Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Am J Pathol ; 191(5): 947-964, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33640319

RESUMO

This study investigated the effects of long-term NF-κB inhibition in mitigating retinal vasculopathy in a type 1 diabetic mouse model (Akita, Ins2Akita). Akita and wild-type (C57BL/6J) male mice, 24 to 26 weeks old, were treated with or without a selective inhibitor of NF-κB, 4-methyl-N1-(3-phenyl-propyl) benzene-1,2-diamine (JSH-23), for 4 weeks. Treatment was given when the mice were at least 24 weeks old. Metabolic parameters, key inflammatory mediators, blood-retinal barrier junction molecules, retinal structure, and function were measured. JSH-23 significantly lowered basal glucose levels and intraocular pressure in Akita. It also mitigated vascular remodeling and microaneurysms significantly. Optical coherence tomography of untreated Akita showed thinning of retinal layers; however, treatment with JSH-23 could prevent it. Electroretinogram demonstrated that A- and B-waves in Akita were significantly smaller than in wild type mice, indicating that JSH-23 intervention prevented loss of retinal function. Protein levels and gene expression of key inflammatory mediators, such as NOD-like receptor family pyrin domain-containing 3, intercellular adhesion molecule-1, inducible nitric oxide synthase, and cyclooxygenase-2, were decreased after JSH-23 treatment. At the same time, connexin-43 and occludin were maintained. Vision-guided behavior also improved significantly. The results show that reducing inflammation could protect the diabetic retina and its vasculature. Findings appear to have broader implications in treating not only ocular conditions but also other vasculopathies.


Assuntos
Diabetes Mellitus Experimental/complicações , Inflamação/patologia , NF-kappa B/antagonistas & inibidores , Fenilenodiaminas/farmacologia , Doenças Retinianas/prevenção & controle , Doenças Vasculares/prevenção & controle , Animais , Apoptose , Glicemia/análise , Modelos Animais de Doenças , Eletrorretinografia , Humanos , Hiperglicemia/patologia , Leucócitos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação , NF-kappa B/metabolismo , Retina/diagnóstico por imagem , Retina/patologia , Doenças Retinianas/diagnóstico por imagem , Doenças Retinianas/etiologia , Doenças Retinianas/patologia , Vasos Retinianos/diagnóstico por imagem , Vasos Retinianos/patologia , Tomografia de Coerência Óptica , Doenças Vasculares/diagnóstico por imagem , Doenças Vasculares/etiologia , Doenças Vasculares/patologia
2.
Mol Cell Biochem ; 476(2): 663-673, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33074445

RESUMO

Epigenetic memory plays crucial roles in gene regulation. It not only modulates the expression of specific genes but also has ripple effects on transcription as well as translation of other genes. Very often an alteration in expression occurs either via methylation or demethylation. In this context, "1-carbon metabolism" assumes a special significance since its dysregulation by higher levels of homocysteine; Hcy (known as hyperhomocysteinemia; HHcy), a byproduct of "1-Carbon Metabolism" during methionine biosynthesis leads to serious implications in cardiovascular, renal, cerebrovascular systems, and a host of other conditions. Currently, the circular RNAs (circRNAs) generated via non-canonical back-splicing events from the pre-mRNA molecules are at the center stage for their essential roles in diseases via their epigenetic manifestations. We recently identified a circular RNA transcript (circGRM4) that is significantly upregulated in the eye of cystathionine ß-synthase-deficient mice. We also discovered a concurrent over-expression of the mGLUR4 receptor in the eyes of these mice. In brief, circGRM4 is selectively transcribed from its parental mGLUR4 receptor gene (GRM4) functions as a "molecular-sponge" for the miRNAs and results into excessive turnover of the mGLUR4 receptor in the eye in response to extremely high circulating glutamate concentration. We opine that this epigenetic manifestation potentially predisposes HHcy people to retinovascular malfunctioning.


Assuntos
Cistationina beta-Sintase/genética , Olho/irrigação sanguínea , Olho/metabolismo , Ácido Glutâmico/metabolismo , MicroRNAs/metabolismo , RNA Circular/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Animais , Cistationina beta-Sintase/metabolismo , Células Endoteliais/metabolismo , Epigênese Genética , Oftalmopatias/induzido quimicamente , Oftalmopatias/genética , Oftalmopatias/metabolismo , Oftalmopatias/patologia , Homocisteína/metabolismo , Humanos , Hiper-Homocisteinemia/genética , MicroRNAs/genética , RNA Circular/genética , Receptores de Glutamato Metabotrópico/genética , Doenças Vasculares/induzido quimicamente , Doenças Vasculares/genética , Doenças Vasculares/metabolismo , Doenças Vasculares/patologia
3.
Can J Physiol Pharmacol ; 99(1): 115-123, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32721223

RESUMO

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.


Assuntos
Cistationina beta-Sintase/deficiência , Hiper-Homocisteinemia/complicações , Periodontite/imunologia , Periodonto/patologia , Animais , Cistationina beta-Sintase/genética , Modelos Animais de Doenças , Ácido Fólico , Homocisteína/sangue , Homocisteína/metabolismo , Humanos , Hiper-Homocisteinemia/sangue , Hiper-Homocisteinemia/imunologia , Hiper-Homocisteinemia/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Estresse Oxidativo/imunologia , Periodontite/patologia , Periodonto/imunologia
4.
Can J Physiol Pharmacol ; 99(2): 161-170, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32721225

RESUMO

Research demonstrates that senescence is associated with tissue and organ dysfunction, and the eye is no exception. Sequelae arising from aging have been well defined as distinct clinical entities and vision impairment has significant psychosocial consequences. Retina and adjacent tissues like retinal pigmented epithelium and choroid are the key structures that are required for visual perception. Any structural and functional changes in retinal layers and blood retinal barrier could lead to age-related macular degeneration, diabetic retinopathy, and glaucoma. Further, there are significant oxygen gradients in the eye that can lead to excessive reactive oxygen species, resulting in endoplasmic reticulum and mitochondrial stress response. These radicals are source of functional and morphological impairment in retinal pigmented epithelium and retinal ganglion cells. Therefore, ocular diseases could be summarized as disturbance in the redox homeostasis. Hyperhomocysteinemia is a risk factor and causes vascular occlusive disease of the retina. Interestingly, hydrogen sulfide (H2S) has been proven to be an effective antioxidant agent, and it can help treat diseases by alleviating stress and inflammation. Concurrent glutamate excitotoxicity, endoplasmic reticulum stress, and microglia activation are also linked to stress; thus, H2S may offer additional interventional strategy. A refined understanding of the aging eye along with H2S biology and pharmacology may help guide newer therapies for the eye.


Assuntos
Envelhecimento/fisiologia , Olho/efeitos dos fármacos , Sulfeto de Hidrogênio/farmacologia , Envelhecimento/efeitos dos fármacos , Animais , Humanos
5.
Can J Physiol Pharmacol ; 99(1): 56-63, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32799662

RESUMO

Epigenetic DNA methylation (1-carbon metabolism) is crucial for gene imprinting/off-printing that ensures epigenetic memory but also generates a copious amount of homocysteine (Hcy), unequivocally. That is why during pregnancy, expectant mothers are recommended "folic acid" preemptively to avoid birth defects in the young ones because of elevated Hcy levels (i.e., hyperhomocysteinemia (HHcy)). As we know, children born with HHcy have several musculoskeletal abnormalities, including growth retardation. Here, we focus on the gut-dysbiotic microbiome implication(s) that we believe instigates the "1-carbon metabolism" and HHcy causing growth retardation along with skeletal muscle abnormalities. We test our hypothesis whether high-methionine diet (HMD) (an amino acid that is high in red meat), a substrate for Hcy, can cause skeletal muscle and growth retardation, and treatment with probiotics (PB) to mitigate skeletal muscle dysfunction. To test this, we employed cystathionine ß-synthase, CBS deficient mouse (CBS+/-) fed with/without HMD and with/without a probiotic (Lactobacillus rhamnosus) in drinking water for 16 weeks. Matrix metalloproteinase (MMP) activity, a hallmark of remodeling, was measured by zymography. Muscle functions were scored via electric stimulation. Our results suggest that compared to the wild-type, CBS+/- mice exhibited reduced growth phenotype. MMP-2 activity was robust in CBS+/- and HMD effects were successfully attenuated by PB intervention. Electrical stimulation magnitude was decreased in CBS+/- and CBS+/- treated with HMD. Interestingly; PB mitigated skeletal muscle growth retardation and atrophy. Collectively, results imply that individuals with mild/moderate HHcy seem more prone to skeletal muscle injury and its dysfunction.


Assuntos
Disbiose/complicações , Transtornos do Crescimento/prevenção & controle , Hiper-Homocisteinemia/complicações , Músculo Esquelético/patologia , Probióticos/administração & dosagem , Animais , Cistationina beta-Sintase/deficiência , Cistationina beta-Sintase/genética , Metilação de DNA , Modelos Animais de Doenças , Disbiose/metabolismo , Disbiose/microbiologia , Disbiose/terapia , Epigênese Genética , Feminino , Microbioma Gastrointestinal/fisiologia , Transtornos do Crescimento/sangue , Transtornos do Crescimento/metabolismo , Transtornos do Crescimento/patologia , Homocisteína/sangue , Homocisteína/metabolismo , Humanos , Hiper-Homocisteinemia/sangue , Hiper-Homocisteinemia/genética , Hiper-Homocisteinemia/metabolismo , Lacticaseibacillus rhamnosus , Masculino , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Metionina/administração & dosagem , Metionina/metabolismo , Camundongos , Camundongos Transgênicos , Músculo Esquelético/metabolismo
6.
Can J Physiol Pharmacol ; 99(1): 9-17, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32706987

RESUMO

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.


