Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Article in English | MEDLINE | ID: mdl-38713615

ABSTRACT

Humans/mammals obtain vitamin B1 from dietary and gut microbiota sources. Considerable amount of the microbiota generated vitamin exists in the form of thiamine pyrophosphate (TPP), and colonocytes are capable of absorbing TPP via a specific carrier-mediated process that involves the colonic TPP transporter (cTPPT; encoded by SLC44A4). Little is known about the relative contribution of SLC44A4 toward total colonic carrier-mediated TPP uptake, and its role in colon physiology. To address these issues, we generated an Slc44a4 knockout (KO) mouse model (by Cre-Lox recombination) and found a near complete inhibition in colonic carrier-mediated 3H-TPP uptake in the Slc44a4 KO compared to Wild-type-littermates (WT). We also observed a significant reduction in KO mice body weight and a shortening of their colon compared to WT. Using RNAseq and Ingenuity Pathway Analysis (IPA) approaches, we found that knocking out the colonic Slc44a4 to lead to changes in level of expression of many genes, including up-regulation in those associated with intestinal inflammation/colitis. Finally, we found that the Slc44a4 KO mice to be more susceptible to the effect of the colitogenic dextran sodium sulfate (DSS) compared to WT animals, a finding that lends support to the recent prediction by multiple genome-wide association studies (GWAS) that the SLC44A4 is a possible colitis susceptibility gene. In summary, results of these investigations show that the Slc44a4 is the predominant/only transporter involved in colonic uptake of TPP, that the transporter is important for colon physiology, and that its deletion increases susceptibility to inflammation.

2.
Am J Physiol Gastrointest Liver Physiol ; 325(4): G347-G355, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37529835

ABSTRACT

Thiamin (vitamin B1) plays a vital role in cellular energy metabolism/ATP production. Pancreatic acinar cells (PACs) obtain thiamin from circulation and convert it to thiamin pyrophosphate (TPP) in the cytoplasm. TPP is then taken up by the mitochondria via a carrier-mediated process that involves the mitochondrial TPP transporter (MTPPT; encoded by the gene SLC25A19). We have previously characterized different aspects of the mitochondrial carrier-mediated TPP uptake process, but nothing is known about its possible regulation at the posttranscriptional level. We address this issue in the current investigations focusing on the role of miRNAs in this regulation. First, we subjected the human (and rat) 3'-untranslated region (3'-UTR) of the SLC25A19 to three in-silico programs, and all have identified putative binding sites for miR-122-5p. Transfecting pmirGLO-hSLC25A19 3'-UTR into rat PAC AR42J resulted in a significant reduction in luciferase activity compared with cells transfected with pmirGLO-empty vector. Mutating as well as truncating the putative miR-122-5p binding sites in the hSLC25A19 3'-UTR led to abrogation of inhibition in luciferase activity in PAC AR42J. Furthermore, transfecting/transducing PAC AR42J and human primary PACs with mimic of miR-122-5p led to a significant inhibition in the level of expression of the MTPPT mRNA and protein as well as in mitochondrial carrier-mediated TPP uptake. Conversely, transfecting PAC AR42J with an inhibitor of miR-122-5p increased MTPPT expression and function. These findings show, for the first time, that expression and function of the MTPPT in PACs are subject to posttranscriptional regulation by miR-122-5p.NEW & NOTEWORTHY This study shows that the expression and function of mitochondrial TPP transporter (MTPPT) are subject to posttranscriptional regulation by miRNA-122-5p in pancreatic acinar cells.


Subject(s)
Acinar Cells , MicroRNAs , Humans , Rats , Animals , Acinar Cells/metabolism , Diphosphates/metabolism , Thiamine/metabolism , Thiamine Pyrophosphate/metabolism , Mitochondria/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Luciferases/metabolism , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Membrane Transport Proteins/metabolism
3.
Neurobiol Dis ; 171: 105799, 2022 09.
Article in English | MEDLINE | ID: mdl-35750148

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disease characterized by Amyloid-ß peptide (Aß) containing plaques and cognitive deficits. The pathophysiology of AD also involves neuroinflammation. Vitamin B1 (thiamin) is indispensable for normal cellular energy metabolism. Thiamin homeostasis is altered in AD, and its deficiency is known to aggravate AD pathology. Little, however, is known about possible alterations in level of expression of thiamin transporters-1 and -2 (THTR-1 and -2) in the brain of AD, and whether pro-inflammatory cytokines affect thiamin uptake by brain cells. We addressed these issues using brain tissue samples [prefrontal cortex (PFC) and hippocampus (HIP)] from AD patients and from 5XFAD mouse model of AD, together with cultured human neuroblastoma SH-SY5Y cells as model. Our results revealed a significantly lower expression of both THTR-1 and THTR-2 in the PFC and HIP of AD patients and 5XFAD mouse model of AD compared to appropriate normal controls. Further, we found that exposure of the SH-SY5Y cells to pro-inflammatory cytokines (IL-1ß, IL-6, and TNF-α) led to a significant inhibition in thiamin uptake. Focusing on IL-1ß, we found the inhibition in thiamin uptake to be time-dependent and reversible; it was also associated with a substantial reduction in expression of THTR-1 (but not THTR-2) protein and mRNA as well as a decrease in promoter activity of the SLC19A2 gene (which encodes THTR-1). Finally, using transcriptomic analysis, we found that thiamin availability in SH-SY5Y cells caused changes in the expression of genes relevant to AD pathways. These studies demonstrate, for the first time, that thiamin transport physiology/molecular biology parameters are negatively impacted in AD brain and that pro-inflammatory cytokines inhibit thiamin uptake by neuroblastoma cells. The results also support a possible role for thiamin in the pathophysiology of AD.


Subject(s)
Alzheimer Disease , Neuroblastoma , Neurodegenerative Diseases , Acinar Cells/metabolism , Acinar Cells/pathology , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Cytokines/metabolism , Humans , Membrane Transport Proteins , Mice , Mice, Transgenic , Neuroblastoma/pathology , Neurodegenerative Diseases/metabolism , Neuroinflammatory Diseases , Thiamine/metabolism
4.
J Biol Chem ; 298(2): 101562, 2022 02.
Article in English | MEDLINE | ID: mdl-34998824

ABSTRACT

Hypoxia exerts profound effects on cell physiology, but its effect on colonic uptake of the microbiota-generated forms of vitamin B1 (i.e., thiamin pyrophosphate [TPP] and free thiamine) has not been described. Here, we used human colonic epithelial NCM460 cells and human differentiated colonoid monolayers as in vitro and ex vivo models, respectively, and were subjected to either chamber (1% O2, 5% CO2, and 94% N2) or chemically (desferrioxamine; 250 µM)-induced hypoxia followed by determination of different physiological-molecular parameters. We showed that hypoxia causes significant inhibition in TPP and free thiamin uptake by colonic NCM460 cells and colonoid monolayers; it also caused a significant reduction in the expression of TPP (SLC44A4) and free thiamin (SLC19A2 and SLC19A3) transporters and in activity of their gene promoters. Furthermore, hypoxia caused a significant induction in levels of hypoxia-inducible transcription factor (HIF)-1α but not HIF-2α. Knocking down HIF-1α using gene-specific siRNAs in NCM460 cells maintained under hypoxic conditions, on the other hand, led to a significant reversal in the inhibitory effect of hypoxia on TPP and free thiamin uptake as well as on the expression of their transporters. Finally, a marked reduction in level of expression of the nuclear factors cAMP responsive element-binding protein 1 and gut-enriched Krüppel-like factor 4 (required for activity of SLC44A4 and SLC19A2 promoters, respectively) was observed under hypoxic conditions. In summary, hypoxia causes severe inhibition in colonic TPP and free thiamin uptake that is mediated at least in part via HIF-1α-mediated transcriptional mechanisms affecting their respective transporters.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit , Microbiota , Thiamine , Biological Transport , Cell Hypoxia/physiology , Humans , Hypoxia/metabolism , Hypoxia/microbiology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Membrane Transport Proteins/metabolism , Thiamine/metabolism , Thiamine Pyrophosphate/metabolism
5.
Am J Physiol Gastrointest Liver Physiol ; 321(2): G123-G133, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34077272

ABSTRACT

Vitamin B7 (biotin) is essential for normal health and its deficiency/suboptimal levels occur in a variety of conditions including chronic alcoholism. Mammals, including humans, obtain biotin from diet and gut-microbiota via absorption along the intestinal tract. The absorption process is carrier mediated and involves the sodium-dependent multivitamin transporter (SMVT; SLC5A6). We have previously shown that chronic alcohol exposure significantly inhibits intestinal/colonic biotin uptake via suppression of Slc5a6 transcription in animal and cell line models. However, little is known about the transcriptional/epigenetic factors that mediate this suppression. In addition, the effect of alcohol metabolites (generated via alcohol metabolism by gut microbiota and host tissues) on biotin uptake is still unknown. To address these questions, we first demonstrated that chronic alcohol exposure inhibits small intestinal and colonic biotin uptake and SMVT expression in human differentiated enteroid and colonoid monolayers. We then showed that chronic alcohol exposures of both, Caco-2 cells and mice, are associated with a significant suppression in expression of the nuclear factor KLF-4 (needed for Slc5a6 promoter activity), as well as with epigenetic alterations (histone modifications). We also found that chronic exposure of NCM460 human colonic epithelial cells as well as human differentiated colonoid monolayers, to alcohol metabolites (acetaldehyde, ethyl palmitate, ethyl oleate) significantly inhibited biotin uptake and SMVT expression. These findings shed light onto the molecular/epigenetic mechanisms that mediate the inhibitory effect of chronic alcohol exposure on intestinal biotin uptake. They further show that alcohol metabolites are also capable of inhibiting biotin uptake in the gut.NEW & NOTEWORTHY Using complementary models, including human differentiated enteroid and colonoid monolayers, this study shows the involvement of molecular and epigenetic mechanisms in mediating the inhibitory effect of chronic alcohol exposure on biotin uptake along the intestinal tract. The study also shows that alcohol metabolites (generated by gut microbiota and host tissues) cause inhibition in gut biotin uptake.


Subject(s)
Biotin/metabolism , DNA Methylation , Epigenesis, Genetic , Ethanol/pharmacology , Intestinal Mucosa/drug effects , Acetaldehyde/pharmacology , Animals , Caco-2 Cells , Cells, Cultured , Ethanol/metabolism , Humans , Intestinal Mucosa/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mice , Mice, Inbred C57BL , Oleic Acids/pharmacology , Palmitic Acids/pharmacology , Symporters/genetics , Symporters/metabolism
6.
Am J Physiol Gastrointest Liver Physiol ; 319(3): G323-G332, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32683950

ABSTRACT

The water-soluble vitamin B1 (thiamin) plays essential roles in normal metabolism and function of all human/mammalian cells, including the pancreatic acinar cells (PACs). PACs obtain thiamin from their surrounding circulation via transport across the plasma membrane, a process that is mediated by thiamin transporter (THTR)-1 and THTR-2. We have previously characterized different aspects of thiamin uptake by mouse and human primary PACs, but little is known about posttranscriptional regulation of the uptake event. We addressed this by focusing on the predominant thiamin transporter THTR-1 (encoded by SLC19A2 gene) in PACs. Transfecting pmirGLO-SLC19A2 3'-untranslated region (UTR) into mouse-derived PAC 266-6 cells leads to a significant reduction in luciferase activity compared with cells transfected with empty vector. Subjecting the SLC19A2 3'-UTR to different in silico algorithms identified multiple putative microRNA binding sites in this region. Focusing on miR-200a-3p (since it is highly expressed in mouse and human pancreas), we found that transfecting PAC 266-6 and human primary PACs (hPACs) with mimic miR-200a-3p leads to a significant inhibition of THTR-1 expression (both protein and mRNA levels) and in thiamin uptake. In contrast, transfection by miR-200a-3p inhibitor leads to an increase in THTR-1 expression and thiamin uptake. Additionally, truncating the region carrying miR-200a-3p binding site in SLC19A2 3'-UTR and mutating the binding site lead to abrogation in the inhibitory effect of this microRNA on luciferase activity in PAC 266-6. These results demonstrate that expression of THTR-1 and thiamin uptake in PACs is subject to posttranscriptional regulation by microRNAs.NEW & NOTEWORTHY The findings of this study show, for the first time, that the membrane transporter of vitamin B1, i.e., thiamin transporter-1 (THTR-1), is subject to regulation by microRNAs (specifically miR-200a-3p) in mouse and human primary pancreatic acinar cells (PACs). The results also show that this posttranscriptional regulation has functional consequences on the ability of PACs to take in the essential micronutrient thiamin.


Subject(s)
Acinar Cells/metabolism , Membrane Transport Proteins/genetics , MicroRNAs/genetics , Pancreas/metabolism , RNA Processing, Post-Transcriptional/genetics , 3' Untranslated Regions/genetics , Animals , Humans , Mice , Mutation , Primary Cell Culture , Thiamine/metabolism
7.
PLoS One ; 12(4): e0175319, 2017.
Article in English | MEDLINE | ID: mdl-28403226

ABSTRACT

TCTP (Translationally Controlled Tumour Protein) is a multifunctional protein that plays a role in the development, immune system, tumour reversion, and maintenance of stem cells. The mRNA of the Tpt1 gene is over-expressed during liver regeneration. But, the function of the protein in regeneration is not known. To study the role of the protein in regeneration, the earthworm Eudrilus eugeniae was chosen. First, the full length cDNA of the Tpt1 gene was sequenced. The size of the cDNA is 504 bp and the protein has 167 amino acids. The highest level of TCTP expression was documented in the worm after three days of regeneration. The protein was found to be expressed specifically in the epithelial layer of the skin. During regeneration, the protein expression was found to be the highest at the tip of blastema. The pharmacological suppression of TCTP using nutlin-3 and TCTP RNAi experiments resulted in the failure of the regeneration process. The suppression of TCTP caused the arrest of proliferation in posterior amputated worms. The severe cell death was documented in the amputated region of nutlin-3 injected worm. The silencing of TCTP has blocked the modification of clitellar segments. The experiments confirm that TCTP has major functions in the upstream signalling of cell proliferation in the early regeneration process in E. eugeniae.


Subject(s)
Biomarkers, Tumor/metabolism , Oligochaeta/physiology , Proteins/metabolism , Regeneration , Amino Acid Sequence , Animals , Base Sequence , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Cell Proliferation , Gene Expression , Mitosis , Oligochaeta/chemistry , Oligochaeta/genetics , Proteins/analysis , Proteins/genetics , RNA Interference , RNA, Messenger/genetics , RNA, Small Interfering/genetics
8.
Invert Neurosci ; 16(3): 6, 2016 09.
Article in English | MEDLINE | ID: mdl-27279085

ABSTRACT

Earthworms are segmented invertebrates that belong to the phylum Annelida. The segments can be divided into the anterior, clitellar and posterior parts. If the anterior part of the earthworm, which includes the brain, is amputated, the worm would essentially survive even in the absence of the brain. In these brain amputee-derived worms, the nerve cord serves as the primary control center for neurological function. In this current work, we studied changes in the expression levels of anti-acetylated tubulin and serotonin as the indicators of neuro-regenerative processes. The data reveal that the blastemal tissues express the acetylated tubulin and serotonin from day four and that the worm amputated at the 7th segment takes 30 days to complete the regeneration of brain. The ability of self-assemblage is one of the specific functions of the earthworm's brain. The brain amputee restored the ability of self-assemblage on the eighth day.


Subject(s)
Central Nervous System/physiology , Oligochaeta/physiology , Regeneration/physiology , Social Behavior , Animals
9.
Stem Cell Res ; 15(2): 354-364, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26313525

ABSTRACT

Pluripotent stem cells are being actively studied as a cell source for regenerating damaged liver. For long-term survival of engrafting cells in the body, not only do the cells have to execute liver-specific function but also withstand the physical strains and invading pathogens. The cellular innate immune system orchestrated by the interferon (IFN) pathway provides the first line of defense against pathogens. The objective of this study is to assess the innate immune function as well as to systematically profile the IFN-induced genes during hepatic differentiation of pluripotent stem cells. To address this objective, we derived endodermal cells (day 5 post-differentiation), hepatoblast (day 15) and hepatocyte-like cells (day 21) from human embryonic stem cells (hESCs). Day 5, 15 and 21 cells were stimulated with IFN-α and subjected to IFN pathway analysis. Transcriptome analysis was carried out by RNA sequencing. The results showed that the IFN-α treatment activated STAT-JAK pathway in differentiating cells. Transcriptome analysis indicated stage specific expression of classical and non-classical IFN-stimulated genes (ISGs). Subsequent validation confirmed the expression of novel ISGs including RASGRP3, CLMP and TRANK1 by differentiated hepatic cells upon IFN treatment. Hepatitis C virus replication in hESC-derived hepatic cells induced the expression of ISGs--LAMP3, ETV7, RASGRP3, and TRANK1. The hESC-derived hepatic cells contain intact innate system and can recognize invading pathogens. Besides assessing the tissue-specific functions for cell therapy applications, it may also be important to test the innate immune function of engrafting cells to ensure adequate defense against infections and improve graft survival.


Subject(s)
Hepacivirus/genetics , Interferon Type I/metabolism , Pluripotent Stem Cells/cytology , Apolipoprotein B-100/metabolism , Cell Differentiation/drug effects , Cell Line , Cell Lineage , Coxsackie and Adenovirus Receptor-Like Membrane Protein/genetics , Coxsackie and Adenovirus Receptor-Like Membrane Protein/metabolism , Cytokines/genetics , Cytokines/metabolism , Gene Expression Profiling , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Hepacivirus/physiology , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Interferon Type I/genetics , Interferon-alpha/pharmacology , Liver/metabolism , Pluripotent Stem Cells/metabolism , RNA/chemistry , RNA/isolation & purification , RNA/metabolism , Sequence Analysis, RNA , Transcriptome/drug effects , Virus Replication , ras Guanine Nucleotide Exchange Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...