Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 161
Filter
Add more filters

Complementary Medicines
Publication year range
1.
Biofactors ; 50(1): 181-200, 2024.
Article in English | MEDLINE | ID: mdl-37650587

ABSTRACT

In the brain, the non-essential amino acid L-serine is produced through the phosphorylated pathway (PP) starting from the glycolytic intermediate 3-phosphoglycerate: among the different roles played by this amino acid, it can be converted into D-serine and glycine, the two main co-agonists of NMDA receptors. In humans, the enzymes of the PP, namely phosphoglycerate dehydrogenase (hPHGDH, which catalyzes the first and rate-limiting step of this pathway), 3-phosphoserine aminotransferase, and 3-phosphoserine phosphatase are likely organized in the cytosol as a metabolic assembly (a "serinosome"). The hPHGDH deficiency is a pathological condition biochemically characterized by reduced levels of L-serine in plasma and cerebrospinal fluid and clinically identified by severe neurological impairment. Here, three single-point variants responsible for hPHGDH deficiency and Neu-Laxova syndrome have been studied. Their biochemical characterization shows that V261M, V425M, and V490M substitutions alter either the kinetic (both maximal activity and Km for 3-phosphoglycerate in the physiological direction) and the structural properties (secondary, tertiary, and quaternary structure, favoring aggregation) of hPHGDH. All the three variants have been successfully ectopically expressed in U251 cells, thus the pathological effect is not due to hindered expression level. At the cellular level, mistargeting and aggregation phenomena have been observed in cells transiently expressing the pathological protein variants, as well as a reduced L-serine cellular level. Previous studies demonstrated that the pharmacological supplementation of L-serine in hPHGDH deficiencies could ameliorate some of the related symptoms: our results now suggest the use of additional and alternative therapeutic approaches.


Subject(s)
Brain Diseases , Glyceric Acids , Serine , Humans , Serine/genetics , Phosphoglycerate Dehydrogenase/genetics , Phosphoglycerate Dehydrogenase/chemistry , Brain Diseases/metabolism , Amino Acids
2.
Int J Mol Sci ; 23(10)2022 May 13.
Article in English | MEDLINE | ID: mdl-35628259

ABSTRACT

BAM15 (a mitochondrial uncoupling agent) was tested on cecal ligation and puncture (CLP) sepsis mice with in vitro experiments. BAM15 attenuated sepsis as indicated by survival, organ histology (kidneys and livers), spleen apoptosis (activated caspase 3), brain injury (SHIRPA score, serum s100ß, serum miR370-3p, brain miR370-3p, brain TNF-α, and apoptosis), systemic inflammation (cytokines, cell-free DNA, endotoxemia, and bacteremia), and blood-brain barrier (BBB) damage (Evan's blue dye and the presence of green fluorescent E. coli in brain after an oral administration). In parallel, brain miR arrays demonstrated miR370-3p at 24 h but not 120 h post-CLP, which was correlated with metabolic pathways. Either lipopolysaccharide (LPS) or TNF-α upregulated miR370-3p in PC12 (neuron cells). An activation by sepsis factors (LPS, TNF-α, or miR370-3p transfection) damaged mitochondria (fluorescent color staining) and reduced cell ATP, possibly through profound mitochondrial activity (extracellular flux analysis) that was attenuated by BAM15. In bone-marrow-derived macrophages, LPS caused mitochondrial injury, decreased cell ATP, enhanced glycolysis activity (extracellular flux analysis), and induced pro-inflammatory macrophages (iNOS and IL-1ß) which were neutralized by BAM15. In conclusion, BAM15 attenuated sepsis through decreased mitochondrial damage, reduced neuronal miR370-3p upregulation, and induced anti-inflammatory macrophages. BAM15 is proposed to be used as an adjuvant therapy against sepsis hyperinflammation.


Subject(s)
Brain Diseases , MicroRNAs , Sepsis , Adenosine Triphosphate/genetics , Adenosine Triphosphate/metabolism , Animals , Brain Diseases/genetics , Brain Diseases/metabolism , Lipopolysaccharides/administration & dosage , Male , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Punctures , Sepsis/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
3.
Pediatr Res ; 91(3): 598-605, 2022 02.
Article in English | MEDLINE | ID: mdl-33953355

ABSTRACT

BACKGROUND: Infants with moderate and severe neonatal encephalopathy (NE) frequently suffer from long-term adverse outcomes. We hypothesize that the urinary metabolome of newborns with NE reflects the evolution of injury patterns observed with magnetic resonance imaging (MRI). METHODS: Eligible patients were newborn infants with perinatal asphyxia evolving to NE and qualifying for therapeutic hypothermia (TH) included in the HYPOTOP trial. MRI was employed for characterizing brain injury. Urine samples of 55 infants were collected before, during, and after TH. Metabolic profiles of samples were recorded employing three complementary mass spectrometry-based assays, and the alteration of detected metabolic features between groups was assessed. RESULTS: The longitudinal assessment revealed significant perturbations of the urinary metabolome. After 24 h of TH, a stable disease pattern evolved characterized by the alterations of 4-8% of metabolic features related to lipid metabolism, metabolism of cofactors and vitamins, glycan biosynthesis and metabolism, amino acid metabolism, and nucleotide metabolism. Characteristic metabolomic fingerprints were observed for different MRI injury patterns. CONCLUSIONS: This study shows the potential of urinary metabolic profiles for the noninvasive monitoring of brain injury of infants with NE during TH. IMPACT: A comprehensive approach for the study of the urinary metabolome was employed involving a semi-targeted capillary electrophoresis-time-of-flight mass spectrometry (TOFMS) assay, an untargeted ultra-performance liquid chromatography (UPLC)-quadrupole TOFMS assay, and a targeted UPLC-tandem MS-based method for the quantification of amino acids. The longitudinal study of the urinary metabolome identified dynamic metabolic changes between birth and until 96 h after the initiation of TH. The identification of altered metabolic pathways in newborns with pathologic MRI outcomes might offer the possibility of developing noninvasive monitoring approaches for personalized adjustment of the treatment and for supporting early outcome prediction.


Subject(s)
Asphyxia Neonatorum , Brain Injuries , Hypothermia, Induced , Asphyxia Neonatorum/metabolism , Asphyxia Neonatorum/urine , Brain Diseases/metabolism , Brain Diseases/urine , Brain Injuries/metabolism , Brain Injuries/urine , Female , Humans , Infant , Infant, Newborn , Longitudinal Studies , Metabolome , Metabolomics/methods , Pregnancy
4.
Molecules ; 26(15)2021 Aug 03.
Article in English | MEDLINE | ID: mdl-34361850

ABSTRACT

Celastrol, the most abundant compound derived from the root of Tripterygium wilfordii, largely used in traditional Chinese medicine, has shown preclinical and clinical efficacy for a broad range of disorders, acting via numerous mechanisms, including the induction of the expression of several neuroprotective factors, the inhibition of cellular apoptosis, and the decrease of reactive oxygen species (ROS). Given the crucial implication of these pathways in the pathogenesis of Central Nervous System disorders, both in vitro and in vivo studies have focused their attention on the possible use of this compound in these diseases. However, although most of the available studies have reported significant neuroprotective effects of celastrol in cellular and animal models of these pathological conditions, some of these data could not be replicated. This review aims to discuss current in vitro and in vivo lines of evidence on the therapeutic potential of celastrol in neurodegenerative diseases, including Alzheimer's and Parkinson's diseases, amyotrophic lateral sclerosis, Huntington's disease, multiple sclerosis, and cadmium-induced neurodegeneration, as well as in psychiatric disorders, such as psychosis and depression. In vitro and in vivo studies focused on celastrol effects in cerebral ischemia, ischemic stroke, traumatic brain injury, and epilepsy are also described.


Subject(s)
Brain Diseases/drug therapy , Pentacyclic Triterpenes , Plant Roots/chemistry , Tripterygium/chemistry , Animals , Brain Diseases/metabolism , Brain Diseases/pathology , Humans , Neuroprotective Agents/chemistry , Neuroprotective Agents/therapeutic use , Pentacyclic Triterpenes/chemistry , Pentacyclic Triterpenes/therapeutic use
5.
Neurochem Int ; 149: 105124, 2021 10.
Article in English | MEDLINE | ID: mdl-34245808

ABSTRACT

Brain and neuronal circuits constitute the most complex organ networks in human body. They not only control and coordinate functions of all other organs, but also represent one of the most-affected systems with stress, lifestyle and age. With global increase in aging populations, these neuropathologies have emerged as major concern for maintaining quality of life. Recent era has witnessed a surge in nutritional remediation of brain dysfunctions primarily by "nutraceuticals" that refer to functional foods and supplements with pharmacological potential. Specific dietary patterns with a balanced intake of carbohydrates, fatty acids, vitamins and micronutrients have also been ascertained to promote brain health. Dietary herbs and their phytochemicals with wide range of biological and pharmacological activities and minimal adverse effects have gained remarkable attention as neuro-nutraceuticals. Neuro-nutraceutical potentials of herbs are often expressed as effects on cognitive response, circadian rhythm, neuromodulatory, antioxidant and anti-inflammatory activities that are mediated by effects on gene expression, epigenetics, protein synthesis along with their turnover and metabolic pathways. Epidemiological and experimental evidence have implicated enormous applications of herbal supplementation in neurodegenerative and psychiatric disorders. The present review highlights the identification, experimental evidence and applications of some herbs including Bacopa monniera, Withania somnifera, Curcuma longa, Helicteres angustifolia, Undaria pinnatifida, Haematococcus pluvialis, and Vitis vinifera, as neuro-nutraceuticals.


Subject(s)
Antioxidants/therapeutic use , Brain Diseases/drug therapy , Brain/drug effects , Dietary Supplements , Nootropic Agents/therapeutic use , Plant Preparations/therapeutic use , Animals , Antioxidants/isolation & purification , Antioxidants/pharmacology , Brain/metabolism , Brain Diseases/metabolism , Humans , Nootropic Agents/isolation & purification , Nootropic Agents/pharmacology , Phytochemicals/isolation & purification , Phytochemicals/pharmacology , Phytochemicals/therapeutic use , Plant Preparations/isolation & purification , Plant Preparations/pharmacology
6.
Front Endocrinol (Lausanne) ; 12: 668396, 2021.
Article in English | MEDLINE | ID: mdl-34122343

ABSTRACT

In mammals, myeloid cells help maintain the homeostasis of peripheral metabolic tissues, and their immunologic dysregulation contributes to the progression of obesity and associated metabolic disease. There is accumulating evidence that innate immune cells also serve as functional regulators within the mediobasal hypothalamus (MBH), a critical brain region controlling both energy and glucose homeostasis. Specifically, microglia, the resident parenchymal myeloid cells of the CNS, play important roles in brain physiology and pathology. Recent studies have revealed an expanding array of microglial functions beyond their established roles as immune sentinels, including roles in brain development, circuit refinement, and synaptic organization. We showed that microglia modulate MBH function by transmitting information resulting from excess nutrient consumption. For instance, microglia can sense the excessive consumption of saturated fats and instruct neurons within the MBH accordingly, leading to responsive alterations in energy balance. Interestingly, the recent emergence of high-resolution single-cell techniques has enabled specific microglial populations and phenotypes to be profiled in unprecedented detail. Such techniques have highlighted specific subsets of microglia notable for their capacity to regulate the expression of lipid metabolic genes, including lipoprotein lipase (LPL), apolipoprotein E (APOE) and Triggering Receptor Expressed on Myeloid Cells 2 (TREM2). The discovery of this transcriptional signature highlights microglial lipid metabolism as a determinant of brain health and disease pathogenesis, with intriguing implications for the treatment of brain disorders and potentially metabolic disease. Here we review our current understanding of how changes in microglial lipid metabolism could influence the hypothalamic control of systemic metabolism.


Subject(s)
Brain Diseases/pathology , Homeostasis , Hypothalamus/pathology , Lipid Metabolism , Lipids/analysis , Metabolic Diseases/pathology , Microglia/metabolism , Animals , Brain Diseases/etiology , Brain Diseases/metabolism , Humans , Hypothalamus/metabolism , Metabolic Diseases/etiology , Metabolic Diseases/metabolism
7.
Mol Med Rep ; 24(2)2021 Aug.
Article in English | MEDLINE | ID: mdl-34132368

ABSTRACT

Although diabetic encephalopathy (DE) is a major late complication of diabetes, the pathophysiology of postural instability in DE remains poorly understood. Prior studies have suggested that neuronal apoptosis is closely associated with cognitive function, but the mechanism remains to be elucidated. Green tea, which is a non­fermented tea, contains a number of tea polyphenols, alkaloids, amino acids, polysaccharides and other components. Some studies have found that drinking green tea can reduce the incidence of neurodegenerative diseases and improve cognitive dysfunction. We previously found that myosin light chain kinase (MLCK) regulates apoptosis in high glucose­induced hippocampal neurons. In neurodegenerative diseases, including Alzheimer's disease and Parkinson's disease, activation of the JNK signaling pathway promotes neuronal apoptosis. However, the relationship between JNK and MLCK remains to be elucidated. Green tea serum was obtained using seropharmacological methods and applied to hippocampal neurons. In addition, a type 1 diabetes rat model was established and green tea extract was administered, and the Morris water maze test, Cell Counting Kit­8 assays, flow cytometry, western blotting and terminal deoxynucleotidyl transferase­mediated dUTP nick end­labelling assays were used to examine the effects of green tea on hippocampal neuronal apoptosis in diabetic rats. The results demonstrated that green tea can protect against hippocampal neuronal apoptosis by inhibiting the JNK/MLCK pathway and ultimately improves cognitive function in diabetic rats. The present study provided novel insights into the neuroprotective effects of green tea.


Subject(s)
Apoptosis/drug effects , Brain Diseases/metabolism , Diabetes Mellitus, Experimental/metabolism , Hippocampus/drug effects , MAP Kinase Signaling System/drug effects , Neurons/drug effects , Plant Extracts/pharmacology , Animals , Brain Diseases/drug therapy , Cells, Cultured , Cognitive Dysfunction/drug therapy , Diabetes Mellitus, Experimental/complications , Hippocampus/cytology , Hippocampus/metabolism , In Situ Nick-End Labeling , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Maze Learning/drug effects , Models, Animal , Myosin-Light-Chain Kinase/metabolism , Neurons/metabolism , Neuroprotective Agents/pharmacology , Rats , Rats, Sprague-Dawley , Tea/chemistry
8.
Int J Mol Sci ; 22(4)2021 Feb 11.
Article in English | MEDLINE | ID: mdl-33670407

ABSTRACT

Natural compounds such as herbal medicines and/or phyto-compounds from foods, have frequently been used to exert synergistic therapeutic effects with anti-brain disorder drugs, supplement the effects of nutrients, and boost the immune system. However, co-administration of natural compounds with the drugs can cause synergistic toxicity or impeditive drug interactions due to changes in pharmacokinetic properties (e.g., absorption, metabolism, and excretion) and various drug transporters, particularly brain transporters. In this review, natural compound-drug interactions (NDIs), which can occur during the treatment of brain disorders, are emphasized from the perspective of pharmacokinetics and cellular transport. In addition, the challenges emanating from NDIs and recent approaches are discussed.


Subject(s)
Blood-Brain Barrier/metabolism , Brain Diseases/drug therapy , Membrane Transport Proteins/metabolism , Phytochemicals , Plants, Medicinal , Animals , Biological Transport , Blood-Brain Barrier/pathology , Brain Diseases/metabolism , Brain Diseases/pathology , Drug Interactions , Humans , Phytochemicals/agonists , Phytochemicals/antagonists & inhibitors , Phytochemicals/pharmacokinetics , Phytochemicals/therapeutic use
9.
Pharmacol Res ; 168: 105581, 2021 06.
Article in English | MEDLINE | ID: mdl-33781873

ABSTRACT

In-depth characterization of heart-brain communication in critically ill patients with severe acute respiratory failure is attracting significant interest in the COronaVIrus Disease 19 (COVID-19) pandemic era during intensive care unit (ICU) stay and after ICU or hospital discharge. Emerging research has provided new insights into pathogenic role of the deregulation of the heart-brain axis (HBA), a bidirectional flow of information, in leading to severe multiorgan disease syndrome (MODS) in patients with confirmed infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Noteworthy, HBA dysfunction may worsen the outcome of the COVID-19 patients. In this review, we discuss the critical role HBA plays in both promoting and limiting MODS in COVID-19. We also highlight the role of HBA as new target for novel therapeutic strategies in COVID-19 in order to open new translational frontiers of care. This is a translational perspective from the Italian Society of Cardiovascular Researches.


Subject(s)
Brain Diseases/therapy , Brain/drug effects , COVID-19/therapy , Heart Diseases/therapy , Heart/drug effects , Adrenal Cortex Hormones/administration & dosage , Anti-Inflammatory Agents/administration & dosage , Antiviral Agents/administration & dosage , Brain/immunology , Brain/metabolism , Brain Diseases/immunology , Brain Diseases/metabolism , COVID-19/immunology , COVID-19/metabolism , Critical Care/methods , Critical Illness/therapy , Dietary Supplements , Functional Food , Heart Diseases/immunology , Heart Diseases/metabolism , Humans , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Microvessels/drug effects , Microvessels/immunology , Microvessels/metabolism , Multiple Organ Failure/immunology , Multiple Organ Failure/metabolism , Multiple Organ Failure/therapy , SARS-CoV-2/drug effects , SARS-CoV-2/immunology , SARS-CoV-2/metabolism
10.
Int J Mol Sci ; 22(2)2021 Jan 15.
Article in English | MEDLINE | ID: mdl-33467663

ABSTRACT

Neonatal hypoxic-ischemic (HI) brain injury is one of the major drawbacks of mortality and causes significant short/long-term neurological dysfunction in newborn infants worldwide. To date, due to multifunctional complex mechanisms of brain injury, there is no well-established effective strategy to completely provide neuroprotection. Although therapeutic hypothermia is the proven treatment for hypoxic-ischemic encephalopathy (HIE), it does not completely chang outcomes in severe forms of HIE. Therefore, there is a critical need for reviewing the effective therapeutic strategies to explore the protective agents and methods. In recent years, it is widely believed that there are neuroprotective possibilities of natural compounds extracted from plants against HIE. These natural agents with the anti-inflammatory, anti-oxidative, anti-apoptotic, and neurofunctional regulatory properties exhibit preventive or therapeutic effects against experimental neonatal HI brain damage. In this study, it was aimed to review the literature in scientific databases that investigate the neuroprotective effects of plant extracts/plant-derived compounds in experimental animal models of neonatal HI brain damage and their possible underlying molecular mechanisms of action.


Subject(s)
Hypothermia, Induced/methods , Hypoxia-Ischemia, Brain/drug therapy , Neuroprotective Agents/therapeutic use , Plant Extracts/therapeutic use , Animals , Animals, Newborn , Apoptosis , Biological Products/therapeutic use , Brain Diseases/metabolism , Brain Injuries/drug therapy , Cytokines/metabolism , Disease Models, Animal , Free Radicals , Humans , Inflammation , Mice , Neurons/metabolism , Oxidative Stress , Polyphenols/chemistry , Rats , Swine
11.
J Nutr ; 150(Suppl 1): 2506S-2517S, 2020 10 01.
Article in English | MEDLINE | ID: mdl-33000152

ABSTRACT

The metabolism of sulfur-containing amino acids (SAAs) requires an orchestrated interplay among several dozen enzymes and transporters, and an adequate dietary intake of methionine (Met), cysteine (Cys), and B vitamins. Known human genetic disorders are due to defects in Met demethylation, homocysteine (Hcy) remethylation, or cobalamin and folate metabolism, in Hcy transsulfuration, and Cys and hydrogen sulfide (H2S) catabolism. These disorders may manifest between the newborn period and late adulthood by a combination of neuropsychiatric abnormalities, thromboembolism, megaloblastic anemia, hepatopathy, myopathy, and bone and connective tissue abnormalities. Biochemical features include metabolite deficiencies (e.g. Met, S-adenosylmethionine (AdoMet), intermediates in 1-carbon metabolism, Cys, or glutathione) and/or their accumulation (e.g. S-adenosylhomocysteine, Hcy, H2S, or sulfite). Treatment should be started as early as possible and may include a low-protein/low-Met diet with Cys-enriched amino acid supplements, pharmacological doses of B vitamins, betaine to stimulate Hcy remethylation, the provision of N-acetylcysteine or AdoMet, or experimental approaches such as liver transplantation or enzyme replacement therapy. In several disorders, patients are exposed to long-term markedly elevated Met concentrations. Although these conditions may inform on Met toxicity, interpretation is difficult due to the presence of additional metabolic changes. Two disorders seem to exhibit Met-associated toxicity in the brain. An increased risk of demyelination in patients with Met adenosyltransferase I/III (MATI/III) deficiency due to biallelic mutations in the MATIA gene has been attributed to very high blood Met concentrations (typically >800 µmol/L) and possibly also to decreased liver AdoMet synthesis. An excessively high Met concentration in some patients with cystathionine ß-synthase deficiency has been associated with encephalopathy and brain edema, and direct toxicity of Met has been postulated. In summary, studies in patients with various disorders of SAA metabolism showed complex metabolic changes with distant cellular consequences, most of which are not attributable to direct Met toxicity.


Subject(s)
Amino Acids, Sulfur/metabolism , Cysteine/metabolism , Homocysteine/metabolism , Metabolic Diseases/genetics , Methionine/metabolism , Sulfur Compounds/metabolism , Sulfur/metabolism , Animals , Brain Diseases/etiology , Brain Diseases/metabolism , Glutathione/metabolism , Homocystinuria/etiology , Homocystinuria/metabolism , Humans , Hydrogen Sulfide/metabolism , Liver/metabolism , Metabolic Diseases/metabolism , Metabolic Diseases/pathology , Metabolic Diseases/therapy , Metabolism, Inborn Errors/pathology , Metabolism, Inborn Errors/therapy , Methionine Adenosyltransferase/metabolism , Methylation , S-Adenosylmethionine/metabolism , Sulfites/metabolism
12.
Brain Dev ; 42(9): 691-695, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32773162

ABSTRACT

INTRODUCTION: KCNT2 was recently recognized as a gene associated with neurodevelopmental disorder and epilepsy. CASE REPORT: We present an additional observation of a 16-year-old male patient with a novel de novo KCNT2 likely pathogenic variant and review the five previously reported cases of de novo variants in this gene. DISCUSSION: Whole exome sequencing identified the missense variant c.725C > A p.(Thr242Asn), which was confirmed by Sanger sequencing. Our patient has a refractory stereotyped and monomorphic type of hyperkinetic focal motor seizure, similar to what is seen in frontal lobe epilepsy, occurring only during sleep. This type of seizure is not usually seen in epileptic encephalopathies.


Subject(s)
Brain Diseases/genetics , Epilepsy, Frontal Lobe/genetics , Potassium Channels, Sodium-Activated/genetics , Adolescent , Brain Diseases/metabolism , Child , Epilepsy, Frontal Lobe/diagnosis , Epilepsy, Generalized/genetics , Female , Humans , Male , Mutation, Missense/genetics , Neurodevelopmental Disorders/diagnosis , Neurodevelopmental Disorders/genetics , Phenotype , Potassium Channels, Sodium-Activated/metabolism , Exome Sequencing , Young Adult
13.
J Neurosci Res ; 98(10): 1889-1904, 2020 10.
Article in English | MEDLINE | ID: mdl-32643793

ABSTRACT

Iron is essential for normal cellular function, however, excessive accumulation of iron in neural tissue has been implicated in both cortical and retinal diseases. The exact role of iron in the pathogenesis of neurodegenerative disorders remains incompletely understood. However, iron-induced damage to the brain and retina is often attributed to the redox ability of iron to generate dangerous free radicals, which exacerbates local oxidative stress and neuronal damage. Iron chelators are compounds designed to scavenge labile iron, aiding to regulate iron bioavailability. Recently there has been growing interest in the application of chelating agents for treatment of diseases including neurodegenerative conditions, characterized by increased oxidative stress. This article reviews both clinical and preclinical evidence relating to the effectiveness of iron chelation therapy in conditions of iron dyshomeostasis linked to neurodegeneration in the brain and retina. The limitations as well as future opportunities iron chelation therapy are discussed.


Subject(s)
Brain Diseases/drug therapy , Brain Diseases/metabolism , Iron Chelating Agents/therapeutic use , Iron/metabolism , Retinal Diseases/drug therapy , Retinal Diseases/metabolism , Animals , Humans , Iron Chelating Agents/pharmacology , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Neuroprotection/drug effects , Neuroprotection/physiology
14.
Nutrients ; 12(6)2020 Jun 21.
Article in English | MEDLINE | ID: mdl-32575852

ABSTRACT

There is mounting evidence that diets supplemented with polyunsaturated fatty acids (PUFA) can impact brain biology and functions. This study investigated whether moderately high-fat diets differing in n-6/n-3 fatty acid ratio could impact fatty acid composition in regions of the brain linked to various psychopathologies. Adult male Sprague Dawley rats consumed isocaloric diets (35% kcal from fat) containing different ratios of linoleic acid (n-6) and alpha-linolenic acid (n-3) for 2 months. It was found that the profiles of PUFA in the prefrontal cortex, hippocampus, and hypothalamus reflected the fatty acid composition of the diet. In addition, region-specific changes in saturated fatty acids and monounsaturated fatty acids were detected in the hypothalamus, but not in the hippocampus or prefrontal cortex. This study in adult rats demonstrates that fatty acid remodeling in the brain by diet can occur within months and provides additional evidence for the suggestion that diet could impact mental health.


Subject(s)
Brain/metabolism , Diet, High-Fat , Dietary Fats/metabolism , Fatty Acids, Omega-3/metabolism , Fatty Acids, Omega-6/metabolism , Feeding Behavior , Nutritional Status , Animals , Brain Diseases/metabolism , Brain Mapping , Fatty Acids/metabolism , Fatty Acids, Monounsaturated/metabolism , Linoleic Acid/metabolism , Male , Rats, Sprague-Dawley , alpha-Linolenic Acid/metabolism
15.
Prog Neurobiol ; 191: 101806, 2020 08.
Article in English | MEDLINE | ID: mdl-32473843

ABSTRACT

Growing evidence highlights the relevance of microbiota-gut-brain axis in the maintenance of brain homeostasis as well as in the pathophysiology of major neurological and psychiatric disorders, including Parkinson's disease (PD), Alzheimer's disease (AD), multiple sclerosis (MS), autism spectrum disorder (ASD) and major depressive disorder (MDD). In particular, changes in gut microbiota can promote enteric and peripheral neurogenic/inflammatory responses, which, in turn, could contribute to neuroinflammation and neurodegeneration in the central nervous system (CNS). Of note, the nucleotide-binding oligomerization domain leucine rich repeat and pyrin domain-containing protein 3 (NLRP3) inflammasome acts as a key player in both coordinating the host physiology and shaping the peripheral and central immune/inflammatory responses in CNS diseases. In this context, there is pioneering evidence supporting the existence of a microbiota-gut-inflammasome-brain axis, in which enteric bacteria modulate, via NLRP3 signaling, inflammatory pathways that, in turn, contribute to influence brain homeostasis. The present review provides an overview of current knowledge on the role of microbiota-gut-inflammasome-brain axis in the major CNS diseases, including PD, AD, MS, ASD and MDD. In particular, though no direct and causal correlation among altered gut microbiota, NLRP3 activation and brain pathology has been demonstrated and in-depth studies are needed in this setting, our purpose was to pave the way to a novel and pioneering perspective on the pathophysiology of CNS disorders. Our intent was also to highlight and discuss whether alterations of microbiota-gut-inflammasome-brain axis support a holistic view of the pathophysiology of CNS diseases, even though each disorder displays a different clinical picture.


Subject(s)
Brain Diseases , Gastrointestinal Microbiome , Inflammasomes , Inflammation , Mental Disorders , NLR Family, Pyrin Domain-Containing 3 Protein , Animals , Brain Diseases/immunology , Brain Diseases/metabolism , Brain Diseases/microbiology , Gastrointestinal Microbiome/immunology , Humans , Inflammasomes/immunology , Inflammasomes/metabolism , Inflammation/immunology , Inflammation/metabolism , Inflammation/microbiology , Mental Disorders/immunology , Mental Disorders/metabolism , Mental Disorders/microbiology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
16.
J Ethnopharmacol ; 257: 112876, 2020 Jul 15.
Article in English | MEDLINE | ID: mdl-32305638

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Withania somnifera (Family: Solanaceae), commonly known as Ashwagandha or Indian ginseng is distributed widely in India, Nepal, China and Yemen. The roots of plant consist of active phytoconstituents mainly withanolides, alkaloids and sitoindosides and are conventionally used for the treatment of multiple brain disorders. AIM OF THE REVIEW: This review aims to critically assess and summarize the current state and implication of Ashwagandha in brain disorders. We have mainly focussed on the reported neuroactive phytoconstituents, available marketed products, pharmacological studies, mechanism of action and recent patents published related to neuroprotective effects of Ashwagandha in brain disorders. MATERIALS AND METHODS: All the information and data was collected on Ashwagandha using keywords "Ashwagandha" along with "Phytoconstituents", "Ayurvedic, Unani and Homeopathy marketed formulation", "Brain disorders", "Mechanism" and "Patents". Following sources were searched for data collection: electronic scientific databases such as Science Direct, Google Scholar, Elsevier, PubMed, Wiley On-line Library, Taylor and Francis, Springer; books such as AYUSH Pharmacopoeia; authentic textbooks and formularies. RESULTS: Identified neuroprotective phytoconstituents of Ashwagandha are sitoindosides VII-X, withaferin A, withanosides IV, withanols, withanolide A, withanolide B, anaferine, beta-sitosterol, withanolide D with key pharmacological effects in brain disorders mainly anxiety, Alzheimer's, Parkinson's, Schizophrenia, Huntington's disease, dyslexia, depression, autism, addiction, amyotrophic lateral sclerosis, attention deficit hyperactivity disorder and bipolar disorders. The literature survey does not highlight any toxic effects of Ashwagandha. Further, multiple available marketed products and patents recognized its beneficial role in various brain disorders; however, very few data is available on mechanistic pathway and clinical studies of Ashwagandha for various brain disorders is scarce and not promising. CONCLUSION: The review concludes the results of recent studies on Ashwagandha suggesting its extensive potential as neuroprotective in various brain disorders as supported by preclinical studies, clinical trials and published patents. However vague understanding of the mechanistic pathways involved in imparting the neuroprotective effect of Ashwagandha warrants further study to promote it as a promising drug candidate.


Subject(s)
Brain Diseases/drug therapy , Brain/drug effects , Neuroprotective Agents/therapeutic use , Phytotherapy , Plant Extracts/therapeutic use , Animals , Brain/metabolism , Brain/pathology , Brain/physiopathology , Brain Diseases/metabolism , Brain Diseases/pathology , Brain Diseases/physiopathology , Drug Development/legislation & jurisprudence , Drug Discovery/legislation & jurisprudence , Humans , Neuroprotective Agents/adverse effects , Neuroprotective Agents/isolation & purification , Patents as Topic , Plant Extracts/adverse effects , Plant Extracts/chemistry , Plant Extracts/isolation & purification , Plant Roots , Signal Transduction
17.
EBioMedicine ; 52: 102663, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32062359

ABSTRACT

BACKGROUND: Although thalamic magnetic resonance (MR) spectroscopy (MRS) accurately predicts adverse outcomes after neonatal encephalopathy, its utility in infants without MR visible deep brain nuclei injury is not known. We examined thalamic MRS metabolite perturbations in encephalopathic infants with white matter (WM) injury with or without cortical injury and its associations with adverse outcomes. METHODS: We performed a subgroup analysis of all infants recruited to the MARBLE study with isolated WM or mixed WM/cortical injury, but no visible injury to the basal ganglia/thalamus (BGT) or posterior limb of the internal capsule (PLIC). We used binary logistic regression to examine the association of MRS biomarkers with three outcomes (i) WM injury score (1 vs. 2/3); (ii) cortical injury scores (0/1 vs. 2/3); and (iii) adverse outcomes (defined as death, moderate/severe disability) at two years (yes/no). We also assessed the accuracy of MRS for predicting adverse outcome. FINDINGS: Of the 107 infants included in the analysis, five had adverse outcome. Reduced thalamic N-acetylaspartate concentration [NAA] (odds ratio 0.4 (95% CI 0.18-0.93)) and elevated thalamic Lactate/NAA peak area ratio (odds ratio 3.37 (95% CI 1.45-7.82)) were significantly associated with higher WM injury scores, but not with cortical injury. Thalamic [NAA] (≤5.6 mmol/kg/wet weight) had the best accuracy for predicting adverse outcomes (sensitivity 1.00 (95% CI 0.16-1.00); specificity 0.95 (95% CI 0.84-0.99)). INTERPRETATION: Thalamic NAA is reduced in encephalopathic infants without MR visible deep brain nuclei injury and may be a useful predictor of adverse outcomes. FUNDING: The National Institute for Health Research (NIHR).


Subject(s)
Brain Diseases/complications , Brain Diseases/metabolism , Brain Injuries/etiology , Brain Injuries/pathology , Energy Metabolism , Thalamus/metabolism , White Matter/pathology , Biomarkers , Brain Diseases/diagnosis , Brain Injuries/diagnostic imaging , Female , Humans , Infant , Infant, Newborn , Infant, Newborn, Diseases , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy , Male , Sensitivity and Specificity , White Matter/diagnostic imaging
18.
Neuropsychobiology ; 79(1): 89-99, 2020.
Article in English | MEDLINE | ID: mdl-30808841

ABSTRACT

In the past, accelerated tryptophan breakdown was considered to be a feature of clinical conditions, such as infection, inflammation, and malignant disease. More recently, however, the focus has changed to include the additional modulation of tryptophan metabolism by changes in nutrition and microbiota composition. The regulation of tryptophan concentration is critical for the maintenance of systemic homeostasis because it integrates essential pathways involved in nutrient sensing, metabolic stress response, and immunity. In addition to tryptophan being important as a precursor for the synthesis of the neurotransmitter serotonin, several catabolites along the kynurenine axis are neuroactive. This emphasizes the importance of the immunometabolic fate of this amino acid for processes relevant to neuropsychiatric symptoms. In humans, besides hepatic catabolism, there is usually a strong relationship between immune activation-associated tryptophan breakdown and increased levels of biomarkers, such as neopterin, which has particular relevance for both acute and chronic diseases. A shift towards neopterin synthesis during oxidative stress may indicate a corresponding decrease in tetrahydrobiopterin, a cofactor of several mono-oxygenases, providing a further link between tryptophan metabolism and serotonergic and catecholaminergic neurotransmission. The psychoneuroimmunological consequences of tryptophan metabolism and the susceptibility of this pathway to modulation by a variety of nutritional and lifestyle-related factors have important implications for the development of both diagnostic and treatment options.


Subject(s)
Brain Diseases , Diet , Gastrointestinal Microbiome , Life Style , Psychoneuroimmunology , Signal Transduction , Tryptophan/metabolism , Brain Diseases/immunology , Brain Diseases/metabolism , Brain Diseases/microbiology , Brain Diseases/therapy , Gastrointestinal Microbiome/physiology , Humans , Signal Transduction/physiology
19.
Inflammopharmacology ; 28(4): 939-948, 2020 Aug.
Article in English | MEDLINE | ID: mdl-31646410

ABSTRACT

Nicotine mediates some of the injurious effects caused by consuming tobacco products. The aim of this work is to investigate the protective effects of Mentha spicata extract (ME) supplementation on the testis and brain of nicotine-induced oxidative damage rats. ME extract showed interesting hydrogen peroxide-scavenging activity. HPLC-DAD analysis of ME revealed the presence of nine compounds among them gallic acid was the major one (165.44 µg/g ME). Thirty-two rats were randomly divided into four groups: control, a nicotine-treated group (1 mg/kg i.p.), a group receiving ME (100 mg/kg), and a group receiving both ME (100 mg/kg) and nicotine (1 mg/kg). After 2 months of treatment, the in vivo results showed that nicotine exhibited an increase in the body, brain, testis and accessory sex organ weights, sperm count and sperm motility. In addition, exposure to nicotine significantly (p < 0.01) increased acetylcholinesterase level (AChE) in brain, lipid peroxidation level in brain and testis as compared to control group. The antioxidant enzymes results showed that nicotine treatment induced a significant decrease (p < 0.01) in brain and testis antioxidant enzymes such as catalase, superoxide dismutase and glutathione peroxidase as compared to control group. Interestingly, pretreatment with ME significantly (p < 0.01) restored the majority of these biological parameters to normal levels, as well as a histological improvement. Obtained results suggest that ME contains promising substances that counteract the nicotine-intoxication and can be efficient in the prevention of brain and testis toxicity complications.


Subject(s)
Acetylcholinesterase/metabolism , Brain Diseases/drug therapy , Mentha spicata/chemistry , Nicotine/adverse effects , Oxidative Stress/drug effects , Plant Extracts/pharmacology , Testis/drug effects , Animals , Antioxidants/pharmacology , Brain/drug effects , Brain/metabolism , Brain Diseases/chemically induced , Brain Diseases/metabolism , Catalase/metabolism , Cholinesterase Inhibitors/adverse effects , Glutathione/metabolism , Glutathione Peroxidase/metabolism , Lipid Peroxidation/drug effects , Male , Rats , Rats, Wistar , Sperm Motility/drug effects , Spermatozoa/drug effects , Superoxide Dismutase/metabolism , Testis/metabolism
20.
Int J Mol Sci ; 20(24)2019 Dec 05.
Article in English | MEDLINE | ID: mdl-31817343

ABSTRACT

There is a huge demand for pro-/anti-angiogenic nanomedicines to treat conditions such as ischemic strokes, brain tumors, and neurodegenerative diseases such as Alzheimer's and Parkinson's. Nanomedicines are therapeutic particles in the size range of 10-1000 nm, where the drug is encapsulated into nano-capsules or adsorbed onto nano-scaffolds. They have good blood-brain barrier permeability, stability and shelf life, and able to rapidly target different sites in the brain. However, the relationship between the nanomedicines' physical and chemical properties and its ability to travel across the brain remains incompletely understood. The main challenge is the lack of a reliable drug testing model for brain angiogenesis. Recently, microfluidic platforms (known as "lab-on-a-chip" or LOCs) have been developed to mimic the brain micro-vasculature related events, such as vasculogenesis, angiogenesis, inflammation, etc. The LOCs are able to closely replicate the dynamic conditions of the human brain and could be reliable platforms for drug screening applications. There are still many technical difficulties in establishing uniform and reproducible conditions, mainly due to the extreme complexity of the human brain. In this paper, we review the prospective of LOCs in the development of nanomedicines for brain angiogenesis-related conditions.


Subject(s)
Angiogenesis Inducing Agents , Angiogenesis Inhibitors , Blood-Brain Barrier/metabolism , Brain Diseases , Lab-On-A-Chip Devices , Models, Biological , Nanomedicine , Angiogenesis Inducing Agents/chemistry , Angiogenesis Inducing Agents/pharmacokinetics , Angiogenesis Inducing Agents/therapeutic use , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/pharmacokinetics , Angiogenesis Inhibitors/therapeutic use , Blood-Brain Barrier/pathology , Brain Diseases/drug therapy , Brain Diseases/metabolism , Brain Diseases/pathology , Drug Evaluation, Preclinical , Humans , Nanomedicine/instrumentation , Nanomedicine/methods , Permeability
SELECTION OF CITATIONS
SEARCH DETAIL