Assuntos
Disbiose/complicações , Microbioma Gastrointestinal/fisiologia , Homocisteína/metabolismo , Hiper-Homocisteinemia/imunologia , Periodontite/imunologia , Disbiose/sangue , Disbiose/imunologia , Disbiose/microbiologia , Ácido Fólico/sangue , Ácido Fólico/metabolismo , Homocisteína/sangue , Homocisteína/imunologia , Humanos , Hiper-Homocisteinemia/sangue , Hiper-Homocisteinemia/metabolismo , Metionina/metabolismo , Periodontite/sangue , Periodontite/metabolismo , Probióticos
7.
Int J Mol Sci ; 22(24)2021 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-34948342

RESUMO

Although blood-heart-barrier (BHB) leakage is the hallmark of congestive (cardio-pulmonary) heart failure (CHF), the primary cause of death in elderly, and during viral myocarditis resulting from the novel coronavirus variants such as the severe acute respiratory syndrome novel corona virus 2 (SARS-CoV-2) known as COVID-19, the mechanism is unclear. The goal of this project is to determine the mechanism of the BHB in CHF. Endocardial endothelium (EE) is the BHB against leakage of blood from endocardium to the interstitium; however, this BHB is broken during CHF. Previous studies from our laboratory, and others have shown a robust activation of matrix metalloproteinase-9 (MMP-9) during CHF. MMP-9 degrades the connexins leading to EE dysfunction. We demonstrated juxtacrine coupling of EE with myocyte and mitochondria (Mito) but how it works still remains at large. To test whether activation of MMP-9 causes EE barrier dysfunction, we hypothesized that if that were the case then treatment with hydroxychloroquine (HCQ) could, in fact, inhibit MMP-9, and thus preserve the EE barrier/juxtacrine signaling, and synchronous endothelial-myocyte coupling. To determine this, CHF was created by aorta-vena cava fistula (AVF) employing the mouse as a model system. The sham, and AVF mice were treated with HCQ. Cardiac hypertrophy, tissue remodeling-induced mitochondrial-myocyte, and endothelial-myocyte contractions were measured. Microvascular leakage was measured using FITC-albumin conjugate. The cardiac function was measured by echocardiography (Echo). Results suggest that MMP-9 activation, endocardial endothelial leakage, endothelial-myocyte (E-M) uncoupling, dyssynchronous mitochondrial fusion-fission (Mfn2/Drp1 ratio), and mito-myocyte uncoupling in the AVF heart failure were found to be rampant; however, treatment with HCQ successfully mitigated some of the deleterious cardiac alterations during CHF. The findings have direct relevance to the gamut of cardiac manifestations, and the resultant phenotypes arising from the ongoing complications of COVID-19 in human subjects.


Assuntos
COVID-19/complicações , Insuficiência Cardíaca/metabolismo , Coração/virologia , Animais , Sangue/virologia , Fenômenos Fisiológicos Sanguíneos/imunologia , COVID-19/fisiopatologia , Cardiomegalia/metabolismo , Doenças Cardiovasculares/metabolismo , Fenômenos Fisiológicos Cardiovasculares/imunologia , Modelos Animais de Doenças , Endotélio/metabolismo , Coração/fisiopatologia , Insuficiência Cardíaca/virologia , Hidroxicloroquina/farmacologia , Masculino , Metaloproteinase 9 da Matriz/efeitos dos fármacos , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Células Musculares/metabolismo , Miocárdio/metabolismo , SARS-CoV-2/metabolismo , SARS-CoV-2/patogenicidade , Remodelação Ventricular/fisiologia
8.
J Cell Physiol ; 235(3): 2590-2598, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31489638

RESUMO

Unless there is a genetic defect/mutation/deletion in a gene, the causation of a given disease is chronic dysregulation of gut metabolism. Most of the time, if not always, starts within the gut; that is what we eat. Recent research shows that the imbalance between good versus bad microbial population, especially in the gut, causes systemic diseases. Thus, an appropriate balance of the gut microbiota (eubiosis over dysbiosis) needs to be maintained for normal health (Veeranki and Tyagi, 2017, Journal of Cellular Physiology, 232, 2929-2930). However, during various diseases such as metabolic syndrome, inflammatory bowel disease, diabetes, obesity, and hypertension the dysbiotic gut environment tends to prevail. Our research focuses on homocysteine (Hcy) metabolism that occupies a center-stage in many biochemically relevant epigenetic mechanisms. For example, dysbiotic bacteria methylate promoters to inhibit gene activities. Interestingly, the product of the 1-carbon metabolism is Hcy, unequivocally. Emerging studies show that host resistance to various antibiotics occurs due to inverton promoter inhibition, presumably because of promoter methylation. This results from modification of host promoters by bacterial products leading to loss of host's ability to drug compatibility and system sensitivity. In this study, we focus on the role of high methionine diet (HMD), an ingredient rich in red meat and measure the effects of a probiotic on cardiac muscle remodeling and its functions. We employed wild type (WT) and cystathionine beta-synthase heterozygote knockout (CBS+/- ) mice with and without HMD and with and without a probiotic; PB (Lactobacillus) in drinking water for 16 weeks. Results indicate that matrix metalloproteinase-2 (MMP-2) activity was robust in CBS+/- fed with HMD and that it was successfully attenuated by the PB treatment. Cardiomyocyte contractility and ECHO data revealed mitigation of the cardiac dysfunction in CBS+/- + HMD mice treated with PB. In conclusion, our data suggest that probiotics can potentially reverse the Hcy-meditated cardiac dysfunction.


Assuntos
Cistationina beta-Sintase/genética , Disbiose/metabolismo , Microbioma Gastrointestinal/genética , Homocisteína/metabolismo , Animais , Carbono/metabolismo , Modelos Animais de Doenças , Disbiose/genética , Disbiose/microbiologia , Epigênese Genética/genética , Homocisteína/genética , Humanos , Lactobacillus/efeitos dos fármacos , Lactobacillus/metabolismo , Metaloproteinase 2 da Matriz/genética , Camundongos , Camundongos Knockout , Miocárdio/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Probióticos/farmacologia
9.
Can J Physiol Pharmacol ; 98(2): 51-60, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31369712

RESUMO

Homocysteine (Hcy), a sulfur-containing nonproteinogenic amino acid, is generated as a metabolic intermediate. Hcy constitutes an important part of the "1-carbon metabolism" during methionine turnover. Elevated levels of Hcy known as hyperhomocysteinemia (HHcy) results from vitamin B deficiency, lack of exercise, smoking, excessive alcohol intake, high-fat and methionine-rich diet, and the underlying genetic defects. These factors directly affect the "1-carbon metabolism (methionine-Hcy-folate)" of a given cell. In fact, the Hcy levels are determined primarily by dietary intake, vitamin status, and the genetic blueprint of the susceptible individual. Although Hcy performs an important role in cellular functions, genetic alterations in any of the key enzymes responsible for the "1-carbon metabolism" could potentially upset the metabolic cycle, thus causing HHcy environment in susceptible people. As such, HHcy relates to several clinical conditions like atherosclerosis, myocardial infarction, stroke, cognitive impairment, dementia, Parkinson's disease, multiple sclerosis, epilepsy, and ocular disorders, among others. This article summarizes the findings from our laboratory and public database regarding genetics of HHcy and its effects on ocular disorders, their respective management during dysregulation of the 1-carbon metabolism.


Assuntos
Carbono/metabolismo , Hiper-Homocisteinemia/genética , Hiper-Homocisteinemia/metabolismo , Retina/patologia , Retina/fisiopatologia , Animais , Humanos , Hiper-Homocisteinemia/patologia , Hiper-Homocisteinemia/fisiopatologia
10.
Physiol Genomics ; 51(12): 613-622, 2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-31709889

RESUMO

Retinal degeneration from inherited gene mutation(s) is a common cause of blindness because of structural and functional alterations in photoreceptors. Accordingly, various approaches are being tested to ameliorate or even cure neuroretinal blinding conditions in susceptible patients by employing neuroprotective agents, gene therapeutics, optogenetics, regenerative therapies, and retinal prostheses. The FVB/NJ mouse strain inherently has a common Pde6b rd1 homozygous allele that renders its progeny blind by the time pups reach weaning age. To study the role matrix metalloproteinase-9 (MMP-9) in retinal structure and function, we examined a global MMP-9 knockout (KO) mouse model that has been engineered on the same FVB/NJ background to test the hypothesis whether lack of MMP-9 activity diminishes neuroretinal degenerative changes and thus helps improve the vision. We compared side-by-side various aspects of the ocular physiology in the wild-type (WT) C57BL/6J, FVB/NJ, and MMP-9 KO strains of mice. The results suggest that MMP-9 KO mice display subdued changes in their retinae as reflected by both structural and functional enhancement in the overall ocular neurophysiological parameters. Altogether, the findings appear to have clinical relevance for targeting conditions wherein MMPs and their overactivities are suspected to play dominant pathophysiological roles in advancing neurodegenerative retinal diseases.


Assuntos
Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Retina/patologia , Degeneração Retiniana/genética , Degeneração Retiniana/fisiopatologia , Remodelação Vascular/genética , Animais , Modelos Animais de Doenças , Feminino , Técnicas de Inativação de Genes , Homeostase/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Inibidor Tecidual de Metaloproteinase-1/metabolismo , Visão Ocular/genética
11.
J Cell Biochem ; 120(1): 77-92, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30272816

RESUMO

Higher levels of nonprotein amino acid homocysteine (Hcy), that is, hyperhomocysteinemia (HHcy) (~5% of general population) has been associated with severe vasculopathies in different organs; however, precise molecular mechanism(s) as to how HHcy plays havoc with body's vascular networks are largely unknown. Interventional modalities have not proven beneficial to counter multifactorial HHcy's effects on the vascular system. An ancient Indian form of exercise called 'yoga' causes transient ischemia as a result of various body postures however the cellular mechanisms are not clear. We discuss a novel perspective wherein we argue that application of remote ischemic conditioning (RIC) could, in fact, deliver anticipated results to patients who are suffering from chronic vascular dysfunction due to HHcy. RIC is the mechanistic phenomenon whereby brief episodes of ischemia-reperfusion events are applied to distant tissues/organs; that could potentially offer a powerful tool in mitigating chronic lethal ischemia in target organs during HHcy condition via simultaneous reduction of inflammation, oxidative and endoplasmic reticulum stress, extracellular matrix remodeling, fibrosis, and angiogenesis. We opine that during ischemic conditioning our organs cross talk by releasing cellular messengers in the form of exosomes containing messenger RNAs, circular RNAs, anti-pyroptotic factors, protective cytokines like musclin, transcription factors, small molecules, anti-inflammatory, antiapoptotic factors, antioxidants, and vasoactive gases. All these could help mobilize the bone marrow-derived stem cells (having tissue healing properties) to target organs. In that context, we argue that RIC could certainly play a savior's role in an unfortunate ischemic or adverse event in people who have higher levels of the circulating Hcy in their systems.


Assuntos
Homocisteína/metabolismo , Hiper-Homocisteinemia/metabolismo , Hiper-Homocisteinemia/terapia , Reperfusão/métodos , Doenças Vasculares/metabolismo , Doenças Vasculares/terapia , Animais , Citocinas/metabolismo , Estresse do Retículo Endoplasmático , Homocisteína S-Metiltransferase/metabolismo , Humanos , Hiper-Homocisteinemia/complicações , Inflamação/metabolismo , Isquemia/metabolismo , Células-Tronco Mesenquimais/metabolismo , Camundongos , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo , Doenças Vasculares/complicações
12.
Rev Cardiovasc Med ; 20(3): 121-128, 2019 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-31601086

RESUMO

Recent studies have shown that the integrity of the gastrointestinal tract and its microbiome impact the functioning of various body systems by regulating immunological responses, extracting energy, remodeling intestinal epithelia, and strengthening the gut itself. The gastrointestinal tract microbiota includes bacteria, fungi, protozoa, viruses, and archaea which collectively comprise a dynamic community prone to alterations via influences such as the environment, illness, and metabolic processes. The idea that the host's diet possesses characteristics that could potentially alter microbiota composition is a novel notion. We hypothesize that a high fat diet leads to the alteration of the gastrointestinal microbiota composition and that metabolic transformation of the compound trimethylamine into trimethylamine-N-oxide promotes vasculopathy such as atherosclerosis and affects cardiovascular functionality. Furthermore, we hypothesize that treatment with probiotics will restore the homeostatic environment (eubiosis) of the gastrointestinal tract.


Assuntos
Aterosclerose/metabolismo , Bactérias/metabolismo , Dieta Hiperlipídica/efeitos adversos , Endotélio Vascular/metabolismo , Microbioma Gastrointestinal , Metilaminas/metabolismo , Animais , Aterosclerose/microbiologia , Aterosclerose/fisiopatologia , Aterosclerose/terapia , Bactérias/crescimento & desenvolvimento , Disbiose , Endotélio Vascular/fisiopatologia , Interações Hospedeiro-Patógeno , Humanos , Lipídeos/sangue , Placa Aterosclerótica , Probióticos/uso terapêutico , Fatores de Risco
13.
Can J Physiol Pharmacol ; 97(6): 441-456, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30422673

RESUMO

Elevated homocysteine (Hcy), i.e., hyperhomocysteinemia (HHcy), causes skeletal muscle myopathy. Among many cellular and metabolic alterations caused by HHcy, oxidative and endoplasmic reticulum (ER) stress are considered the major ones; however, the precise molecular mechanism(s) in this process is unclear. Nevertheless, there is no treatment option available to treat HHcy-mediated muscle injury. Hydrogen sulfide (H2S) is increasingly recognized as a potent anti-oxidant, anti-apoptotic/necrotic/pyroptotic, and anti-inflammatory compound and also has been shown to improve angiogenesis during ischemic injury. Patients with CBS mutation produce less H2S, making them vulnerable to Hcy-mediated cellular damage. Many studies have reported bidirectional regulation of ER stress in apoptosis through JNK activation and concomitant attenuation of cell proliferation and protein synthesis via PI3K/AKT axis. Whether H2S mitigates these detrimental effects of HHcy on muscle remains unexplored. In this review, we discuss molecular mechanisms of HHcy-mediated oxidative/ER stress responses, apoptosis, angiogenesis, and atrophic changes in skeletal muscle and how H2S can restore skeletal muscle homeostasis during HHcy condition. This review also highlights the molecular mechanisms on how H2S could be developed as a clinically relevant therapeutic option for chronic conditions that are aggravated by HHcy.


Assuntos
Estresse do Retículo Endoplasmático , Homeostase , Sulfeto de Hidrogênio/metabolismo , Hiper-Homocisteinemia/metabolismo , Músculo Esquelético/metabolismo , Estresse Oxidativo , Animais , Humanos , Hiper-Homocisteinemia/patologia , Músculo Esquelético/patologia
14.
Can J Physiol Pharmacol ; 97(11): 1013-1017, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31269408

RESUMO

Homocysteine, a non-proteinogenic amino acid but an important metabolic intermediate is generated as an integral component for the "1-carbon metabolism" during normal physiology. It is catabolized to cysteine via the transulfuration pathway resulting in the generation of hydrogen sulfide, a naturally endogenous byproduct. Genetics or metabolic derangement can alter homocysteine concentration leading to hyperhomocysteinemia (HHcy), a physiologically unfavorable condition that causes serious medical conditions including muscle wasting. HHcy environment can derail physiological processes by targeting biomolecules such as Akt; however, not much is known regarding the effects of HHcy on regulation of transcription factors such as forkhead box O (FOXO) proteins. Recently, hydrogen sulfide has been shown to be highly effective in alleviating the effects of HHcy by serving as an antiapoptotic factor, but role of FOXO and its interaction with hydrogen sulfide are yet to be established. In this review, we discuss role(s) of HHcy in skeletal muscle atrophy and how HHcy interact with FOXO and peroxisome proliferator-activated receptor gamma coactivator 1-alpha expressions that are relevant in musculoskeletal atrophy. Further, therapeutic intervention with hydrogen sulfide for harnessing its beneficial effects might help mitigate the dysregulated 1-carbon metabolism that happens to be the hallmark of HHcy-induced pathologies such as muscle atrophy.


Assuntos
Carbono/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Atrofia Muscular/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Animais , Humanos , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Atrofia Muscular/patologia
15.
Can J Physiol Pharmacol ; 97(6): 463-472, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30444648

RESUMO

Circular RNAs (circRNAs) are being hailed as a newly rediscovered class of covalently closed transcripts that are produced via alternative, noncanonical pre-mRNA back-splicing events. These single-stranded RNA molecules have been identified in organisms ranging from the worm (Cortés-López et al. 2018. BMC Genomics, 19: 8; Ivanov et al. 2015. Cell Rep. 10: 170-177) to higher eukaryotes (Yang et al. 2017. Cell Res. 27: 626-641) to plants (Li et al. 2017. Biochem. Biophys. Res. Commun. 488: 382-386). At present, research on circRNAs is an active area because of their diverse roles in development, health, and diseases. Partly because their circularity makes them resistant to degradation, they hold great promise as unique biomarkers for ocular and central nervous system (CNS) disorders. We believe that further work on their applications could help in developing them as "first-in-class" diagnostics, therapeutics, and prognostic targets for numerous eye conditions. Interestingly, many circRNAs play key roles in transcriptional regulation by acting as miRNAs sponges, meaning that they serve as master regulators of RNA and protein expression. Since the retina is an extension of the brain and is part of the CNS, we highlight the current state of circRNA biogenesis, properties, and function and we review the crucial roles that they play in the eye and the brain. We also discuss their regulatory roles as miRNA sponges, regulation of their parental genes or linear mRNAs, translation into micropeptides or proteins, and responses to cellular stress. We posit that future advances will provide newer insights into the fields of RNA metabolism in general and diseases of the aging eye and brain in particular. Furthermore, in keeping pace with the rapidly evolving discipline of RNA"omics"-centered metabolism and to achieve uniformity among researchers, we recently introduced the term "cromics" (circular ribonucleic acids based omics) (Singh et al. 2018. Exp. Eye Res. 174: 80-92).


Assuntos
Encéfalo/metabolismo , Olho/metabolismo , Regulação da Expressão Gênica , Mamíferos/genética , RNA/genética , RNA/metabolismo , Animais , Humanos , RNA/biossíntese , RNA Circular
16.
Am J Physiol Cell Physiol ; 315(5): C609-C622, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30110564

RESUMO

Although hyperhomocysteinemia (HHcy) occurs because of the deficiency in cystathionine-ß-synthase (CBS) causing skeletal muscle dysfunction, it is still unclear whether this effect is mediated through oxidative stress, endoplasmic reticulum (ER) stress, or both. Nevertheless, there is no treatment option available to improve HHcy-mediated muscle injury. Hydrogen sulfide (H2S) is an antioxidant compound, and patients with CBS mutation do not produce H2S. In this study, we hypothesized that H2S mitigates HHcy-induced redox imbalance/ER stress during skeletal muscle atrophy via JNK phosphorylation. We used CBS+/- mice to study HHcy-mediated muscle atrophy, and treated them with sodium hydrogen sulfide (NaHS; an H2S donor). Proteins and mRNAs were examined by Western blots and quantitative PCR. Proinflammatory cytokines were also measured. Muscle mass and strength were studied via fatigue susceptibility test. Our data revealed that HHcy was detrimental to skeletal mass, particularly gastrocnemius and quadriceps muscle weight. We noticed that oxidative stress was reversed by NaHS in homocysteine (Hcy)-treated C2C12 cells. Interestingly, ER stress markers (GRP78, ATF6, pIRE1α, and pJNK) were elevated in vivo and in vitro, and NaHS mitigated these effects. Additionally, we observed that JNK phosphorylation was upregulated in C2C12 after Hcy treatment, but NaHS could not reduce this effect. Furthermore, inflammatory cytokines IL-6 and TNF-α were higher in plasma from CBS as compared with wild-type mice. FOXO1-mediated Atrogin-1 and MuRF-1 upregulation were attenuated by NaHS. Functional studies revealed that NaHS administration improved muscle fatigability in CBS+/- mice. In conclusion, our work provides evidence that NaHS is beneficial in mitigating HHcy-mediated skeletal injury incited by oxidative/ER stress responses.


Assuntos
Cistationina beta-Sintase/genética , Hiper-Homocisteinemia/tratamento farmacológico , Atrofia Muscular/tratamento farmacológico , Sulfitos/administração & dosagem , Fator 6 Ativador da Transcrição/genética , Animais , Antioxidantes/administração & dosagem , Chaperona BiP do Retículo Endoplasmático , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Endorribonucleases/genética , Proteína Forkhead Box O1/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas de Choque Térmico/genética , Humanos , Hiper-Homocisteinemia/sangue , Hiper-Homocisteinemia/complicações , Hiper-Homocisteinemia/genética , Interleucina-6/sangue , MAP Quinase Quinase 4/genética , Camundongos , Proteínas Musculares/genética , Força Muscular/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/patologia , Atrofia Muscular/sangue , Atrofia Muscular/etiologia , Atrofia Muscular/genética , Estresse Oxidativo/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/genética , Proteínas Ligases SKP Culina F-Box/genética , Proteínas com Motivo Tripartido/genética , Fator de Necrose Tumoral alfa/sangue , Ubiquitina-Proteína Ligases/genética
17.
J Cell Physiol ; 233(4): 3080-3092, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-28833102

RESUMO

Homocysteine (Hcy) causes endothelial dysfunction by inducing oxidative stress in most neurodegenerative disorders. This dysfunction is highly correlated with mitochondrial dynamics such as fusion and fission. However, there are no strategies to prevent Hcy-induced mitochondrial remodeling. Tetrahydrocurcumin (THC) is an anti-inflammatory and anti-oxidant compound. We hypothesized that THC may ameliorates Hcy-induced mitochondria remodeling in mouse brain endothelial cells (bEnd3) cells. bEnd3 cells were exposed to Hcy treatment in the presence or absence of THC. Cell viability and autophagic cell death were measured with MTT and MDC staining assay. Reactive oxygen species (ROS) production was determined using DCFH-DA staining by confocal microscopy. Autophagy flux was assessed using a conventional GFP-microtubule-associated protein 1 light chain 3 (LC3) dot assay. Interaction of phagophore marker LC-3 with mitochondrial receptor NIX was observed by confocal imaging. Mitochondrial fusion and fission were evaluated by western blot and RT-PCR. Our results demonstrated that Hcy resulted in cell toxicity in a dose-dependent manner and supplementation of THC prevented the detrimental effects of Hcy on cell survival. Furthermore, Hcy also upregulated fission marker (DRP-1), fusion marker (Mfn2), and autophagy marker (LC-3). Finally, we observed that Hcy activated mitochondrial specific phagophore marker (LC-3) and co-localized with the mitochondrial receptor NIX, as viewed by confocal microscopy. Pretreatment of bEnd3 with THC (15 µM) ameliorated Hcy-induced oxidative damage, mitochondrial fission/fusion, and mitophagy. Our studies strongly suggest that THC has beneficial effects on mitochondrial remodeling and could be developed as a potential therapeutic agent against hyperhomocysteinemia (HHcy) induced mitochondrial dysfunction.


Assuntos
Curcumina/análogos & derivados , Células Endoteliais/metabolismo , Homocisteína/toxicidade , Mitocôndrias/metabolismo , Animais , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Encéfalo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Curcumina/farmacologia , Citoproteção/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Espaço Intracelular/metabolismo , Camundongos , Mitocôndrias/efeitos dos fármacos , Dinâmica Mitocondrial/efeitos dos fármacos , Proteínas Mitocondriais/metabolismo , Modelos Biológicos , Espécies Reativas de Oxigênio/metabolismo
18.
Exp Eye Res ; 174: 80-92, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29803556

RESUMO

Cystathionine-ß-synthase (CBS) gene encodes L-serine hydrolyase which catalyzes ß-reaction to condense serine with homocysteine (Hcy) by pyridoxal-5'-phosphate helps to form cystathionine which in turn is converted to cysteine. CBS resides at the intersection of transmethylation, transsulfuration, and remethylation pathways, thus lack of CBS fundamentally blocks Hcy degradation; an essential step in glutathione synthesis. Redox homeostasis, free-radical detoxification and one-carbon metabolism (Methionine-Hcy-Folate cycle) require CBS and its deficiency leads to hyperhomocysteinemia (HHcy) causing retinovascular thromboembolism and eye-lens dislocation along with vascular cognitive impairment and dementia. HHcy results in retinovascular, coronary, cerebral and peripheral vessels' dysfunction and how it causes metabolic dysregulation predisposing patients to serious eye conditions remains unknown. HHcy orchestrates inflammation and redox imbalance via epigenetic remodeling leading to neurovascular pathologies. Although circular RNAs (circRNAs) are dominant players regulating their parental genes' expression dynamics, their importance in ocular biology has not been appreciated. Progress in gene-centered analytics via improved microarray and bioinformatics are enabling dissection of genomic pathways however there is an acute under-representation of circular RNAs in ocular disorders. This study undertook circRNAs' analysis in the eyes of CBS deficient mice identifying a pool of 12532 circRNAs, 74 exhibited differential expression profile, ∼27% were down-regulated while most were up-regulated (∼73%). Findings also revealed several microRNAs that are specific to each circRNA suggesting their roles in HHcy induced ocular disorders. Further analysis of circRNAs helped identify novel parental genes that seem to influence certain eye disease phenotypes.


Assuntos
Cistationina beta-Sintase/genética , Hiper-Homocisteinemia/metabolismo , Subluxação do Cristalino/metabolismo , RNA/metabolismo , Animais , Cistationina beta-Sintase/metabolismo , Epigenômica , Perfilação da Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Estresse Oxidativo/fisiologia , RNA Circular
19.
Alcohol Clin Exp Res ; 40(12): 2474-2481, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27805256

RESUMO

Alcohol is the most socially accepted addictive drug. Alcohol consumption is associated with some health problems such as neurological, cognitive, behavioral deficits, cancer, heart, and liver disease. Mechanisms of alcohol-induced toxicity are presently not yet clear. One of the mechanisms underlying alcohol toxicity has to do with its interaction with amino acid homocysteine (Hcy), which has been linked with brain neurotoxicity. Elevated Hcy impairs with various physiological mechanisms in the body, especially metabolic pathways. Hcy metabolism is predominantly controlled by epigenetic regulation such as DNA methylation, histone modifications, and acetylation. An alteration in these processes leads to epigenetic modification. Therefore, in this review, we summarize the role of Hcy metabolism abnormalities in alcohol-induced toxicity with epigenetic adaptation and their influences on cerebrovascular pathology.


Assuntos
Alcoolismo/genética , Epigênese Genética/efeitos dos fármacos , Etanol/toxicidade , Homocisteína/metabolismo , Hiper-Homocisteinemia/induzido quimicamente , Hiper-Homocisteinemia/metabolismo , Redes e Vias Metabólicas/efeitos dos fármacos , Redes e Vias Metabólicas/genética , Animais , Humanos , Modelos Biológicos , Modelos Neurológicos
20.
Mol Neurobiol ; 58(8): 3614-3627, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33774742

RESUMO

Traumatic brain injury (TBI) is a damage to the brain from an external force that results in temporary or permanent impairment in brain functions. Unfortunately, not many treatment options are available to TBI patients. Therefore, knowledge of the complex interplay between gut microbiome (GM) and brain health may shed novel insights as it is a rapidly expanding field of research around the world. Recent studies show that GM plays important roles in shaping neurogenerative processes such as blood-brain-barrier (BBB), myelination, neurogenesis, and microglial maturation. In addition, GM is also known to modulate many aspects of neurological behavior and cognition; however, not much is known about the role of GM in brain injuries. Since GM has been shown to improve cellular and molecular functions via mitigating TBI-induced pathologies such as BBB permeability, neuroinflammation, astroglia activation, and mitochondrial dysfunction, herein we discuss how a dysbiotic gut environment, which in fact, contributes to central nervous system (CNS) disorders during brain injury and how to potentially ward off these harmful effects. We further opine that a better understanding of GM-brain (GMB) axis could help assist in designing better treatment and management strategies in future for the patients who are faced with limited options.


Assuntos
Lesões Encefálicas Traumáticas/metabolismo , Eixo Encéfalo-Intestino/fisiologia , Encéfalo/metabolismo , Disbiose/metabolismo , Microbioma Gastrointestinal/fisiologia , Animais , Encéfalo/imunologia , Lesões Encefálicas Traumáticas/imunologia , Lesões Encefálicas Traumáticas/terapia , Disbiose/imunologia , Disbiose/terapia , Humanos , Mediadores da Inflamação/imunologia , Mediadores da Inflamação/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA