Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
Front Immunol ; 12: 632333, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33717169

RESUMEN

Background: Murine monocytes (MC) are classified into Ly6Chigh and Ly6Clow MC. Ly6Chigh MC is the pro-inflammatory subset and the counterpart of human CD14++CD16+ intermediate MC which contributes to systemic and tissue inflammation in various metabolic disorders, including hyperhomocysteinemia (HHcy). This study aims to explore molecule signaling mediating MC subset differentiation in HHcy and control mice. Methods: RNA-seq was performed in blood Ly6Chigh and Ly6Clow MC sorted by flow cytometry from control and HHcy cystathionine ß-synthase gene-deficient (Cbs-/-) mice. Transcriptome data were analyzed by comparing Ly6Chigh vs. Ly6Clow in control mice, Ly6Chigh vs. Ly6Clow in Cbs-/- mice, Cbs-/- Ly6Chigh vs. control Ly6Chigh MC and Cbs-/- Ly6Clow vs. control Ly6Clow MC by using intensive bioinformatic strategies. Significantly differentially expressed (SDE) immunological genes and transcription factor (TF) were selected for functional pathways and transcriptional signaling identification. Results: A total of 7,928 SDE genes and 46 canonical pathways derived from it were identified. Ly6Chigh MC exhibited activated neutrophil degranulation, lysosome, cytokine production/receptor interaction and myeloid cell activation pathways, and Ly6Clow MC presented features of lymphocyte immunity pathways in both mice. Twenty-four potential transcriptional regulatory pathways were identified based on SDE TFs matched with their corresponding SDE immunological genes. Ly6Chigh MC presented downregulated co-stimulatory receptors (CD2, GITR, and TIM1) which direct immune cell proliferation, and upregulated co-stimulatory ligands (LIGHT and SEMA4A) which trigger antigen priming and differentiation. Ly6Chigh MC expressed higher levels of macrophage (MΦ) markers, whereas, Ly6Clow MC highly expressed lymphocyte markers in both mice. HHcy in Cbs-/- mice reinforced inflammatory features in Ly6Chigh MC by upregulating inflammatory TFs (Ets1 and Tbx21) and strengthened lymphocytes functional adaptation in Ly6Clow MC by increased expression of CD3, DR3, ICOS, and Fos. Finally, we established 3 groups of transcriptional models to describe Ly6Chigh to Ly6Clow MC subset differentiation, immune checkpoint regulation, Ly6Chigh MC to MΦ subset differentiation and Ly6Clow MC to lymphocyte functional adaptation. Conclusions: Ly6Chigh MC displayed enriched inflammatory pathways and favored to be differentiated into MΦ. Ly6Clow MC manifested activated T-cell signaling pathways and potentially can adapt the function of lymphocytes. HHcy reinforced inflammatory feature in Ly6Chigh MC and strengthened lymphocytes functional adaptation in Ly6Clow MC.


Asunto(s)
Antígenos Ly/inmunología , Hiperhomocisteinemia/inmunología , Monocitos/inmunología , Animales , Antígenos Ly/metabolismo , Diferenciación Celular/inmunología , Cistationina betasintasa/deficiencia , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Hiperhomocisteinemia/metabolismo , Proteínas de Punto de Control Inmunitario/genética , Inflamación , Linfocitos/inmunología , Lisosomas/inmunología , Macrófagos/inmunología , Ratones , Monocitos/metabolismo , Transducción de Señal , Factores de Transcripción/genética
2.
Can J Physiol Pharmacol ; 99(1): 115-123, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32721223

RESUMEN

Hyperhomocysteinemia (HHcy) affects bone remodeling, since a destructive process in cortical alveolar bone has been linked to it; however, the mechanism remains at large. HHcy increases proinflammatory cytokines viz. TNF-α, IL-1b, IL-6, and IL-8 that leads to a cascade that negatively impacts methionine metabolism and homocysteine cycling. Further, chronic inflammation decreases vitamins B12, B6, and folic acid that are required for methionine homocysteine homeostasis. This study aims to investigate a HHcy mouse model (cystathionine ß-synthase deficient, CBS+/-) for studying the potential pathophysiological changes, if any, in the periodontium (gingiva, periodontal ligament, cement, and alveolar bone). We compared the periodontium side-by-side in the CBS+/- model with that of the wild-type (C57BL/6J) mice. Histology and histomorphometry of the mandibular bone along with gene expression analyses were carried out. Also, proangiogenic proteins and metalloproteinases were studied. To our knowledge, this research shows, for the first time, a direct connection between periodontal disease during CBS deficiency, thereby suggesting the existence of disease drivers during the hyperhomocysteinemic condition. Our findings offer opportunities to develop diagnostics/therapeutics for people who suffer from chronic metabolic disorders like HHcy.


Asunto(s)
Cistationina betasintasa/deficiencia , Hiperhomocisteinemia/complicaciones , Periodontitis/inmunología , Periodoncio/patología , Animales , Cistationina betasintasa/genética , Modelos Animales de Enfermedad , Ácido Fólico , Homocisteína/sangre , Homocisteína/metabolismo , Humanos , Hiperhomocisteinemia/sangre , Hiperhomocisteinemia/inmunología , Hiperhomocisteinemia/metabolismo , Masculino , Ratones , Ratones Transgénicos , Estrés Oxidativo/inmunología , Periodontitis/patología , Periodoncio/inmunología
3.
Can J Physiol Pharmacol ; 99(1): 9-17, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32706987

RESUMEN

Periodontal disease is one of the most common conditions resulting from poor oral hygiene and is characterized by a destructive process in the periodontium that essentially includes gingiva, alveolar mucosa, cementum, periodontal ligament, and alveolar bone. Notably, the destructive event in the alveolar bone has been linked to homocysteine (Hcy) metabolism; however, it has not been fully investigated. Therefore; the implication of Hcy towards initiation, progression, and maintenance of the periodontal disease remains incompletely understood. Higher levels of Hcy (also known as hyperhomocysteinemia (HHcy)) exerts deleterious effects on gum health and teeth in distinct ways. Firstly, increased production of proinflammatory cytokines such as TNF-α, IL-1ß, IL-6, and IL-8 leads to an inflammatory cascade of events that affect methionine (Met) and Hcy metabolism (i.e., 1-carbon metabolism) leading to HHcy. Secondly, metabolic dysregulation during chronic medical conditions increases systemic inflammation leading to a decrease in vitamins, more specifically B6, B12, and folic acid, that play important roles as cofactors in Hcy metabolism. Also, given the folate level in the HHcy state that is important during dysbiosis, these two conditions appear to be intimately related, and in this context, HHcy-induced dysbiosis may be one of the potential causes of periodontal disease. This paper sums up the link between periodontitis and HHcy, with a special emphasis on the "oral-gut microbiome axis" and the potential probiotic intervention towards warding off some of the serious periodontal disease conditions.


Asunto(s)
Disbiosis/complicaciones , Microbioma Gastrointestinal/fisiología , Homocisteína/metabolismo , Hiperhomocisteinemia/inmunología , Periodontitis/inmunología , Disbiosis/sangre , Disbiosis/inmunología , Disbiosis/microbiología , Ácido Fólico/sangre , Ácido Fólico/metabolismo , Homocisteína/sangre , Homocisteína/inmunología , Humanos , Hiperhomocisteinemia/sangre , Hiperhomocisteinemia/metabolismo , Metionina/metabolismo , Periodontitis/sangre , Periodontitis/metabolismo , Probióticos
4.
Cardiovasc Res ; 116(11): 1897-1909, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31782769

RESUMEN

AIMS: Overactivated B cells secrete pathological antibodies, which in turn accelerate the formation of abdominal aortic aneurysms (AAAs). Hyperhomocysteinaemia (HHcy) aggravates AAA in mice; however, the underlying mechanisms remain largely elusive. In this study, we further investigated whether homocysteine (Hcy)-activated B cells produce antigen-specific antibodies that ultimately contribute to AAA formation. METHODS AND RESULTS: ELISA assays showed that HHcy induced the secretion of anti-beta 2 glycoprotein I (anti-ß2GPI) antibody from B cells both in vitro and in vivo. Mechanistically, Hcy increased the accumulation of various lipid metabolites in B cells tested by liquid chromatography-tandem mass spectrometry, which contributed to elevated anti-ß2GPI IgG secretion. By using the toll-like receptor 4 (TLR4)-specific inhibitor TAK-242 or TLR4-deficient macrophages, we found that culture supernatants from Hcy-activated B cells and HHcy plasma IgG polarized inflammatory macrophages in a TLR4-dependent manner. In addition, HHcy markedly increased the incidence of elastase- and CaPO4-induced AAA in male BALB/c mice, which was prevented in µMT mice. To further determine the importance of IgG in HHcy-aggravated AAA formation, we purified plasma IgG from HHcy or control mice and then transferred the IgG into µMT mice, which were subsequently subjected to elastase- or CaPO4-induced AAA. Compared with µMT mice that received plasma IgG from control mice, µMT mice that received HHcy plasma IgG developed significantly exacerbated elastase- or CaPO4-induced AAA accompanied by increased elastin degradation, MMP2/9 expression, and anti-ß2GPI IgG deposition in vascular lesions, as shown by immunofluorescence histochemical staining. CONCLUSION: Our findings reveal a novel mechanism by which Hcy-induced B cell-derived pathogenic anti-ß2GPI IgG might, at least in part, contribute to HHcy-aggravated chronic vascular inflammation and AAA formation.


Asunto(s)
Aorta Abdominal/inmunología , Aneurisma de la Aorta Abdominal/inmunología , Autoanticuerpos/metabolismo , Linfocitos B/inmunología , Hiperhomocisteinemia/inmunología , beta 2 Glicoproteína I/inmunología , Animales , Aorta Abdominal/metabolismo , Aorta Abdominal/patología , Aneurisma de la Aorta Abdominal/inducido químicamente , Aneurisma de la Aorta Abdominal/metabolismo , Aneurisma de la Aorta Abdominal/patología , Linfocitos B/metabolismo , Fosfatos de Calcio , Células Cultivadas , Modelos Animales de Enfermedad , Elastina/metabolismo , Hiperhomocisteinemia/complicaciones , Hiperhomocisteinemia/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Elastasa Pancreática , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
5.
Zh Nevrol Psikhiatr Im S S Korsakova ; 119(11): 133-138, 2019.
Artículo en Ruso | MEDLINE | ID: mdl-31851185

RESUMEN

Endothelial dysfunction today is recognized as one of the leading factors in the pathogenesis of diseases of the central nervous system of various etiologies. Numerous studies have shown the role of hyperhomocysteinemia in the development of endothelial dysfunction and prothrombogenic state. The most important condition in the development of multiple sclerosis (MS) is dysregulation of the blood-brain barrier (BBB) and transendothelial leukocyte migration. It has been proven that homocysteine also contributes to the damage of neurons by the mechanism of excitotoxicity and induction of apoptosis of neurons. These processes can be one of the factors of neurodegenerative brain damage, which plays a leading role in the progression of MS. This review describes the pleiotropic effect of homocysteine on these processes and its role in the pathogenesis of MS.


Asunto(s)
Endotelio , Hiperhomocisteinemia , Esclerosis Múltiple , Barrera Hematoencefálica , Sistema Nervioso Central , Endotelio/fisiopatología , Humanos , Hiperhomocisteinemia/inmunología , Hiperhomocisteinemia/fisiopatología , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/fisiopatología , Neuronas
6.
J Cell Biochem ; 120(6): 9421-9429, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30681198

RESUMEN

BACKGROUND: Elevated plasma homocysteine (Hcy) concentration is considered as the diagnostic criteria of Hyperhomocysteinemia (HHcy), which is associated with the inflammatory response and blood-brain barrier disruption. Previous studies have proposed that HHcy with hypertension was associated with the brain injury by enhancing the cerebrovascular permeability, however, the immune mechanism remains obscure. The purpose of the study is to explore the immunomodulatory mechanism of brain injury in spontaneously hypertensive rats (SHRs) induced by HHcy. MATERIALS AND METHODS: Sixty SHRs were randomly assigned to three groups: SHR-C (control group), SHR-M (methionine group) and SHR-T (treatment group). Physical examination of body weight, systolic blood pressure (SBP) and plasma Hcy content was measured every 4 weeks. Besides, T-helper cell 17 and regulatory T cells (Treg)-related inflammatory cytokines (interleukin [IL]-6, IL-17, IL-10, and transforming growth factor beta [TGF-ß]) and genes (RORγt and FoxP3) were detected by enzyme-linked immunosorbent assay, quantitative polymerase chain reaction , Western blot, and immunohistochemistry. RESULTS: High methionine diet could cause weight loss, SBP rising, and plasma Hcy content significantly elevated. IL-16 and IL-17A levels in peripheral blood and in brain tissue both lifted, while IL-10 and TGF-ß levels dropped; RORγt expression raised in brain, nevertheless, FoxP3 levels were the opposite. After the intervention with vitamin B6, B12, and folic acid in SHR-T group, these trends would be eased or completely changed. Furthermore, brain tissue slices showed that IL-17-positive cells tended to decrease, and IL-10-positive cells increased in SHR-T group, which was reversed in SHR-M group. CONCLUSIONS: HHcy may promote inflammation that can lead to brain lesions and down-regulate immune response to protect the brain.


Asunto(s)
Lesiones Encefálicas/dietoterapia , Hiperhomocisteinemia/dietoterapia , Inflamación/dietoterapia , Ratas Endogámicas SHR/genética , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/inmunología , Lesiones Encefálicas/etiología , Lesiones Encefálicas/inmunología , Lesiones Encefálicas/patología , Factores de Transcripción Forkhead/genética , Homocisteína/sangre , Humanos , Hiperhomocisteinemia/sangre , Hiperhomocisteinemia/complicaciones , Hiperhomocisteinemia/inmunología , Inmunomodulación/genética , Inmunomodulación/inmunología , Inflamación/etiología , Inflamación/genética , Inflamación/inmunología , Interleucina-10/genética , Interleucina-17/genética , Interleucina-6/genética , Metionina/farmacología , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Ratas , Ratas Endogámicas SHR/sangre , Ratas Endogámicas SHR/inmunología , Linfocitos T Colaboradores-Inductores/efectos de los fármacos , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Reguladores/inmunología , Factor de Crecimiento Transformador beta/genética
7.
J Endocrinol Invest ; 41(7): 831-837, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29288439

RESUMEN

PURPOSE: Hyperhomocysteinemia is a known cardiovascular risk factor and a key player in the inflammatory activation of autoimmune diseases. Hashimoto's thyroiditis (HT) is the leading cause of hypothyroidism which, in itself, has been associated with a significant raise of homocysteine (Hcy) levels and increased cardiovascular risk. Our aim was to assess the impact of HT on Hcy levels in patients with acute hypothyroidism. METHODS: We prospectively enrolled 121 patients (mean age: 46 years, F/M = 102/19) with acute post-surgical hypothyroidism. Based on the presence of anti-thyroid antibodies and the histological description of an inflammatory infiltrate, 26 and 95 patients were classified as HT and non-HT, respectively. Several parameters including thyroid-stimulating hormone (TSH), levels of serum free T3 and free T4, weight, glucose levels, total cholesterol, creatinine, vitamin B12, ferritin and erythrocyte sedimentation rate were obtained from all patients and correlated with Hcy levels. RESULTS: Median Hcy level in the whole cohort was 16.8 µmol/L (normal values: < 12 µmol/l). Among all parameters analysed, only Hcy levels were significantly different between HT and non-HT patients (median Hcy = 19.7 vs 16.2 µmol/L, respectively; p = 0.018, Mann-Whitney U test). Analysis of covariance showed the presence of HT to be the strongest predictor of Hcy levels (coefficient = 0.25534, p = 0.001). Serum TSH was not significantly associated with Hcy levels (p = 0.943). CONCLUSION: In patients with iatrogenic hypothyroidism, those with HT have significantly higher Hcy levels than those without HT. The increase of Hcy levels appears to be mainly determined by the HT-related immune-inflammatory condition.


Asunto(s)
Autoinmunidad , Hiperhomocisteinemia/etiología , Hipotiroidismo/complicaciones , Glándula Tiroides/inmunología , Enfermedad Aguda , Adulto , Femenino , Enfermedad de Hashimoto/complicaciones , Enfermedad de Hashimoto/epidemiología , Enfermedad de Hashimoto/inmunología , Enfermedad de Hashimoto/cirugía , Humanos , Hiperhomocisteinemia/epidemiología , Hiperhomocisteinemia/inmunología , Hipotiroidismo/epidemiología , Hipotiroidismo/inmunología , Enfermedad Iatrogénica/epidemiología , Radioisótopos de Yodo/uso terapéutico , Masculino , Persona de Mediana Edad , Factores de Riesgo , Glándula Tiroides/efectos de la radiación
8.
Mol Cell Biochem ; 433(1-2): 177-194, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28386844

RESUMEN

Although hyperhomocysteinemia (HHcy) is known to promote downstream pro-inflammatory cytokine elevation, the precise mechanism is still unknown. One of the possible receptors that could have significant attention in the field of hypertension is toll-like receptor 4 (TLR-4). TLR-4 is a cellular membrane protein that is ubiquitously expressed in all cell types of the vasculature. Its mutation can attenuate the effects of HHcy-mediated vascular inflammation and mitochondria- dependent cell death that suppresses hypertension. In this review, we observed that HHcy induces vascular remodeling through immunological adaptation, promoting inflammatory cytokine up-regulation (IL-1ß, IL-6, TNF-α) and initiation of mitochondrial dysfunction leading to cell death and chronic vascular inflammation. The literature suggests that HHcy promotes TLR-4-driven chronic vascular inflammation and mitochondria-mediated cell death inducing peripheral vascular remodeling. In the previous studies, we have characterized the role of TLR-4 mutation in attenuating vascular remodeling in hyperhomocysteinemia. This review includes, but is not limited to, the physiological synergistic aspects of the downstream elevation of cytokines found within the vascular inflammatory cascade. These events subsequently induce mitochondrial dysfunction defined by excessive mitochondrial fission and mitochondrial apoptosis contributing to vascular remodeling followed by hypertension.


Asunto(s)
Citocinas/inmunología , Hiperhomocisteinemia/inmunología , Receptor Toll-Like 4/inmunología , Remodelación Vascular/inmunología , Vasculitis/inmunología , Animales , Enfermedad Crónica , Citocinas/genética , Humanos , Hiperhomocisteinemia/genética , Hiperhomocisteinemia/patología , Receptor Toll-Like 4/genética , Vasculitis/genética , Vasculitis/patología
9.
Can J Physiol Pharmacol ; 95(3): 239-246, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27398734

RESUMEN

Although hyperhomocysteinemia (HHcy) is an independent risk factor for cardiovascular diseases (CVD), there is a debate on whether HHcy is a risk factor or just a biomarker. Interestingly, homocysteine lowering strategies in humans had very little effect on reducing the cardiovascular risk, as compared with animals; this may suggest heterogeneity in human population and epigenetic alterations. Moreover, there are only few studies that suggest the idea that HHcy contributes to CVD in the presence of other risk factors such as inflammation, a known risk factor for CVD. Elevated levels of homocysteine have been shown to contribute to inflammation. Here, we highlight possible relationships between homocysteine, T cell immunity, and hypertension, and summarize the evidence that suggested these factors act together in increasing the risk for CVD. In light of this new evidence, we further propose that there is a need for evaluation of the causes of HHcy, defective remethylation or defective transsulfuration, which may differentially modulate hypertension progression, not just the homocysteine levels.


Asunto(s)
Presión Sanguínea , Homocisteína/sangre , Hiperhomocisteinemia/complicaciones , Hipertensión/etiología , Inmunidad Celular , Linfocitos T/inmunología , Animales , Biomarcadores/sangre , Humanos , Hiperhomocisteinemia/sangre , Hiperhomocisteinemia/inmunología , Hipertensión/sangre , Hipertensión/inmunología , Hipertensión/fisiopatología , Mediadores de Inflamación/sangre , Mediadores de Inflamación/inmunología , Activación de Linfocitos , Pronóstico , Medición de Riesgo , Factores de Riesgo , Transducción de Señal , Linfocitos T/metabolismo , Regulación hacia Arriba
10.
Neuroscience ; 341: 42-51, 2017 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-27890830

RESUMEN

Vascular cognitive impairment and dementia (VCID) is the second leading cause of dementia behind Alzheimer's disease (AD) and is a frequent co-morbidity with AD. Despite its prevalence, little is known about the molecular mechanisms underlying the cognitive dysfunction resulting from cerebrovascular disease. Astrocytic end-feet almost completely surround intraparenchymal blood vessels in the brain and express a variety of channels and markers indicative of their specialized functions in the maintenance of ionic and osmotic homeostasis and gliovascular signaling. These functions are mediated by end-foot enrichment of the aquaporin 4 water channel (AQP4), the inward rectifying potassium channel Kir4.1 and the calcium-dependent potassium channel MaxiK. Using our hyperhomocysteinemia (HHcy) model of VCID we examined the time-course of astrocytic end-foot changes along with cognitive and neuroinflammatory outcomes. We found that there were significant astrocytic end-foot disruptions in the HHcy model. AQP4 becomes dislocalized from the end-feet, there is a loss of Kir4.1 and MaxiK protein expression, as well as a loss of the Dp71 protein known to anchor the Kir4.1, MaxiK and AQP4 channels to the end-foot membrane. Neuroinflammation occurs prior to the astrocytic changes, while cognitive impairment continues to decline with the exacerbation of the astrocytic changes. We have previously reported similar astrocytic changes in models of cerebral amyloid angiopathy (CAA) and therefore, we believe astrocytic end-foot disruption could represent a common cellular mechanism of VCID and may be a target for therapeutic development.


Asunto(s)
Astrocitos/inmunología , Astrocitos/patología , Disfunción Cognitiva/inmunología , Disfunción Cognitiva/patología , Demencia Vascular/inmunología , Demencia Vascular/patología , Animales , Encéfalo/irrigación sanguínea , Encéfalo/inmunología , Encéfalo/patología , Antígeno CD11b/metabolismo , Hemorragia Cerebral/inmunología , Hemorragia Cerebral/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Distrofina/metabolismo , Gliosis/inmunología , Gliosis/patología , Hiperhomocisteinemia/inmunología , Hiperhomocisteinemia/patología , Subunidad p35 de la Interleucina-12/metabolismo , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Aprendizaje por Laberinto/fisiología , Ratones Endogámicos C57BL , Canales de Potasio de Rectificación Interna/metabolismo , Factores de Tiempo , Factor de Necrosis Tumoral alfa/metabolismo
11.
J Immunol ; 198(1): 170-183, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-27903739

RESUMEN

The overactivation of immune cells plays an important role in the pathogenesis of hyperhomocysteinemia (HHcy)-accelerated atherosclerosis. Homocysteine (Hcy) activates B cell proliferation and Ab secretion; however, the underlying mechanisms for these effects remain largely unknown. Metabolic reprogramming is critical for lymphocyte activation and effector function. In this study, we showed that Hcy-activated B cells displayed an increase in both oxidative phosphorylation and glycolysis, with a tendency to shift toward the latter, as well as an accumulation of intermediates in the pentose phosphate pathway, to provide energy and biosynthetic substrates for cell growth and function. Mechanistically, Hcy increased both the protein expression and glycolytic enzyme activity of the pyruvate kinase muscle isozyme 2 (PKM2) in B cells, whereas the PKM2 inhibitor shikonin restored Hcy-induced metabolic changes, as well as B cell proliferation and Ab secretion both in vivo and in vitro, indicating that PKM2 plays a critical role in metabolic reprogramming in Hcy-activated B cells. Further investigation revealed that the Akt-mechanistic target of rapamycin signaling pathway was involved in this process, as the mechanistic target of rapamycin inhibitor rapamycin inhibited Hcy-induced changes in PKM2 enzyme activity and B cell activation. Notably, shikonin treatment effectively attenuated HHcy-accelerated atherosclerotic lesion formation in apolipoprotein E-deficient mice. In conclusion, our results demonstrate that PKM2 is required to support metabolic reprogramming for Hcy-induced B cell activation and function, and it might serve as a critical regulator in HHcy-accelerated initiation of atherosclerosis.


Asunto(s)
Linfocitos B/metabolismo , Homocisteína/metabolismo , Piruvato Quinasa/metabolismo , Animales , Aterosclerosis/inmunología , Linfocitos B/inmunología , Cromatografía Liquida , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Técnicas de Silenciamiento del Gen , Homocisteína/inmunología , Hiperhomocisteinemia/inmunología , Hiperhomocisteinemia/metabolismo , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa , Espectrometría de Masas en Tándem
12.
Clin Nutr ; 35(3): 706-12, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26071632

RESUMEN

BACKGROUND & AIMS: Critically ill patients develop severe stress, inflammation and a clinical state that may raise the utilization and metabolic replacement of pyridoxal-5'-phosphate decreasing their body reserves. This study was designed to assess the nutritional pyridoxal-5'-phosphate status in critical care patients with systemic inflammatory response syndrome, comparing them with a group of healthy people, and studying it's association with factors involved in the pyridoxine and other B vitamins metabolism, as the total antioxidant capacity and Hcy as cardiovascular risk biomarker. METHODS: Prospective, multicentre, comparative, observational and analytic study. One hundred and three critically ill patients from different hospitals, and eighty four healthy subjects from Granada, Spain, all with informed consent. Data from daily nutritional assessment, ICU severity scores, clinical and nutritional parameters, antioxidant status and homocysteine levels was taken at admission and at the seventh day of the ICU stay. RESULTS: Thiamine, riboflavin, pyridoxine and folate status proved deficient in a large number of patients, being significantly lower in comparison with control group, and significantly decreased at 7th day of ICU stay. Higher homocysteine was observed in patients compared with control group (p < 0.05) where 31.5 and 26.8 percent of subjects presented hyperhomocysteinemia at initial and final of study, respectively. Antioxidant status was lower than control group in two periods analysed, and decreased at 7th day of ICU stay (p < 0.05) being associated with PLP deficiency. PLP deficiency was also correlated with hyperhomocysteinemia at two times measured (r. -0.73, p < 0.001; r. -0.69, p < 0.001, respectively), showing at day 7 an odds ratio of 6.62 in our multivariate model. CONCLUSION: Critically ill patients with SIRS show deficient B vitamin and low antioxidant statuses. Despite association found between PLP deficiency and low antioxidant status in critically ill patients, PLP deficiency was only associated with hyperhomocysteinemia regardless of antioxidant, riboflavin, cobalamine, and folate statuses in critically ill patients with SIRS at seventh day of ICU stay. PLP deficient patients presented about six times more risk of cardiovascular disease than non deficients.


Asunto(s)
Enfermedades Cardiovasculares/etiología , Enfermedad Crítica , Hiperhomocisteinemia/etiología , Estado Nutricional , Estrés Oxidativo , Fosfato de Piridoxal/deficiencia , Deficiencia de Vitamina B 6/etiología , APACHE , Adulto , Biomarcadores/sangre , Enfermedades Cardiovasculares/epidemiología , Enfermedades Cardiovasculares/inmunología , Enfermedades Cardiovasculares/metabolismo , Femenino , Homocisteína/sangre , Homocisteína/metabolismo , Humanos , Hiperhomocisteinemia/epidemiología , Hiperhomocisteinemia/inmunología , Hiperhomocisteinemia/metabolismo , Unidades de Cuidados Intensivos , Masculino , Persona de Mediana Edad , Prevalencia , Estudios Prospectivos , Fosfato de Piridoxal/metabolismo , Riesgo , España/epidemiología , Deficiencia de Vitamina B 6/epidemiología , Deficiencia de Vitamina B 6/inmunología , Deficiencia de Vitamina B 6/metabolismo
13.
Clin Exp Rheumatol ; 33(6): 900-3, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26690891

RESUMEN

OBJECTIVES: The increase of homocysteine (Hcy) is an independent risk factor related to the development of atherosclerosis (AS) and the cardiovascular disease (CVD) in patients with rheumatoid arthritis (RA). Cardiovascular disease is a leading cause of mortality in RA. The aim of the study was to determine its relationship with homocysteine and the clinical immunological-inflammatory and metabolic laboratory markers and evaluate the factors in relation to hyperhomocysteinaemia in RA. METHODS: Analysis of total serum homocysteine (Hcy) concentrations was carried out in fifty patients with RA compared with 50 matched health controls. In patients with RA, numerous immunological-inflammatory and metabolic laboratory makers included, folate, vitamin B12 complement (i.e. C3 and C4), C-reactive protein (CRP), Rheumatoid factor (RF), anticyclic citrullinated peptide (anti-CCP) antibody, cystatin C (CysC), triglycerides, cholesterol and total leukocyte count. We also assessed imaging makers, common carotid intima-media thickness (IMT) and disease activity makers such as Disease Activity Score in 28 joints (DAS28). RESULTS: Median concentration of Hcy was significantly greater in patients with RA than in controls: 9.09 (5.38-33.91) vs. 7.45 (4.89-25.77) µmol/L (p<0.001). In RA patients, homocysteine was inversely associated with folate (rho=-0.672, p<0.0001), vitamin B12 (rho=-0.424, p=0.002), C3 (rho=-0.612, p<0.0001), C4 (rho=-0.323, p=0.022), correlate with CRP (rho=0.342, p=0.015), CysC (rho=0.430, p=0.002), but not with anti-CCP antibody (rho=0.205, p=0.152), RF (rho=0.214, p=0.135), triglycerides (rho=-0.107, p=0.459), cholesterol (rho=0.160, p=0.268), total leukocyte count (rho=-0.157, p=0.276), IMT (rho=0.134, p=0.156), DAS28 (rho=0.211, p=0.148). There was association between hyperomocysteinaemia in RA with vitamin B12 (p=0.037), folate (p<0.001), and CRP (p=0.018). There was no association with male gender, anti-CCP antibody, RF, IMT and DAS28. CONCLUSIONS: Results from this study suggested that Hcy concentration was increased in RA patients and associated with RA-related immunological-inflammatory and metabolic laboratory markers, including folate, vitamin B12, CRP, CysC, C3, C4. Early determination of Hcy and correlated clinical laboratory markers may be useful to evaluate RA patients with high cardiovascular risk.


Asunto(s)
Artritis Reumatoide , Enfermedades Cardiovasculares/epidemiología , Cistatina C/sangre , Homocisteína/sangre , Hiperhomocisteinemia , Factor Reumatoide/sangre , Artritis Reumatoide/sangre , Artritis Reumatoide/complicaciones , Artritis Reumatoide/diagnóstico , Artritis Reumatoide/epidemiología , Biomarcadores/sangre , Proteína C-Reactiva/metabolismo , Grosor Intima-Media Carotídeo , Femenino , Humanos , Hiperhomocisteinemia/epidemiología , Hiperhomocisteinemia/etiología , Hiperhomocisteinemia/inmunología , Inflamación/inmunología , Masculino , Persona de Mediana Edad , Monitorización Inmunológica/métodos , Péptidos Cíclicos/inmunología , Factores de Riesgo , Estadística como Asunto
14.
Bull Exp Biol Med ; 159(3): 358-60, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26212809

RESUMEN

Moderate exogenous hyperhomocysteinemia is associated with an increase in lymphocyte count (mainly at the expense of cytotoxic T cells), a 2-fold drop of the immunoregulatory index, and increase in TNF-α, IFN-γ, and IL-17α concentrations in the serum and in mononuclear cells. Homocysteine concentration in the mononuclears remains unchanged.


Asunto(s)
Citocinas/metabolismo , Hiperhomocisteinemia/inmunología , Hiperhomocisteinemia/metabolismo , Subgrupos Linfocitarios/metabolismo , Animales , Interferón gamma/metabolismo , Interleucina-17/metabolismo , Masculino , Ratas , Factor de Necrosis Tumoral alfa/metabolismo
15.
Int J Mol Sci ; 16(6): 12560-77, 2015 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-26047341

RESUMEN

Hyperhomocysteinemia (HHcy) is an independent risk factor of atherosclerosis and other cardiovascular diseases. Unfortunately, Hcy-lowering strategies were found to have limited effects in reducing cardiovascular events. The underlying mechanisms remain unclear. Increasing evidence reveals a role of inflammation in the pathogenesis of HHcy. Homocysteine (Hcy) is a precursor of hydrogen sulfide (H2S), which is formed via the transsulfuration pathway catalyzed by cystathionine ß-synthase and cystathionine γ-lyase (CSE) and serves as a novel modulator of inflammation. In the present study, we showed that methionine supplementation induced mild HHcy in mice, associated with the elevations of TNF-α and IL-1ß in the plasma and reductions of plasma H2S level and CSE expression in the peritoneal macrophages. H2S-releasing compound GYY4137 attenuated the increases of TNF-α and IL-1ß in the plasma of HHcy mice and Hcy-treated raw264.7 cells while CSE inhibitor PAG exacerbated it. Moreover, the in vitro study showed that Hcy inhibited CSE expression and H2S production in macrophages, accompanied by the increases of DNA methyltransferase (DNMT) expression and DNA hypermethylation in cse promoter region. DNMT inhibition or knockdown reversed the decrease of CSE transcription induced by Hcy in macrophages. In sum, our findings demonstrate that Hcy may trigger inflammation through inhibiting CSE-H2S signaling, associated with increased promoter DNA methylation and transcriptional repression of cse in macrophages.


Asunto(s)
Cistationina gamma-Liasa/genética , Metilación de ADN/efectos de los fármacos , Homocisteína/farmacología , Hiperhomocisteinemia/inducido químicamente , Mediadores de Inflamación/farmacología , Macrófagos/efectos de los fármacos , Animales , Células Cultivadas , Cistationina gamma-Liasa/metabolismo , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Sulfuro de Hidrógeno/metabolismo , Hiperhomocisteinemia/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Metionina/administración & dosificación , Metionina/efectos adversos , Ratones , Morfolinas/farmacología , Compuestos Organotiofosforados/farmacología , Regiones Promotoras Genéticas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
16.
Diabetes ; 63(12): 4275-90, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25008174

RESUMEN

Hyperhomocysteinemia (HHcy) is associated with increased diabetic cardiovascular diseases. However, the role of HHcy in atherogenesis associated with hyperglycemia (HG) remains unknown. To examine the role and mechanisms by which HHcy accelerates HG-induced atherosclerosis, we established an atherosclerosis-susceptible HHcy and HG mouse model. HHcy was established in mice deficient in cystathionine ß-synthase (Cbs) in which the homocysteine (Hcy) level could be lowered by inducing transgenic human CBS (Tg-hCBS) using Zn supplementation. HG was induced by streptozotocin injection. Atherosclerosis was induced by crossing Tg-hCBS Cbs mice with apolipoprotein E-deficient (ApoE(-/-)) mice and feeding them a high-fat diet for 2 weeks. We demonstrated that HHcy and HG accelerated atherosclerosis and increased lesion monocytes (MCs) and macrophages (MØs) and further increased inflammatory MC and MØ levels in peripheral tissues. Furthermore, Hcy-lowering reversed circulating mononuclear cells, MC, and inflammatory MC and MC-derived MØ levels. In addition, inflammatory MC correlated positively with plasma Hcy levels and negatively with plasma s-adenosylmethionine-to-s-adenosylhomocysteine ratios. Finally, l-Hcy and d-glucose promoted inflammatory MC differentiation in primary mouse splenocytes, which was reversed by adenoviral DNA methyltransferase-1. HHcy and HG, individually and synergistically, accelerated atherosclerosis and inflammatory MC and MØ differentiation, at least in part, via DNA hypomethylation.


Asunto(s)
Aterosclerosis/inmunología , Diferenciación Celular/inmunología , Hiperglucemia/inmunología , Hiperhomocisteinemia/inmunología , Macrófagos/inmunología , Monocitos/inmunología , Animales , Apolipoproteínas E/genética , Aterosclerosis/complicaciones , Cistationina betasintasa/genética , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Humanos , Hiperglucemia/complicaciones , Hiperhomocisteinemia/complicaciones , Inflamación/inmunología , Ratones , Ratones Transgénicos
17.
Eur Rev Med Pharmacol Sci ; 18(3): 359-64, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24563435

RESUMEN

AIM: The current study aims to explore the possible molecular mechanism of hyperhomocysteinemia (HHcy) mediated atherosclerosis (AS) and to find an effective intervention method for AS. MATERIALS AND METHODS: A total of 40 Wistar rats were equalized into four groups: blank control, HHcy, folacin intervention, and lutein intervention groups. HHcy rat models were established. The intervention groups were respectively lavaged with folacin and lutein. Oxidative stress states, the levels of nitric oxide (NO) and endothelin-1 (ET-1), as well as the expression of nuclear factor (NF)-κB p65 and intercellular adhesion molecule (ICAM)-1 were compared. RESULTS: In the HHcy rats, the activity of serum superoxide dismutase (SOD) and glutathione peroxidase (GPx) significantly decreased, whereas the malondialdehyde content and hydroxyl radical level noticeably increased, indicating that the rats stayed in aggravated oxidative stress states. Lutein intervention inhibited HHcy-induced oxidative stress excitement. In the HHcy rats, the NO level significantly decreased, whereas the ET-1 level significantly increased, indicating that HHcy mediated vascular endothelial dysfunction. Lutein reversed such dysfunction. In the HHcy rats, the mRNA and protein expression of SOD2 and GPX1 in the aortic wall tissue decreased, whereas that of NF-κB p65 and ICAM-1 increased. Lutein significantly upregulated the mRNA and protein expression of SOD2 and GPx1 and downregulated the expression of NF-κB p65 and ICAM-1. CONCLUSIONS: Oxidative stress and inflammation are the important mechanisms of HHcy-mediated AS. In particular, HHcy-induced aggravated oxidative stress may function as the initial AS-mediating mechanism, upregulating the expression of NF-κB p65 and ICAM-1 and thereby becoming associated with AS. Lutein noticeably intervenes in and inhibits Hcy-mediated oxidative stress excitement and downregulates the expression of inflammation-associated informational molecules.


Asunto(s)
Antioxidantes/uso terapéutico , Aterosclerosis/prevención & control , Hiperhomocisteinemia/tratamiento farmacológico , Luteína/uso terapéutico , Estrés Oxidativo/efectos de los fármacos , Animales , Antioxidantes/administración & dosificación , Aterosclerosis/etiología , Aterosclerosis/inmunología , Aterosclerosis/metabolismo , Modelos Animales de Enfermedad , Regulación hacia Abajo , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/inmunología , Endotelio Vascular/metabolismo , Hiperhomocisteinemia/complicaciones , Hiperhomocisteinemia/inmunología , Hiperhomocisteinemia/metabolismo , Molécula 1 de Adhesión Intercelular/biosíntesis , Luteína/administración & dosificación , Masculino , Ratas Wistar , Factor de Transcripción ReIA/biosíntesis
18.
Lik Sprava ; (5): 54-9, 2013.
Artículo en Ucraniano | MEDLINE | ID: mdl-24605633

RESUMEN

In article described research the results of the prevalence of the genetic polymorphism of the gene Methylentetrahydrofolatereductase C677T (MTHFR) in 130 patients with pseudarthrosis of long bones and in those with consolidated fractures. The incidence of allele-T among patients with pseudarthrosis was 1.4 times higher than among those with consolidated fractures. Pathological genotype MTHFR 677-TT was associated with the development avital types of pseudarthrosis and increase the proportion of people with hyperhomocysteinemia, high content of inflammatory mediators and development refracture.


Asunto(s)
Hiperhomocisteinemia/enzimología , Mediadores de Inflamación/sangre , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Polimorfismo Genético , Seudoartrosis/enzimología , Fracturas de la Tibia/enzimología , Adulto , Proteína C-Reactiva/inmunología , Femenino , Curación de Fractura/genética , Curación de Fractura/fisiología , Frecuencia de los Genes , Heterocigoto , Fracturas de Cadera/enzimología , Fracturas de Cadera/epidemiología , Homocisteína/sangre , Homocigoto , Humanos , Hiperhomocisteinemia/complicaciones , Hiperhomocisteinemia/epidemiología , Hiperhomocisteinemia/inmunología , Mediadores de Inflamación/inmunología , Interleucina-6/sangre , Interleucina-6/inmunología , Masculino , Seudoartrosis/complicaciones , Seudoartrosis/epidemiología , Seudoartrosis/inmunología , Fracturas de la Tibia/complicaciones , Fracturas de la Tibia/epidemiología , Fracturas de la Tibia/inmunología
19.
Eur J Nutr ; 52(3): 1243-50, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22899103

RESUMEN

PURPOSE: Hyperhomocysteinemia is well recognized as an independent risk factor for the development of premature atherosclerosis. Atherosclerosis, however, may be prevented by polyphenols, potent antioxidant compounds with anti-atherogenic properties. Previously, we used cystathionine beta synthase-deficient mice [Cbs (±)] fed a high-methionine diet-a murine model of hyperhomocysteinemia-to show that daily intake of a red wine polyphenolic extract, mainly comprised of catechin and epicatechin, has a beneficial effect on aortic expression of endothelial dysfunction biomarkers and pro-inflammatory cytokines. The aim of the present study was to understand whether catechin and epicatechin, in purified forms, have anti-atherogenic effects in hyperhomocysteinemia. METHODS: Cbs (±) mice received 50 µg of catechin and/or epicatechin daily in drinking water for 1 month. Plasma homocysteine (Hcy) level and aortic expression of several endothelial dysfunction biomarkers (Vcam-1, Icam-1, E-selectin, and Lox-1) and pro-inflammatory cytokines (Tnf-α, Il-6) were assessed. RESULTS: We found that both catechin and epicatechin had a beneficial effect on plasma homocysteine levels and endothelial dysfunction biomarker expression; however, only catechin had a beneficial effect on pro-inflammatory cytokine expression. Further, when both polyphenols were given, a beneficial effect was observed only on pro-inflammatory cytokine expression. CONCLUSIONS: Catechin seems to be a more potent anti-atherogenic compound than epicatechin in hyperhomocysteinemia and should be considered as a novel therapeutic approach against endothelial dysfunction induced by this condition.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Antioxidantes/uso terapéutico , Aorta/fisiopatología , Catequina/uso terapéutico , Citocinas/metabolismo , Endotelio Vascular/fisiopatología , Hiperhomocisteinemia/dietoterapia , Animales , Antiinflamatorios no Esteroideos/química , Antioxidantes/química , Aorta/inmunología , Aorta/metabolismo , Aterosclerosis/etiología , Aterosclerosis/prevención & control , Biomarcadores/metabolismo , Catequina/análogos & derivados , Cruzamientos Genéticos , Cistationina betasintasa/genética , Cistationina betasintasa/metabolismo , Suplementos Dietéticos , Regulación hacia Abajo , Endotelio Vascular/inmunología , Endotelio Vascular/metabolismo , Femenino , Hiperhomocisteinemia/inmunología , Hiperhomocisteinemia/metabolismo , Hiperhomocisteinemia/fisiopatología , Metionina/efectos adversos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Estereoisomerismo
20.
Circ Res ; 111(10): 1261-73, 2012 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-22912384

RESUMEN

RATIONALE: A number of epidemiological studies have suggested an association of hyperhomocysteinemia (HHcy) and abdominal aortic aneurysm (AAA), but discrepancies exist. In addition, we lack direct evidence supporting a causal role. OBJECTIVE: We determined the association and contribution of HHcy to AAA formation. METHODS AND RESULTS: We first performed a meta-analysis of studies involving 1489 subjects and found a strong association of HHcy and AAA (odds ratio, 7.39). Next, we used angiotensin II-infused male apolipoprotein E-deficient mice and tested whether HHcy contributes to AAA pathogenesis. Homocysteine (Hcy) supplement (1.8 g/L) in drinking water resulted in mild HHcy. Intriguingly, HHcy greatly increased the incidence of angiotensin II-induced AAA and aortic dissection in apolipoprotein E-deficient mice (vehicle versus Hcy: 50% versus 100%; P<0.05). Histology indicated HHcy markedly exaggerated aortic adventitial inflammation. Increased levels of proinflammatory interleukin-6 and monocyte chemoattractant protein-1 were preferentially colocalized within adventitial fibroblasts in HHcy plus angiotensin II mice, which suggested the importance of adventitial fibroblasts activation in Hcy-aggravated AAA. Hcy sequentially stimulated adventitial fibroblasts transformation into myofibroblasts, secretion of interleukin-6 and monocyte chemoattractant protein-1, and consequent recruitment of monocytes/macrophages to adventitial fibroblasts, which was abolished by the NADPH oxidase inhibitor diphenyliodonium. NADPH oxidase 4, but not other homologs of NADPH oxidase, was significantly upregulated by Hcy in adventitial fibroblasts, whereas NADPH oxidase 4 small interfering RNA silencing diminished Hcy-induced adventitial fibroblasts activation. Finally, folic acid supplement (0.071 µg/g per day) markedly reduced HHcy-aggravated angiotensin II-induced AAA formation in apolipoprotein E-deficient mice. CONCLUSIONS: HHcy may aggravate AAA formation at least partially via activating adventitial fibroblast NADPH oxidase 4.


Asunto(s)
Aneurisma de la Aorta Abdominal/epidemiología , Aneurisma de la Aorta Abdominal/inmunología , Hiperhomocisteinemia/epidemiología , Hiperhomocisteinemia/inmunología , Vasculitis/epidemiología , Vasculitis/inmunología , Adventicia/inmunología , Angiotensina II/farmacología , Animales , Aneurisma de la Aorta Abdominal/inducido químicamente , Apolipoproteínas E/genética , Quimiocina CCL2/metabolismo , Modelos Animales de Enfermedad , Femenino , Fibroblastos/enzimología , Fibroblastos/inmunología , Fibroblastos/metabolismo , Humanos , Hiperhomocisteinemia/metabolismo , Incidencia , Interleucina-6/metabolismo , Masculino , Ratones , Ratones Mutantes , Persona de Mediana Edad , NADPH Oxidasa 4 , NADPH Oxidasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Factores de Riesgo , Transducción de Señal/inmunología , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Vasculitis/metabolismo , Vasoconstrictores/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA