategies.


Alzheimer Disease , Hyperhomocysteinemia , Hypertension , Humans , Female , Male , Alzheimer Disease/pathology , Hyperhomocysteinemia/complications , Hyperhomocysteinemia/pathology , Hypertension/drug therapy , Hypertension/epidemiology , Hypertension/complications , Risk Factors , Biomarkers
5.
Rev Port Cardiol ; 41(10): 813-819, 2022 10.
Article En, Pt | MEDLINE | ID: mdl-36210587

OBJECTIVES: Hyperhomocysteinemia (HHcy) can induce vascular inflammatory and oxidative damage and accelerate intimal hyperplasia. This study investigated the protective effect of pirfenidone (PFD) on the recovery process of injured endothelial arteries during HHcy. MATERIALS AND METHODS: Thirty rabbits were randomly separated into three groups: A control group (n=10, standard rabbit chow), a model group (n=10, control diet plus 30 g methionine/kg food), and a PFD group (n=10, model diet plus oral administration of 90 mg/day of PFD). After 14 weeks of arterial injury, histopathological changes were determined. Plasma homocysteine (Hcy) concentrations, lipid profiles and oxidant and antioxidant status were evaluated. Macrophage infiltration was assessed using immunohistochemical staining. RESULTS: PFD supplementation decreased macrophage infiltration of iliac artery significantly without changes in blood lipids and Hcy concentrations. Compared with the model group, PFD restored superoxide dismutase and glutathione peroxidase activities and reduced malondialdehyde and reactive oxygen species levels. A high-methionine diet significantly increased neointimal area and the ratio between neointimal and media area. Systemic administration of PFD inhibited neointimal formation. CONCLUSIONS: PFD can partly alleviate intimal hyperplasia by inhibiting inflammatory and oxidative stress response induced by HHcy during endothelial injury. It may be a potential therapeutic agent for the prevention and treatment of endothelial injury-associated diseases such as atherosclerosis.


Hyperhomocysteinemia , Animals , Antioxidants/pharmacology , Antioxidants/therapeutic use , Glutathione Peroxidase/pharmacology , Glutathione Peroxidase/therapeutic use , Homocysteine/pharmacology , Homocysteine/therapeutic use , Hyperhomocysteinemia/complications , Hyperhomocysteinemia/drug therapy , Hyperhomocysteinemia/pathology , Hyperplasia/pathology , Lipids , Malondialdehyde/pharmacology , Methionine/pharmacology , Methionine/therapeutic use , Oxidants/pharmacology , Oxidants/therapeutic use , Pyridones , Rabbits , Reactive Oxygen Species/pharmacology , Reactive Oxygen Species/therapeutic use , Superoxide Dismutase/pharmacology , Superoxide Dismutase/therapeutic use , Tunica Intima/pathology
6.
Folia Histochem Cytobiol ; 60(2): 111-124, 2022.
Article En | MEDLINE | ID: mdl-35603572

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.


Cardiovascular Diseases , Hyperhomocysteinemia , Animals , Cardiovascular Diseases/chemically induced , Cardiovascular Diseases/complications , Gerbillinae , Hyperhomocysteinemia/chemically induced , Hyperhomocysteinemia/complications , Hyperhomocysteinemia/pathology , Methionine , Phenotype
7.
Nutrients ; 14(7)2022 Apr 01.
Article En | MEDLINE | ID: mdl-35406088

Hyperhomocysteinemia (HHcy) is a methionine metabolism problem that causes a variety of inflammatory illnesses. Oxidative stress is among the processes thought to be involved in the pathophysiology of the damage produced by HHcy. HHcy is likely to involve the dysfunction of several organs, such as the kidney, liver, or gut, which are currently poorly understood. Nuts are regarded as an important part of a balanced diet since they include protein, good fatty acids, and critical nutrients. The aim of this work was to evaluate the anti-inflammatory and antioxidant effects of cashew nuts in HHcy induced by oral methionine administration for 30 days, and to examine the possible pathways involved. In HHcy rats, cashew nuts (100 mg/kg orally, daily) were able to counteract clinical biochemical changes, oxidative and nitrosative stress, reduced antioxidant enzyme levels, lipid peroxidation, proinflammatory cytokine release, histological tissue injuries, and apoptosis in the kidney, colon, and liver, possibly by the modulation of the antioxidant nuclear factor erythroid 2-related factor 2 NRF-2 and inflammatory nuclear factor NF-kB pathways. Thus, the results suggest that the consumption of cashew nuts may be beneficial for the treatment of inflammatory conditions associated with HHcy.


Anacardium , Hyperhomocysteinemia , Animals , Antioxidants/metabolism , Hyperhomocysteinemia/pathology , Inflammation , Methionine/metabolism , Nuts/metabolism , Oxidative Stress , Rats
8.
Am J Pathol ; 192(1): 43-55, 2022 01.
Article En | MEDLINE | ID: mdl-34717894

Nucleotide-binding oligomerization domain-like receptor containing pyrin domain 3 (NLRP3) inflammasome activation in podocytes is reportedly associated with enhanced release of exosomes containing NLRP3 inflammasome products from these cells during hyperhomocysteinemia (hHcy). This study examined the possible role of increased exosome secretion during podocyte NLRP3 inflammasome activation in the glomerular inflammatory response. Whether exosome biogenesis and lysosome function are involved in the regulation of exosome release from podocytes during hHcy in mice and upon stimulation of homocysteine (Hcy) in podocytes was tested. By nanoparticle tracking analysis, treatments of mice with amitriptyline (acid sphingomyelinase inhibitor), GW4869 (exosome biogenesis inhibitor), and rapamycin (lysosome function enhancer) were found to inhibit elevated urinary exosomes during hHcy. By examining NLRP3 inflammasome activation in glomeruli during hHcy, amitriptyline (but not GW4869 and rapamycin) was shown to have an inhibitory effect. However, all treatments attenuated glomerular inflammation and injury during hHcy. In cell studies, Hcy treatment stimulated exosome release from podocytes, which was prevented by amitriptyline, GW4869, and rapamycin. Structured illumination microscopy revealed that Hcy inhibited lysosome-multivesicular body interactions in podocytes, which was prevented by amitriptyline or rapamycin but not GW4869. Thus, the data from this study shows that activation of exosome biogenesis and dysregulated lysosome function are critically implicated in the enhancement of exosome release from podocytes leading to glomerular inflammation and injury during hHcy.


Exosomes/metabolism , Hyperhomocysteinemia/pathology , Inflammation/pathology , Kidney Glomerulus/pathology , Lysosomes/metabolism , Podocytes/metabolism , Animals , Homocysteine/metabolism , Inflammasomes/metabolism , Male , Mice, Inbred C57BL , Multivesicular Bodies/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Podocytes/pathology , Sphingomyelin Phosphodiesterase/metabolism
9.
Sci Rep ; 11(1): 21430, 2021 11 02.
Article En | MEDLINE | ID: mdl-34728708

Artificial neural network (ANN) is the main tool to dig data and was inspired by the human brain and nervous system. Several studies clarified its application in medicine. However, none has applied ANN to predict the efficacy of folic acid treatment to Hyperhomocysteinemia (HHcy). The efficacy has been proved to associate with both genetic and environmental factors while previous studies just focused on the latter one. The explained variance genetic risk score (EV-GRS) had better power and could represent the effect of genetic architectures. Our aim was to add EV-GRS into environmental factors to establish ANN to predict the efficacy of folic acid therapy to HHcy. We performed the prospective cohort research enrolling 638 HHcy patients. The multilayer perception algorithm was applied to construct ANN. To evaluate the effect of ANN, we also established logistic regression (LR) model to compare with ANN. According to our results, EV-GRS was statistically associated with the efficacy no matter analyzed as a continuous variable (OR = 3.301, 95%CI 1.954-5.576, P < 0.001) or category variable (OR = 3.870, 95%CI 2.092-7.159, P < 0.001). In our ANN model, the accuracy was 84.78%, the Youden's index was 0.7073 and the AUC was 0.938. These indexes above indicated higher power. When compared with LR, the AUC, accuracy, and Youden's index of the ANN model (84.78%, 0.938, 0.7073) were all slightly higher than the LR model (83.33% 0.910, 0.6687). Therefore, clinical application of the ANN model may be able to better predict the folic acid efficacy to HHcy than the traditional LR model. When testing two models in the validation set, we got the same conclusion. This study appears to be the first one to establish the ANN model which added EV-GRS into environmental factors to predict the efficacy of folic acid to HHcy. This model would be able to offer clinicians a new method to make decisions and individual therapeutic plans.


Algorithms , Folic Acid/therapeutic use , Genetic Markers , Genetic Predisposition to Disease , Hyperhomocysteinemia/drug therapy , Neural Networks, Computer , Aged , Female , Humans , Hyperhomocysteinemia/genetics , Hyperhomocysteinemia/pathology , Male , Prospective Studies , Risk Factors , Treatment Outcome
10.
Int J Mol Sci ; 22(12)2021 Jun 10.
Article En | MEDLINE | ID: mdl-34200792

Hyperhomocysteinemia (HHcy) is remarkably common among the aging population. The relation between HHcy and the development of neurodegenerative diseases, such as Alzheimer's disease (AD) and eye diseases, and age-related macular degeneration (AMD) and diabetic retinopathy (DR) in elderly people, has been established. Disruption of the blood barrier function of the brain and retina is one of the most important underlying mechanisms associated with HHcy-induced neurodegenerative and retinal disorders. Impairment of the barrier function triggers inflammatory events that worsen disease pathology. Studies have shown that AD patients also suffer from visual impairments. As an extension of the central nervous system, the retina has been suggested as a prominent site of AD pathology. This review highlights inflammation as a possible underlying mechanism of HHcy-induced barrier dysfunction and neurovascular injury in aging diseases accompanied by HHcy, focusing on AD.


Central Nervous System Diseases/pathology , Homocysteine/metabolism , Hyperhomocysteinemia/pathology , Inflammation/physiopathology , Age Factors , Animals , Central Nervous System Diseases/etiology , Central Nervous System Diseases/metabolism , Humans , Hyperhomocysteinemia/etiology , Hyperhomocysteinemia/metabolism
11.
Cells ; 10(6)2021 06 18.
Article En | MEDLINE | ID: mdl-34207057

Maternal hyperhomocysteinemia is one of the common complications of pregnancy that causes offspring cognitive deficits during postnatal development. In this study, we investigated the effect of prenatal hyperhomocysteinemia (PHHC) on inflammatory, glial activation, and neuronal cell death markers in the hippocampus of infant rats. Female Wistar rats received L-methionine (0.6 g/kg b.w.) by oral administration during pregnancy. On postnatal days 5 and 20, the offspring's hippocampus was removed to perform histological and biochemical studies. After PHHC, the offspring exhibited increased brain interleukin-1ß and interleukin-6 levels and glial activation, as well as reduced anti-inflammatory interleukin-10 level in the hippocampus. Additionally, the activity of acetylcholinesterase was increased in the hippocampus of the pups. Exposure to PHHC also resulted in the reduced number of neurons and disrupted neuronal ultrastructure. At the same time, no changes in the content and activity of caspase-3 were found in the hippocampus of the pups. In conclusion, our findings support the hypothesis that neuroinflammation and glial activation could be involved in altering the hippocampus cellular composition following PHHC, and these alterations could be associated with cognitive disorders later in life.


Biomarkers/metabolism , Hippocampus/metabolism , Hyperhomocysteinemia/metabolism , Inflammation/metabolism , Neuroglia/metabolism , Neurons/metabolism , Prenatal Exposure Delayed Effects/metabolism , Acetylcholinesterase/metabolism , Animals , Animals, Newborn , Brain/metabolism , Brain/pathology , Caspase 3/metabolism , Cytokines/metabolism , Female , Hippocampus/pathology , Hyperhomocysteinemia/pathology , Inflammation/pathology , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Methionine/metabolism , Neuroglia/pathology , Neurons/pathology , Pregnancy , Prenatal Exposure Delayed Effects/pathology , Rats , Rats, Wistar
12.
Int J Mol Sci ; 22(9)2021 May 07.
Article En | MEDLINE | ID: mdl-34066973

L-methionine, an essential amino acid, plays a critical role in cell physiology. High intake and/or dysregulation in methionine (Met) metabolism results in accumulation of its intermediate(s) or breakdown products in plasma, including homocysteine (Hcy). High level of Hcy in plasma, hyperhomocysteinemia (hHcy), is considered to be an independent risk factor for cerebrovascular diseases, stroke and dementias. To evoke a mild hHcy in adult male Wistar rats we used an enriched Met diet at a dose of 2 g/kg of animal weight/day in duration of 4 weeks. The study contributes to the exploration of the impact of Met enriched diet inducing mild hHcy on nervous tissue by detecting the histo-morphological, metabolomic and behavioural alterations. We found an altered plasma metabolomic profile, modified spatial and learning memory acquisition as well as remarkable histo-morphological changes such as a decrease in neurons' vitality, alterations in the morphology of neurons in the selective vulnerable hippocampal CA 1 area of animals treated with Met enriched diet. Results of these approaches suggest that the mild hHcy alters plasma metabolome and behavioural and histo-morphological patterns in rats, likely due to the potential Met induced changes in "methylation index" of hippocampal brain area, which eventually aggravates the noxious effect of high methionine intake.


Behavior, Animal , Hippocampus/pathology , Hyperhomocysteinemia/blood , Hyperhomocysteinemia/metabolism , Metabolomics , Animals , Homocysteine/blood , Hyperhomocysteinemia/pathology , In Situ Nick-End Labeling , Magnetic Resonance Spectroscopy , Male , Methionine , Rats, Wistar , Staining and Labeling
13.
Curr Med Sci ; 41(3): 548-554, 2021 Jun.
Article En | MEDLINE | ID: mdl-34169425

Ligustrazine, an alkaloid extracted from the traditional Chinese herbal medicine Ligusticum Chuanxiong Hort, has been clinically applied to treat the cerebrovascular diseases. Hyperhomocysteinemia (Hhcy) is an independent risk factor for Alzheimer's disease (AD). Memory deficits can be caused by Hhcy via pathologies of AD-like tau and amyloid-ß (Aß) in the hippocampus. Here, we investigated whether homocysteine (Hcy) can induce AD-like pathologies and the effects of ligustrazine on these pathologies. The Hcy rat model was constructed by 14-day Hcy injection via vena caudalis, and rats were treated with daily intragastric administration of ligustrazine at the same time. We found that the pathologies of tau and Aß were induced by Hcy in the hippocampus, while the Hcy-induced tau hyperphosphorylation and Aß accumulation could be markedly attenuated by simultaneous ligustrazine treatment. Our data demonstrate that ligustrazine may be used as a promising neuroprotective agent to treat the Hcy-induced AD-like pathologies.


Alzheimer Disease/drug therapy , Hyperhomocysteinemia/drug therapy , Memory Disorders/drug therapy , Pyrazines/pharmacology , Alzheimer Disease/etiology , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides , Animals , Brain/drug effects , Brain/pathology , Disease Models, Animal , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Hyperhomocysteinemia/complications , Hyperhomocysteinemia/genetics , Hyperhomocysteinemia/pathology , Memory Disorders/etiology , Memory Disorders/genetics , Memory Disorders/pathology , Neurons/drug effects , Neurons/pathology , Neuroprotective Agents/pharmacology , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley
14.
Neurotox Res ; 39(3): 966-974, 2021 Jun.
Article En | MEDLINE | ID: mdl-33786757

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.


Homocysteine/metabolism , Homocysteine/toxicity , Neurodegenerative Diseases/chemically induced , Neurodegenerative Diseases/metabolism , Neuroglia/drug effects , Neuroglia/metabolism , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Gliosis/chemically induced , Gliosis/metabolism , Gliosis/pathology , Humans , Hyperhomocysteinemia/chemically induced , Hyperhomocysteinemia/metabolism , Hyperhomocysteinemia/pathology , Neurodegenerative Diseases/pathology , Neuroglia/pathology
15.
Biomark Med ; 15(5): 337-345, 2021 04.
Article En | MEDLINE | ID: mdl-33666517

Aim: The present study observed the relationship between the methylenetetrahydrofolate reductase genotypes and clinical outcome in children with sickle cell disorder. Methodology: A total of 249 children were recruited for the study and evaluated clinically for calculating severity score, homocysteine levels and C677T and A1298C genotyping. Results: The frequencies of variant genotypes were 28.1% CT/TT677 and 69.1% AC/CC1298. Plasma homocysteine was significantly elevated in variant groups (p < 0.001). Both the genotypes accorded significant association with homocysteinemia (p < 0.001). Vascular crisis (p = 0.04), frequency of hospitalization (p < 0.001) and severity score (p = 0.02) revealed association with C677T and not with A1298C. The CT/TT677 genotypes showed 3.39-times (p = 0.032) increase in a higher score for severity. Conclusion: C677T depicted significant association with clinical severity in study population.


Anemia, Sickle Cell/complications , Genetic Markers , Hyperhomocysteinemia/pathology , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Polymorphism, Single Nucleotide , Severity of Illness Index , Adolescent , Child , Child, Preschool , Cross-Sectional Studies , Female , Genotype , Homocysteine/metabolism , Humans , Hyperhomocysteinemia/etiology , Hyperhomocysteinemia/metabolism , Male
16.
Biochem Biophys Res Commun ; 547: 125-130, 2021 04 02.
Article En | MEDLINE | ID: mdl-33610040

ABJECTIVE: Interaction of hypertension and hyperhomocysteinemia (HHcy) leads to enhanced cardiac remodeling in hypertensive heart disease. However, the mechanism of collagen accumulation and cardiac remodeling remains unclear. In this study, we attempted to evaluate the relationship between hypertension and HHcy in the context of cardiac remodeling and to explore its mechanism of action. METHODS: Wistar Kyoto (WKY) and spontaneous hypertension rats (SHR) were randomly divided into four groups, namely WKY group, WKY + HHcy group, SHR group and SHR + HHcy group. We measured blood pressure (BP), plasma homocysteine (Hcy), serum superoxide dismutase (SOD) and serum malondialdehyde (MDA). We also examined cardiac histopathology and gene and protein expression levels of Nrf2 and HO-1. RESULTS: Compared with the WKY group, myocardial interstitial and perivascular collagen deposition in the WKY + HHcy group, the SHR group and the SHR + HHcy group increased successively, indicating that cardiac remodeling gradually increased, and HHcy aggravated cardiac remodeling was more serious in hypertensive rats. SOD decreased gradually in the four groups, while MDA was on the contrary. WKY + HHcy and SHR + HHcy groups both suppressed Nrf2 and HO-1 expression and inhibited the translocation of Nrf2 from cytoplasm to nucleus compared with their control groups, and the SHR + HHcy group had a stronger inhibitory effect. CONCLUSION: HHcy enhanced cardiac remodeling in rats by enhancing oxidative stress, suppressing the Nrf2/HO-1 pathway and Nrf2 nuclear transport, and this inhibitory effect was stronger in the context of hypertension.


Heart Diseases/metabolism , Heme Oxygenase (Decyclizing)/metabolism , Hyperhomocysteinemia/metabolism , Hypertension/metabolism , NF-E2-Related Factor 2/metabolism , Ventricular Remodeling , Animals , Disease Models, Animal , Heart Diseases/etiology , Heart Diseases/pathology , Hyperhomocysteinemia/pathology , Hypertension/pathology , Male , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Signal Transduction
17.
Int J Mol Sci ; 22(1)2021 Jan 03.
Article En | MEDLINE | ID: mdl-33401548

Erectile Dysfunction (ED) is defined as the inability to maintain and/or achieve a satisfactory erection. This condition can be influenced by the presence of atherosclerosis, a systemic pathology of the vessels that also affects the cavernous arteries and which can cause an alteration of blood flow at penile level. Among the cardiovascular risk factors affecting the genesis of atherosclerosis, hyperhomocysteinemia (HHcys) plays a central role, which is associated with oxidative stress and endothelial dysfunction. This review focuses on the biological processes that lead to homocysteine-induced endothelial damage and discusses the consequences of HHcys on male sexual function.


Endothelium, Vascular/physiopathology , Erectile Dysfunction/complications , Hyperhomocysteinemia/pathology , Oxidative Stress , Animals , Humans , Hyperhomocysteinemia/etiology , Male , Risk Factors
18.
Cell Mol Life Sci ; 78(4): 1709-1727, 2021 Feb.
Article En | MEDLINE | ID: mdl-32734582

Abdominal aortic aneurysm (AAA) is characterized by inflammatory cell infiltration and aggravated by hyperhomocysteinemia (HHcy). It is unknown whether the homocysteine (Hcy)-activated RNA methyltransferase NOP2/Sun domain family member 2 (NSun2) is associated with AAA. Here, we found that NSun2 deficiency significantly attenuated elastase-induced and HHcy-aggravated murine AAA with decreased T cell infiltration in the vessel walls. T cell labeling and adoptive transfer experiments confirmed that NSun2 deficiency inhibited the chemotaxis of vessels to T cells. RNA sequencing of endothelial cells showed that Hcy induced the accumulation of various metabolic enzymes of the phospholipid PC-LPC-LPA metabolic pathway, especially autotaxin (ATX). In the elastase-induced mouse model of AAA, ATX was specifically expressed in the endothelium and the plasma ATX concentration was upregulated and even higher in the HHcy group, which were decreased dramatically by NSun2 knockdown. In vitro Transwell experiments showed that ATX dose-dependently promoted T cell migration. HHcy may upregulate endothelial ATX expression and secretion and in turn recruit T cells into the vessel walls to induce vascular inflammation and consequently accelerate the pathogenesis of AAA. Mechanistically, secreted ATX interacted with T cells by binding to integrin α4, which subsequently activated downstream FAK/Src-RhoA signaling pathways and then induced T cell chemokinesis and adhesion. ATX overexpression in the vessel walls reversed the inhibited development of AAA in the NSun2-deficient mice. Therefore, NSun2 mediates the development of HHcy-aggravated AAA primarily by increasing endothelial ATX expression, secretion and T cell migration, which is a novel mechanism for HHcy-aggravated vascular inflammation and pathogenesis of AAA.


Aortic Aneurysm, Abdominal/genetics , Hyperhomocysteinemia/genetics , Inflammation/genetics , Methyltransferases/genetics , Phosphoric Diester Hydrolases/genetics , Animals , Aortic Aneurysm, Abdominal/complications , Aortic Aneurysm, Abdominal/immunology , Aortic Aneurysm, Abdominal/pathology , Cell Movement/genetics , Disease Models, Animal , Endothelial Cells/immunology , Endothelial Cells/metabolism , Gene Expression Regulation/genetics , Humans , Hyperhomocysteinemia/complications , Hyperhomocysteinemia/pathology , Inflammation/complications , Inflammation/pathology , Mice , Signal Transduction/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
19.
Curr Res Transl Med ; 69(1): 103270, 2021 01.
Article En | MEDLINE | ID: mdl-33268288

BACKGROUND: Hyperhomocysteinemia (HHcy) is one of the major risk factors of cardiovascular diseases. Metformin acts as a cardioprotective role in several cardiovascular diseases, including ischemia/reperfusion, atherosclerosis, and myocardial infarction. However, whether metformin protects against HHcy-induced cardiac hypertrophy is unclear. METHODS AND RESULTS: HHcy model was established in C57BL/6 mice with high L-methionine (L-MET) diet for 12 weeks. AC16 cells were exposed to homocysteine (Hcy) and then intervened with different concentrations of metformin in in vitro studies. The results showed that HHcy was able to induce cardiac hypertrophy, and metformin could abrogate this effect. HHcy increased the fibrosis area and induced apoptosis in the myocardium, whereas metformin could reverse the detrimental effects above. TUNEL assay showed that metformin was able to decrease Hcy-induced apoptosis in AC16 cells. Moreover, western blotting assay revealed that metformin could decrease Hcy-induced expression of Bax and cleaved caspase3, and increase the expression of Bcl-2. CONCLUSIONS: This study demonstrates that metformin is able to attenuate HHcy-induced cardiac hypertrophy by decreasing myocardial fibrosis and apoptosis.


Apoptosis/drug effects , Cardiomegaly , Hyperhomocysteinemia , Metformin/pharmacology , Myocardium/pathology , Adult , Animals , Cardiomegaly/drug therapy , Cardiomegaly/etiology , Cardiomegaly/pathology , Cells, Cultured , Fibrosis , Heart/drug effects , Humans , Hyperhomocysteinemia/complications , Hyperhomocysteinemia/drug therapy , Hyperhomocysteinemia/pathology , Male , Metformin/therapeutic use , Mice , Mice, Inbred C57BL
20.
J Neuroinflammation ; 17(1): 274, 2020 Sep 17.
Article En | MEDLINE | ID: mdl-32943069

BACKGROUND: Elevated blood homocysteine levels, termed hyperhomocysteinemia (HHcy), is a prevalent risk factor for Alzheimer's disease (AD) in elderly populations. While dietary supplementation of B-vitamins is a generally effective method to lower homocysteine levels, there is little if any benefit to cognition. In the context of amyloid pathology, dietary-induced HHcy is known to enhance amyloid deposition and certain inflammatory responses. Little is known, however, about whether there is a more specific effect on microglia resulting from combined amyloid and HHcy pathologies. METHODS: The present study used a knock-in mouse model of amyloidosis, aged to 12 months, given 8 weeks of B-vitamin deficiency-induced HHcy to better understand how microglia are affected in this comorbidity context. RESULTS: We found that HHcy-inducing diet increased amyloid plaque burden, altered the neuroinflammatory milieu, and upregulated the expression of multiple damage-associated and "homeostatic" microglial genes. CONCLUSIONS: Taken together, these data indicate complex effects of comorbid pathologies on microglial function that are not driven solely by increased amyloid burden. Given the highly dynamic nature of microglia, their central role in AD pathology, and the frequent occurrence of various comorbidities in AD patients, it is increasingly important to understand how microglia respond to mixed pathological processes.


Aging/metabolism , Alzheimer Disease/metabolism , Gene Knock-In Techniques/methods , Hyperhomocysteinemia/metabolism , Microglia/metabolism , Plaque, Amyloid/metabolism , Aging/genetics , Aging/pathology , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Animals , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Disease Models, Animal , Hippocampus/metabolism , Hippocampus/pathology , Hyperhomocysteinemia/genetics , Hyperhomocysteinemia/pathology , Mice , Mice, Transgenic , Microglia/pathology , Plaque, Amyloid/genetics , Plaque, Amyloid/pathology
...