Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 510
Filter
Add more filters

Publication year range
1.
Curr Opin Clin Nutr Metab Care ; 27(2): 155-162, 2024 03 01.
Article in English | MEDLINE | ID: mdl-38205831

ABSTRACT

PURPOSE OF REVIEW: Thiamine is a crucial component in cellular energy metabolism, serving as a cofactor for multiple enzymatic processes and also having a role in regulating neuronal and neuromuscular transmission. Also it exerts antioxidant proprieties. The objective of this review is to consolidate and assess the most recent research concerning the consequences of insufficient thiamine levels for critically ill patients and to examine thiamine-related interventions. RECENT FINDINGS: Recent studies have unveiled a noteworthy association between thiamine deficiency and unfavorable consequences, such as heightened morbidity and fatality rates. The aforementioned deficiency exhibits a significant presence in medical situations such as starvation and alcohol use disorder, but also in patients during critical illness. Thiamine deficiency can have significant metabolic implications resulting in compromised energy generation and organ dysfunction, warranting prompt recognition and management. SUMMARY: Thiamine deficiency may not be recognized in critical care. Timely identification and management are imperative to mitigate adverse outcomes and improve patient prognosis. Thiamine may offer benefits for specific patient groups at higher risk of deficiency. Future studies should focus to establish optimal dosing, timing, and monitoring strategies on understanding the pathophysiological changes associated with thiamine deficiency in ICU patients and clarify its role in improving clinical outcomes.


Subject(s)
Critical Illness , Thiamine Deficiency , Humans , Thiamine Deficiency/complications , Thiamine Deficiency/metabolism , Thiamine , Critical Care/methods , Energy Metabolism
2.
Mol Cell Neurosci ; 123: 103785, 2022 12.
Article in English | MEDLINE | ID: mdl-36241022

ABSTRACT

Insufficiencies of the micronutrient thiamine (Vitamin B1) have been associated with inducing Alzheimer's disease (AD)-like neuropathology. The hypometabolic state associated with chronic thiamine insufficiency (TI) has been demonstrated to be a contributor towards the development of amyloid plaque deposition and neurotoxicity. However, the molecular mechanism underlying TI induced AD pathology is still unresolved. Previously, we have established that TI stabilizes the metabolic stress transcriptional factor, Hypoxia Inducible Factor-1α (HIF1α). Utilizing neuronal hippocampal cells (HT22), TI-induced HIF1α activation triggered the amyloidogenic cascade through transcriptional expression and increased activity of ß-secretase (BACE1). Knockdown and pharmacological inhibition of HIF1α during TI significantly reduced BACE1 and C-terminal Fragment of 99 amino acids (C99) formation. TI also increased the expression of the HIF1α regulated pro-apoptotic protein, BCL2/adenovirus E1B 19 kDa protein-interacting protein (BNIP3). Correspondingly, cell toxicity during TI conditions was significantly reduced with HIF1α and BNIP3 knockdown. The role of BNIP3 in TI-mediated toxicity was further highlighted by localization of dimeric BNIP3 into the mitochondria and nuclear accumulation of Endonuclease G. Subsequently, TI decreased mitochondrial membrane potential and enhanced chromatin fragmentation. However, cell toxicity via the HIF1α/BNIP3 cascade required TI induced oxidative stress. HIF1α, BACE1 and BNIP3 expression was induced in 3xTg-AD mice after TI and administration with the HIF1α inhibitor YC1 significantly attenuated HIF1α and target genes levels in vivo. Overall, these findings demonstrate a critical stress response during TI involving the induction of HIF1α transcriptional activity that directly promotes neurotoxicity and AD-like pathology.


Subject(s)
Alzheimer Disease , Hypoxia-Inducible Factor 1, alpha Subunit , Thiamine Deficiency , Animals , Mice , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Thiamine/pharmacology , Thiamine Deficiency/metabolism
3.
Int J Mol Sci ; 24(5)2023 Feb 24.
Article in English | MEDLINE | ID: mdl-36901896

ABSTRACT

Mild thiamine deficiency aggravates Zn accumulation in cholinergic neurons. It leads to the augmentation of Zn toxicity by its interaction with the enzymes of energy metabolism. Within this study, we tested the effect of Zn on microglial cells cultivated in a thiamine-deficient medium, containing 0.003 mmol/L of thiamine vs. 0.009 mmol/L in a control medium. In such conditions, a subtoxic 0.10 mmol/L Zn concentration caused non-significant alterations in the survival and energy metabolism of N9 microglial cells. Both activities of the tricarboxylic acid cycle and the acetyl-CoA level were not decreased in these culture conditions. Amprolium augmented thiamine pyrophosphate deficits in N9 cells. This led to an increase in the intracellular accumulation of free Zn and partially aggravated its toxicity. There was differential sensitivity of neuronal and glial cells to thiamine-deficiency-Zn-evoked toxicity. The co-culture of neuronal SN56 with microglial N9 cells reduced the thiamine-deficiency-Zn-evoked inhibition of acetyl-CoA metabolism and restored the viability of the former. The differential sensitivity of SN56 and N9 cells to borderline thiamine deficiency combined with marginal Zn excess may result from the strong inhibition of pyruvate dehydrogenase in neuronal cells and no inhibition of this enzyme in the glial ones. Therefore, ThDP supplementation can make any brain cell more resistant to Zn excess.


Subject(s)
Microglia , Thiamine Deficiency , Humans , Microglia/metabolism , Acetyl Coenzyme A/metabolism , Thiamine Deficiency/metabolism , Cholinergic Neurons/metabolism , Thiamine Pyrophosphate/metabolism , Cholinergic Agents/metabolism , Zinc/metabolism
4.
J Neuroinflammation ; 19(1): 272, 2022 Nov 14.
Article in English | MEDLINE | ID: mdl-36376954

ABSTRACT

BACKGROUND: Thiamine (vitamin B1) is a cofactor for enzymes of central energy metabolism and its deficiency (TD) impairs oxidative phosphorylation, increases oxidative stress, and activates inflammatory processes that can lead to neurodegeneration. Wernicke-Korsakoff syndrome (WKS) is a consequence of chronic TD, which leads to extensive neuronal death, and is associated with neuropathological disorders, including cognitive deficits and amnesia. The hippocampus is one of the brain areas most affected by WKS. B1 replacement may not be enough to prevent the irreversible cognitive deficit associated with WKS. MATERIALS AND METHODS: An organotypic hippocampal slice culture (OHC) model was developed to investigate, using immunofluorescence and confocal microscopy and transcriptome analysis, the molecular mechanisms underlying the neurodegeneration associated with TD. The effect of anti-inflammatory pharmacological intervention with resveratrol (RSV) was also assessed in B1-deprived OHCs. RESULTS: In OHCs cultured without B1, neuronal density decayed after 5 days and, on the 7th day, the epigenetic markings H3K4me3 and H3K9me3 were altered in mature neurons likely favoring gene transcription. Between the 7th and the 14th day, a pulse of neurogenesis was observed followed by a further massive neuron loss. Transcriptome analysis at day nine disclosed 89 differentially expressed genes in response to B1 deprivation. Genes involved in tryptophan metabolism and lysine degradation KEGG pathways, and those with Gene Ontology (GO) annotations related to the organization of the extracellular matrix, cell adhesion, and positive regulation of synaptic transmission were upregulated. Several genes of the TNF and FoxO signaling pathways and with GO terms related to inflammation were inhibited in response to B1 deprivation. Nsd1, whose product methylates histone H3 lysine 36, was upregulated and the epigenetic marking H3K36me3, associated with negative regulation of neurogenesis, was increased in neurons. Treating B1-deprived OHCs with RSV promoted an earlier neurogenesis pulse. CONCLUSION: Neuroregeneration occurs in B1-deficient hippocampal tissue during a time window. This phenomenon depends on reducing neuroinflammation and, likely, on metabolic changes, allowing acetyl-CoA synthesis from amino acids to ensure energy supply via oxidative phosphorylation. Thus, neuroinflammation is implicated as a major regulator of hippocampal neurogenesis in TD opening a new search space for treating WKS.


Subject(s)
Neuroinflammatory Diseases , Thiamine Deficiency , Humans , Lysine/metabolism , Thiamine Deficiency/complications , Thiamine Deficiency/metabolism , Thiamine Deficiency/pathology , Neurogenesis/physiology , Hippocampus/metabolism , Thiamine/metabolism , Neurons/metabolism
5.
Nutr Cancer ; 74(6): 1942-1957, 2022.
Article in English | MEDLINE | ID: mdl-34854769

ABSTRACT

The malignant neoplastic cell is characterized by its diverse metabolic changes. It occurs in order to maintain the high rate of proliferation. The possibility of new pharmacological targets has inserted tumor metabolism as a target for recent research, emphasizing the enzymatic activity of thiamin. This review aims to elucidate the behavior of thiamin against tumor development. This is a systematic review in which studies indexed in Pubmed, Scopus, SciELO and BVS were searched using the descriptors (Thiamin OR Vitamin B1) AND (Cancer OR Malignant neoplasia) AND (Tumor metabolism). Title and abstract were read. Duplicates, literary reviews, books, conference abstracts, editorials, and papers published prior to 2010 were eliminated. 23 records were included in this review. Low doses of thiamin have been shown to be enough to stimulate tumor growth. Another population studies has shown evidence of tumor regression after correction of vitamin B1 deficiency. There is an open path for the development of new research to better assess the influence of thiamin on cancer cells. Once the connections between thiamin and the metabolism of cancer cells are fully established, new opportunities for therapeutic intervention and dietary modification will appear to reduce the progression of the disease in patients with cancer.


Subject(s)
Neoplasms , Thiamine Deficiency , Carcinogenesis , Cell Transformation, Neoplastic , Dietary Supplements , Humans , Neoplasms/drug therapy , Thiamine/pharmacology , Thiamine Deficiency/drug therapy , Thiamine Deficiency/metabolism
6.
PLoS Biol ; 16(4): e2002907, 2018 04.
Article in English | MEDLINE | ID: mdl-29659562

ABSTRACT

A constellation of metabolic disorders, including obesity, dysregulated lipids, and elevations in blood glucose levels, has been associated with cardiovascular disease and diabetes. Analysis of data from recently published genome-wide association studies (GWAS) demonstrated that reduced-function polymorphisms in the organic cation transporter, OCT1 (SLC22A1), are significantly associated with higher total cholesterol, low-density lipoprotein (LDL) cholesterol, and triglyceride (TG) levels and an increased risk for type 2 diabetes mellitus, yet the mechanism linking OCT1 to these metabolic traits remains puzzling. Here, we show that OCT1, widely characterized as a drug transporter, plays a key role in modulating hepatic glucose and lipid metabolism, potentially by mediating thiamine (vitamin B1) uptake and hence its levels in the liver. Deletion of Oct1 in mice resulted in reduced activity of thiamine-dependent enzymes, including pyruvate dehydrogenase (PDH), which disrupted the hepatic glucose-fatty acid cycle and shifted the source of energy production from glucose to fatty acids, leading to a reduction in glucose utilization, increased gluconeogenesis, and altered lipid metabolism. In turn, these effects resulted in increased total body adiposity and systemic levels of glucose and lipids. Importantly, wild-type mice on thiamine deficient diets (TDs) exhibited impaired glucose metabolism that phenocopied Oct1 deficient mice. Collectively, our study reveals a critical role of hepatic thiamine deficiency through OCT1 deficiency in promoting the metabolic inflexibility that leads to the pathogenesis of cardiometabolic disease.


Subject(s)
Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Type 2/genetics , Longevity/genetics , Obesity/genetics , Octamer Transcription Factor-1/genetics , Thiamine Deficiency/genetics , Thiamine/metabolism , Animals , Blood Glucose/metabolism , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Fatty Acids/metabolism , Gene Expression Regulation , Gluconeogenesis/genetics , Humans , Ketone Oxidoreductases/genetics , Ketone Oxidoreductases/metabolism , Lipid Metabolism/genetics , Liver/metabolism , Liver/pathology , Mice , Mice, Knockout , Obesity/metabolism , Obesity/pathology , Octamer Transcription Factor-1/deficiency , Signal Transduction , Thiamine Deficiency/metabolism , Thiamine Deficiency/pathology , Triglycerides/blood
7.
Alcohol Clin Exp Res ; 45(5): 1013-1027, 2021 05.
Article in English | MEDLINE | ID: mdl-33690917

ABSTRACT

BACKGROUND: Few studies have investigated differences in the vulnerabilities of males and females to alcohol use disorder and alcohol-related brain damage (ARBD). According to epidemiological and clinical findings, females appear to be more sensitive to the effects of alcohol and thiamine deficiency and have a worse prognosis in recovery from neurocognitive deficits compared with males. This study aimed to characterize the effects of chronic ethanol (EtOH) toxicity and thiamine deficiency across the sexes using rodent models. METHODS: Male and female Sprague Dawley rats were assigned to chronic forced EtOH treatment (CET), pyrithiamine-induced thiamine deficiency (PTD), combined CET-PTD, or pair-fed (PF) control treatment conditions. Following treatments, spatial working memory was assessed during a spontaneous alternation task while measuring acetylcholine (ACh) in the prefrontal cortex (PFC) and the hippocampus (HPC). The animals also underwent an operant-based attentional set-shifting task (ASST) for the analysis of behavioral flexibility. RESULTS: Female and male rats did not differ in terms of EtOH consumption; however, the CET and CET-PTD-treated female rats had lower BECs than male rats. Compared with the PF group, the CET, PTD, and CET-PTD groups exhibited spatial working memory impairments with corresponding reductions in ACh efflux in the PFC and HPC. The ASST revealed that CET-PTD-treated males and females displayed impairments marked by increased latency to make decisions. Thalamic shrinkage was prominent only in the CET-PTD and PTD treatment conditions, but no sex-specific effects were observed. CONCLUSIONS: Although the CET and CET-PTD-treated females had lower BECs than the males, they demonstrated similar cognitive impairments. These results provide evidence that female rats experience behavioral and neurochemical disruptions at lower levels of alcohol exposure than males and that chronic EtOH and thiamine deficiencies produce a unique behavioral profile.


Subject(s)
Acetylcholine/metabolism , Alcoholism/metabolism , Central Nervous System Depressants/pharmacology , Ethanol/pharmacology , Hippocampus/drug effects , Prefrontal Cortex/drug effects , Thiamine Deficiency/metabolism , Alcoholism/complications , Alcoholism/physiopathology , Animals , Antimetabolites/toxicity , Attention/drug effects , Behavior, Animal/drug effects , Case-Control Studies , Female , Hippocampus/metabolism , Male , Microdialysis , Prefrontal Cortex/metabolism , Pyrithiamine/toxicity , Rats , Sex Factors , Thiamine Deficiency/chemically induced , Thiamine Deficiency/complications , Thiamine Deficiency/physiopathology
8.
Nutr Neurosci ; 24(6): 432-442, 2021 Jun.
Article in English | MEDLINE | ID: mdl-31331253

ABSTRACT

Objectives: Zinc excitotoxicity and thiamine pyrophosphate deficiency (TD) are known pathogenic signals contributing to mechanism of different encephalopathies through inhibition of enzymes responsible for energy metabolism such as pyruvate dehydrogenase, aconitase or ketoglutarate dehydrogenase. The aim of this work was to investigate whether subclinical Zn excess and TD, frequent in aging brain, may combine yielding overt neuronal impairment.Results: Clonal SN56 cholinergic neuronal cells of septal origin were used as the model of brain cholinergic neurons, which are particularly susceptible to neurodegeneration in the course of Alzheimer's disease, hypoxia and other dementia-linked brain pathologies. Neither subtoxic concentration of Zn (0.10 mM) nor mild 20-25% TD deficits alone caused significant negative changes in cultured cholinergic neurons viability and their acetyl-CoA/acetylcholine metabolism. However, cells with mild TD accumulated Zn in excess, which impaired their energy metabolism causing a loss of neurons viability and their function as neurotransmitters. These negative effects of Zn were aggravated by amprolium which is an inhibitor of thiamine intracellular transport.Conclusion: Our data indicate that TD may amplify otherwise non-harmful border-line Zn excitotoxic signals yielding progress of neurodegeneration.


Subject(s)
Cholinergic Neurons/drug effects , Cholinergic Neurons/metabolism , Thiamine Deficiency/metabolism , Zinc/toxicity , Animals , Cell Line , Cell Survival/drug effects , Energy Metabolism/drug effects , Mice , Mitochondria/drug effects
9.
Metab Brain Dis ; 36(2): 239-246, 2021 02.
Article in English | MEDLINE | ID: mdl-33245475

ABSTRACT

Thiamine deficiency (TD) results in focal lesions in several regions of the rat brain including the thalamus and inferior colliculus. Since alterations in blood-brain barrier (BBB) integrity may play a role in this damage, we have examined the influence of TD on the unidirectional blood-to-brain transfer constant (Ki) of the low molecular weight species α-aminoisobutyric acid (AIB) in vulnerable and non-vulnerable brain regions at different stages during progression of the disorder, and following its reversal with thiamine. Analysis of the regional distribution of Ki values showed early (day 10) increased transfer of [14C]-AIB across the BBB in the vulnerable medial thalamus as well as the non-vulnerable caudate and hippocampus. At the acute symptomatic stage (day 14), more widespread BBB permeability changes were detected in most areas including the lateral thalamus, inferior colliculus, and non-vulnerable cerebellum and pons. Twenty-four hours following thiamine replenishment, a heterogeneous pattern of increased BBB permeability was observed in which many structures maintained increased uptake of [14C]-AIB. No increase in the [3H]-dextran space, a marker of intravascular volume, was detected in brain regions during the progress of TD, suggesting that BBB permeability to this large tracer was unaffected. These results indicate that BBB opening i) occurs early during TD, ii) is not restricted to vulnerable areas of the brain, iii) is progressive, iv) persists for at least 24 h following treatment with thiamine, and v) is likely selective in nature, depending on the molecular species being transported.


Subject(s)
Aminoisobutyric Acids/pharmacology , Blood-Brain Barrier/drug effects , Brain/drug effects , Thiamine Deficiency/metabolism , Animals , Blood-Brain Barrier/metabolism , Brain/metabolism , Male , Permeability/drug effects , Rats , Rats, Sprague-Dawley
10.
Int J Mol Sci ; 22(24)2021 Dec 11.
Article in English | MEDLINE | ID: mdl-34948135

ABSTRACT

Brain pathologies evoked by thiamine deficiency can be aggravated by mild zinc excess. Cholinergic neurons are the most susceptible to such cytotoxic signals. Sub-toxic zinc excess aggravates the injury of neuronal SN56 cholinergic cells under mild thiamine deficiency. The excessive cell loss is caused by Zn interference with acetyl-CoA metabolism. The aim of this work was to investigate whether and how astroglial C6 cells alleviated the neurotoxicity of Zn to cultured SN56 cells in thiamine-deficient media. Low Zn concentrations did not affect astroglial C6 and primary glial cell viability in thiamine-deficient conditions. Additionally, parameters of energy metabolism were not significantly changed. Amprolium (a competitive inhibitor of thiamine uptake) augmented thiamine pyrophosphate deficits in cells, while co-treatment with Zn enhanced the toxic effect on acetyl-CoA metabolism. SN56 cholinergic neuronal cells were more susceptible to these combined insults than C6 and primary glial cells, which affected pyruvate dehydrogenase activity and the acetyl-CoA level. A co-culture of SN56 neurons with astroglial cells in thiamine-deficient medium eliminated Zn-evoked neuronal loss. These data indicate that astroglial cells protect neurons against Zn and thiamine deficiency neurotoxicity by preserving the acetyl-CoA level.


Subject(s)
Cholinergic Neurons/metabolism , Neuroglia/metabolism , Thiamine Deficiency/prevention & control , Zinc/toxicity , Animals , Cell Line, Tumor , Culture Media , Mice , Thiamine/metabolism , Thiamine/pharmacology , Thiamine Deficiency/metabolism
11.
Appl Environ Microbiol ; 86(3)2020 01 21.
Article in English | MEDLINE | ID: mdl-31704686

ABSTRACT

Thiamine is a vitamin that functions as a cofactor for key enzymes in carbon and energy metabolism in all living cells. While most plants, fungi, and bacteria can synthesize thiamine de novo, the oleaginous yeast Yarrowia lipolytica cannot. In this study, we used proteomics together with physiological characterization to elucidate key metabolic processes influenced and regulated by thiamine availability and to identify the genetic basis of thiamine auxotrophy in Y. lipolytica Specifically, we found that thiamine depletion results in decreased protein abundance for the lipid biosynthesis pathway and energy metabolism (i.e., ATP synthase), leading to the negligible growth and poor sugar assimilation observed in our study. Using comparative genomics, we identified the missing 4-amino-5-hydroxymethyl-2-methylpyrimidine phosphate synthase (THI13) gene for the de novo thiamine biosynthesis in Y. lipolytica and discovered an exceptional promoter, P3, that exhibits strong activation and tight repression by low and high thiamine concentrations, respectively. Capitalizing on the strength of our thiamine-regulated promoter (P3) to express the missing gene from Saccharomyces cerevisiae (scTHI13), we engineered a thiamine-prototrophic Y. lipolytica strain. By comparing this engineered strain to the wild-type strain, we revealed the tight relationship between thiamine availability and lipid biosynthesis and demonstrated enhanced lipid production with thiamine supplementation in the engineered thiamine-prototrophic Y. lipolytica strain.IMPORTANCE Thiamine plays a crucial role as an essential cofactor for enzymes involved in carbon and energy metabolism in all living cells. Thiamine deficiency has detrimental consequences for cellular health. Yarrowia lipolytica, a nonconventional oleaginous yeast with broad biotechnological applications, is a native thiamine auxotroph whose affected cellular metabolism is not well understood. Therefore, Y. lipolytica is an ideal eukaryotic host for the study of thiamine metabolism, especially because mammalian cells are also thiamine auxotrophic and thiamine deficiency is implicated in several human diseases. This study elucidates the fundamental effects of thiamine deficiency on cellular metabolism in Y. lipolytica and identifies genes and novel thiamine-regulated elements that eliminate thiamine auxotrophy in Y. lipolytica Furthermore, the discovery of thiamine-regulated elements enables the development of thiamine biosensors with useful applications in synthetic biology and metabolic engineering.


Subject(s)
Fungal Proteins/metabolism , Proteome , Thiamine Deficiency/metabolism , Thiamine/metabolism , Yarrowia/metabolism
12.
Muscle Nerve ; 62(1): 13-29, 2020 07.
Article in English | MEDLINE | ID: mdl-31837157

ABSTRACT

Neuropathies associated with nutritional deficiencies are routinely encountered by the practicing neurologist. Although these neuropathies assume different patterns, most are length-dependent, sensory axonopathies. Cobalamin deficiency neuropathy is the exception, often presenting with a non-length-dependent sensory neuropathy. Patients with cobalamin and copper deficiency neuropathy characteristically have concomitant myelopathy, whereas vitamin E deficiency is uniquely associated with a spinocerebellar syndrome. In contrast to those nutrients for which deficiencies produce neuropathies, pyridoxine toxicity results in a non-length-dependent sensory neuronopathy. Deficiencies occur in the context of malnutrition, malabsorption, increased nutrient loss (such as with dialysis), autoimmune conditions such as pernicious anemia, and with certain drugs that inhibit nutrient absorption. When promptly identified, therapeutic nutrient supplementation may result in stabilization or improvement of these neuropathies.


Subject(s)
Avitaminosis/diagnosis , Avitaminosis/metabolism , Dietary Supplements , Nutritional Status/physiology , Peripheral Nervous System Diseases/diagnosis , Peripheral Nervous System Diseases/metabolism , Anemia, Pernicious/diagnosis , Anemia, Pernicious/drug therapy , Anemia, Pernicious/metabolism , Avitaminosis/drug therapy , Humans , Nutritional Status/drug effects , Peripheral Nervous System Diseases/drug therapy , Thiamine Deficiency/diagnosis , Thiamine Deficiency/drug therapy , Thiamine Deficiency/metabolism , Vitamin B 12 Deficiency/diagnosis , Vitamin B 12 Deficiency/drug therapy , Vitamin B 12 Deficiency/metabolism , Vitamins/administration & dosage
13.
Alzheimers Dement ; 16(11): 1591-1595, 2020 11.
Article in English | MEDLINE | ID: mdl-32808749

ABSTRACT

Alcohol-related dementia (ARD) is a common and severe co-morbidity in alcohol use disorder (AUD). We propose brain iron overload (BIO) to be an important and previously neglected pathogenic process, accelerating cognitive decline in AUD. Furthermore, we suggest thiamine, which is frequently depleted in AUD, to be a key modulator in this process: Thiamine deficiency impairs the integrity of the blood-brain barrier, thereby enabling iron to pass through and accumulate in the brain. This hypothesis is based on findings from animal, translational, and neuroimaging studies, discussed in this article. To validate this hypothesis, translational studies focusing on brain iron homeostasis in AUD, as well as prospective clinical studies investigating prevalence and clinical impact of BIO in AUD, should be conducted. If proven right, this would change the understanding of ARD and may lead to novel therapeutic interventions in prevention and treatment of ARD.


Subject(s)
Alcoholism/complications , Dementia/etiology , Iron Overload/etiology , Thiamine Deficiency/etiology , Thiamine/metabolism , Alcoholism/metabolism , Alcoholism/pathology , Animals , Brain/metabolism , Brain/pathology , Dementia/metabolism , Dementia/pathology , Humans , Iron Overload/metabolism , Iron Overload/pathology , Thiamine Deficiency/metabolism , Thiamine Deficiency/pathology
14.
Alcohol Clin Exp Res ; 43(3): 425-438, 2019 03.
Article in English | MEDLINE | ID: mdl-30589435

ABSTRACT

BACKGROUND: Alcohol-related brain damage (ARBD) is associated with neurotoxic effects of heavy alcohol use and nutritional deficiency, in particular thiamine deficiency (TD), both of which induce inflammatory responses in brain. Although neuroinflammation is a critical factor in the induction of ARBD, few studies have addressed the specific contribution(s) of ethanol (EtOH) versus TD. METHODS: Adult rats were randomly divided into 6 conditions: chronic EtOH treatment (CET) where rats consumed a 20% v/v solution of EtOH for 6 months; CET with injections of thiamine (CET + T); severe pyrithiamine-induced TD (PTD); moderate PTD; moderate PTD during CET; and pair-fed controls. After the treatments, the rats were split into 3 recovery phase time points: the last day of treatment (time point 1), acute recovery (time point 2: 24 hours posttreatment), and delayed recovery (time point 3: 3 weeks posttreatment). At these time points, vulnerable brain regions (thalamus, hippocampus, frontal cortex) were collected and changes in neuroimmune markers were assessed using a combination of reverse transcription polymerase chain reaction and protein analysis. RESULTS: CET led to minor fluctuations in neuroimmune genes, regardless of the structure being examined. In contrast, PTD treatment led to a profound increase in neuroimmune genes and proteins within the thalamus. Cytokine changes in the thalamus ranged in magnitude from moderate (3-fold and 4-fold increase in interleukin-1ß [IL-1ß] and IκBα) to severe (8-fold and 26-fold increase in tumor necrosis factor-α and IL-6, respectively). Though a similar pattern was observed in the hippocampus and frontal cortex, overall fold increases were moderate relative to the thalamus. Importantly, neuroimmune gene induction varied significantly as a function of severity of TD, and most genes displayed a gradual recovery across time. CONCLUSIONS: These data suggest an overt brain inflammatory response by TD and a subtle change by CET alone. Also, the prominent role of TD in the immune-related signaling pathways leads to unique regional and temporal profiles of induction of neuroimmune genes.


Subject(s)
Brain/drug effects , Brain/pathology , Ethanol/adverse effects , Inflammation Mediators/metabolism , Thiamine Deficiency/metabolism , Thiamine/pharmacology , Animals , Biomarkers/metabolism , Frontal Lobe/metabolism , Hippocampus/metabolism , Male , Pyrithiamine , Rats , Thalamus/metabolism , Thiamine Deficiency/chemically induced , Time Factors , Transcriptional Activation/drug effects
15.
Int J Med Sci ; 16(1): 1-7, 2019.
Article in English | MEDLINE | ID: mdl-30662322

ABSTRACT

The objective of this study was to determine the relationship between glucose dosage in parenteral nutrition and reductions in levels of body thiamine in rats. Vitamin-free infusions with differing amounts of glucose were administered to normal or thiamine-deficient rats for 5 days, after which urinary thiamine excretion and the amounts of thiamine in the blood, liver, brain, and skeletal muscles were measured. The total energy dosage was set at three levels (98, 140, and 196 kcal/kg), and the dose of amino acids was constant among all groups. Urinary thiamine excretions on Day 5 decreased with increasing glucose dosage in the infusions. In normal rats, the amount of thiamine in the blood and all organs decreased compared with the diet group; however, no significant differences were found among the infusion groups. In thiamine-deficient rats, on the other hand, the amount of thiamine in the liver and skeletal muscles did not differ significantly among infusion groups; however, the amount of thiamine in the brain and blood decreased with increasing glucose dosage. An organ-specific correlation was found between glucose dosage in infusions and reductions in levels of thiamine. To prevent thiamine deficiencies from affecting the central nervous system, greater caution must be exercised during high-caloric parenteral nutrition. However, a constant supply of thiamine seemed to be essential, irrespective of the amount of energy supplied via parenteral nutrition, to maintain a sufficient level of thiamine in the body.


Subject(s)
Glucose/administration & dosage , Sweetening Agents/metabolism , Thiamine Deficiency , Thiamine/blood , Thiamine/urine , Vitamin B Complex/blood , Vitamin B Complex/urine , Amino Acids/administration & dosage , Animals , Brain/metabolism , Electrolytes/administration & dosage , Glucose/therapeutic use , Japan , Liver/metabolism , Male , Muscle, Skeletal/metabolism , Organ Specificity , Parenteral Nutrition , Rats , Rats, Sprague-Dawley , Sweetening Agents/administration & dosage , Sweetening Agents/therapeutic use , Thiamine/metabolism , Thiamine Deficiency/drug therapy , Thiamine Deficiency/metabolism
16.
Cell Physiol Biochem ; 47(5): 1989-1997, 2018.
Article in English | MEDLINE | ID: mdl-29969779

ABSTRACT

BACKGROUND/AIMS: Thiamine-responsive megaloblastic anemia syndrome is a rare autosomal recessive disorder resulting from mutations in SLC19A2, and is mainly characterized by megaloblastic anemia, diabetes, and progressive sensorineural hearing loss. METHODS: We study a Chinese Zhuang ethnicity family with thiamine-responsive megaloblastic anemia. The proband of the study presented with anemia and diabetes, similar to his late brother, as well as visual impairment. All clinical manifestations were corrected with thiamine (30 mg/d) supplementation for 1-3 months, except for visual impairment, which was irreversible. The presence of mutations in all exons and the flanking sequences of the SLC19A2 gene were analyzed in this family based on the proband's and his brother's clinical data. Computer analysis and prediction of the protein conformation of mutant THTR-1. The relative concentration of thiamine pyrophosphate in the proband's whole blood before and after initiation of thiamine supplement was measured by high performance liquid chromatography (HPLC). RESULTS: Gene sequencing showed a homozygous mutation in exon 6 of the SLC19A2 gene (c.1409insT) in the proband. His parents and sister were diagnosed as heterozygous carriers of the c.1409insT mutation. Computer simulation showed that the mutations caused a change in protein conformation. HPLC results suggested that the relative concentration of thiamine pyrophosphate in the proband's whole blood after thiamine supplement was significantly different (P=0.016) from that at baseline. CONCLUSIONS: This novel homozygous mutation (c.1409insT) caused the onset of thiamine-responsive megaloblastic anemia in the proband.


Subject(s)
Anemia, Megaloblastic/genetics , Diabetes Mellitus/genetics , Exons , Hearing Loss, Sensorineural/genetics , Membrane Transport Proteins/genetics , Mutation , Thiamine Deficiency/congenital , Anemia, Megaloblastic/ethnology , Anemia, Megaloblastic/metabolism , Anemia, Megaloblastic/pathology , Asian People , China/ethnology , Diabetes Mellitus/ethnology , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Female , Hearing Loss, Sensorineural/ethnology , Hearing Loss, Sensorineural/metabolism , Hearing Loss, Sensorineural/pathology , Humans , Infant , Male , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/metabolism , Thiamine Deficiency/ethnology , Thiamine Deficiency/genetics , Thiamine Deficiency/metabolism , Thiamine Deficiency/pathology
17.
Nutr Metab Cardiovasc Dis ; 28(10): 965-972, 2018 10.
Article in English | MEDLINE | ID: mdl-30143411

ABSTRACT

BACKGROUND AND AIM: Thiamine, also known as vitamin B1, functions as a cofactor in the metabolism of carbohydrates and amino acids. Thiamine deficiency has been suggested to be associated with many cardiovascular diseases (CVDs) and risk factors including type 1 and type 2 diabetes (T1D and T2D, respectively), obesity, chronic vascular inflammation, dyslipidemia, heart failure (HF), myocardial infarction (MI) and conduction defects, and depression. The aim of this review was to explore the evidence of thiamine deficiency among subjects with CVDs or risk factors, illustrate the theories explaining the thiamine-CVDs associations, and describe the effect of thiamine supplementation. METHODS: Human and animal studies were collected from various scientific databases following the PRISMA guidelines without limitation regarding the publication year. Studies investigating the prevalence of thiamine deficiency among patients with CVDs and the effect of thiamine supplementation on their conditions were summarized. RESULTS AND CONCLUSIONS: Thiamine deficiency could have a role in the development of CVDs. Future studies should focus on the impact of thiamine supplementation on reversing CVDs and risk factors associated with its deficiency.


Subject(s)
Cardiovascular Diseases/epidemiology , Energy Metabolism , Thiamine Deficiency/epidemiology , Thiamine/metabolism , Adult , Aged , Aged, 80 and over , Animals , Cardiovascular Diseases/diagnosis , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/prevention & control , Dietary Supplements , Energy Metabolism/drug effects , Female , Humans , Male , Middle Aged , Risk Factors , Thiamine/administration & dosage , Thiamine Deficiency/diagnosis , Thiamine Deficiency/drug therapy , Thiamine Deficiency/metabolism , Treatment Outcome
18.
Biol Res ; 51(1): 35, 2018 Sep 19.
Article in English | MEDLINE | ID: mdl-30231926

ABSTRACT

BACKGROUND: The previous studies have demonstrated the reduction of thiamine diphosphate is specific to Alzheimer's disease (AD) and causal factor of brain glucose hypometabolism, which is considered as a neurodegenerative index of AD and closely correlates with the degree of cognitive impairment. The reduction of thiamine diphosphate may contribute to the dysfunction of synapses and neural circuits, finally leading to cognitive decline. RESULTS: To demonstrate this hypothesis, we established abnormalities in the glucose metabolism utilizing thiamine deficiency in vitro and in vivo, and we found dramatically reduced dendrite spine density. We further detected lowered excitatory neurotransmission and impaired hippocampal long-term potentiation, which are induced by TPK RNAi in vitro. Importantly, via treatment with benfotiamine, Aß induced spines density decrease was considerably ameliorated. CONCLUSIONS: These results revealed that thiamine deficiency contributed to synaptic dysfunction which strongly related to AD pathogenesis. Our results provide new insights into pathogenesis of synaptic and neuronal dysfunction in AD.


Subject(s)
Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Neurons/physiology , Synapses/physiology , Thiamine Deficiency/complications , Thiamine Deficiency/metabolism , Thiamine Pyrophosphate/deficiency , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Animals , Blotting, Western , Dendritic Spines/metabolism , Diphosphotransferases/metabolism , Glucose/metabolism , Hippocampus/metabolism , Hippocampus/physiopathology , Male , Mice, Inbred C57BL , Random Allocation , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Synaptic Transmission/physiology , Thiamine Deficiency/physiopathology , Thiamine Pyrophosphate/metabolism
19.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 35(1): 121-124, 2018 Feb 10.
Article in Zh | MEDLINE | ID: mdl-29419877

ABSTRACT

Thiamine, also known as vitamin B1, is an important vitamin for the body. The activated form of thiamine pyrophosphate is involved in cell metabolism as an important co-enzyme. Defects of thiamine transport and activation may cause lack of thiamine and affection of cell metabolism, leading a variety of diseases. This review has summarized defects of thiamine transport and activation and related diseases.


Subject(s)
Membrane Transport Proteins/genetics , Mutation , Thiamine Pyrophosphate/metabolism , Thiamine/metabolism , Anemia, Megaloblastic/genetics , Anemia, Megaloblastic/metabolism , Biological Transport/genetics , Diabetes Mellitus/genetics , Diabetes Mellitus/metabolism , Hearing Loss, Sensorineural/genetics , Hearing Loss, Sensorineural/metabolism , Humans , Membrane Transport Proteins/metabolism , Thiamine Deficiency/congenital , Thiamine Deficiency/genetics , Thiamine Deficiency/metabolism
20.
Am J Physiol Gastrointest Liver Physiol ; 313(5): G448-G455, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-28729247

ABSTRACT

The essentiality of thiamin stems from its roles as a cofactor [mainly in the form of thiamin pyrophosphate (TPP)] in critical metabolic reactions including oxidative energy metabolism and reduction of cellular oxidative stress. Like other mammalian cells, pancreatic acinar cells (PAC) obtain thiamin from their surroundings and convert it to TPP; mitochondria then take up TPP by a carrier-mediated process that involves the mitochondrial TPP (MTPP) transporter (MTPPT; product of SLC25A19 gene). Previous studies have characterized different physiological/biological aspects of the MTPP uptake process, but little is known about its possible adaptive regulation. We addressed this issue using pancreatic acinar 266-6 cells (PAC 266-6) maintained under thiamin-deficient (DEF) and oversupplemented (OS) conditions, as well as thiamin-DEF and -OS transgenic mice carrying the SLC25A19 promoter. We found that maintaining PAC 266-6 under the thiamin-DEF condition leads to a significant induction in mitochondrial [3H]TPP uptake, as well as in the level of expression of the MTPPT protein and mRNA compared with thiamin-OS cells. Similar findings were observed in mitochondria from thiamin-DEF mice compared with thiamin-OS. Subsequently, we demonstrated that adaptive regulation of MTTP protein was partly mediated via transcriptional mechanism(s) via studies with PAC 266-6 transfected with the SLC25A19 promoter and transgenic mice carrying the SLC25A19 promoter. This transcriptional regulation appeared to be, at least in part, mediated via epigenetic mechanism(s) involving histone modifications. These studies report, for the first time, that the PAC mitochondrial TPP uptake process is adaptively regulated by the prevailing thiamin level and that this regulation is transcriptionally mediated and involves epigenetic mechanism(s).NEW & NOTEWORTHY Our findings show, for the first time, that the mitochondrial thiamin pyrophosphate (MTPP) uptake process is adaptively regulated by the prevailing thiamin level in pancreatic acinar cells and this regulation is mediated, at least in part, by transcriptional and epigenetic mechanism(s) affecting the SLC25A19 promoter.


Subject(s)
Acinar Cells/metabolism , Anion Transport Proteins , Membrane Transport Proteins , Mitochondria/metabolism , Mitochondrial Proteins , Pancreas, Exocrine , Thiamine Pyrophosphate/metabolism , Thiamine , Adaptation, Biological/physiology , Animals , Anion Transport Proteins/genetics , Anion Transport Proteins/metabolism , Biological Transport, Active/physiology , Energy Metabolism/physiology , Humans , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mice , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Oxidative Stress/physiology , Pancreas, Exocrine/metabolism , Pancreas, Exocrine/pathology , Thiamine/analysis , Thiamine/metabolism , Thiamine Deficiency/etiology , Thiamine Deficiency/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL