Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 179
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 187(4): 846-860.e17, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38262409

ABSTRACT

RNAs localizing to the outer cell surface have been recently identified in mammalian cells, including RNAs with glycan modifications known as glycoRNAs. However, the functional significance of cell surface RNAs and their production are poorly known. We report that cell surface RNAs are critical for neutrophil recruitment and that the mammalian homologs of the sid-1 RNA transporter are required for glycoRNA expression. Cell surface RNAs can be readily detected in murine neutrophils, the elimination of which substantially impairs neutrophil recruitment to inflammatory sites in vivo and reduces neutrophils' adhesion to and migration through endothelial cells. Neutrophil glycoRNAs are predominantly on cell surface, important for neutrophil-endothelial interactions, and can be recognized by P-selectin (Selp). Knockdown of the murine Sidt genes abolishes neutrophil glycoRNAs and functionally mimics the loss of cell surface RNAs. Our data demonstrate the biological importance of cell surface glycoRNAs and highlight a noncanonical dimension of RNA-mediated cellular functions.


Subject(s)
Endothelial Cells , Neutrophil Infiltration , Neutrophils , RNA , Animals , Mice , Endothelial Cells/metabolism , Neutrophils/metabolism , RNA/chemistry , RNA/metabolism , Nucleotide Transport Proteins/genetics , Nucleotide Transport Proteins/metabolism
2.
Nature ; 588(7836): 174-179, 2020 12.
Article in English | MEDLINE | ID: mdl-32906142

ABSTRACT

Mitochondria require nicotinamide adenine dinucleotide (NAD+) to carry out the fundamental processes that fuel respiration and mediate cellular energy transduction. Mitochondrial NAD+ transporters have been identified in yeast and plants1,2, but their existence in mammals remains controversial3-5. Here we demonstrate that mammalian mitochondria can take up intact NAD+, and identify SLC25A51 (also known as MCART1)-an essential6,7 mitochondrial protein of previously unknown function-as a mammalian mitochondrial NAD+ transporter. Loss of SLC25A51 decreases mitochondrial-but not whole-cell-NAD+ content, impairs mitochondrial respiration, and blocks the uptake of NAD+ into isolated mitochondria. Conversely, overexpression of SLC25A51 or SLC25A52 (a nearly identical paralogue of SLC25A51) increases mitochondrial NAD+ levels and restores NAD+ uptake into yeast mitochondria lacking endogenous NAD+ transporters. Together, these findings identify SLC25A51 as a mammalian transporter capable of importing NAD+ into mitochondria.


Subject(s)
Mitochondria/metabolism , Mitochondrial Proteins/metabolism , NAD/metabolism , Animals , Biological Transport , Cell Line , Cell Respiration/genetics , Genetic Complementation Test , Humans , Mice , Mitochondria/genetics , Mitochondria/pathology , Mitochondrial Proteins/deficiency , Mitochondrial Proteins/genetics , Nucleotide Transport Proteins/genetics , Organic Cation Transport Proteins/deficiency , Organic Cation Transport Proteins/genetics , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics
3.
Proc Natl Acad Sci U S A ; 119(30): e2122158119, 2022 07 26.
Article in English | MEDLINE | ID: mdl-35858418

ABSTRACT

Eicosapentaenoic acid (EPA), an omega-3 (ω-3) polyunsaturated fatty acid, is an essential nutrient that exhibits antiinflammatory, neuroprotective, and cardiovascular-protective activities. Although EPA is used as a nutrient-based pharmaceutical agent or dietary supplement, its molecular target(s) is debatable. Here, we showed that EPA and its metabolites strongly and reversibly inhibit vesicular nucleotide transporter (VNUT), a key molecule for vesicular storage and release of adenosine triphosphate (ATP) in purinergic chemical transmission. In vitro analysis showed that EPA inhibits human VNUT-mediated ATP uptake at a half-maximal inhibitory concentration (IC50) of 67 nM, acting as an allosteric modulator through competition with Cl-. EPA impaired vesicular ATP release from neurons without affecting the vesicular release of other neurotransmitters. In vivo, VNUT-/- mice showed a delay in the onset of neuropathic pain and resistance to both neuropathic and inflammatory pain. EPA potently attenuated neuropathic and inflammatory pain in wild-type mice but not in VNUT-/- mice without affecting the basal nociception. The analgesic effect of EPA was canceled by the intrathecal injection of purinoceptor agonists and was stronger than that of existing drugs used for neuropathic pain treatment, with few side effects. Neuropathic pain impaired insulin sensitivity in previous studies, which was improved by EPA in the wild-type mice but not in the VNUT-/- mice. Our results showed that VNUT is a molecular target of EPA that attenuates neuropathic and inflammatory pain and insulin resistance. EPA may represent a unique nutrient-based treatment and prevention strategy for neurological, immunological, and metabolic diseases by targeting purinergic chemical transmission.


Subject(s)
Eicosapentaenoic Acid , Neuralgia , Nucleotide Transport Proteins , Adenosine Triphosphate/metabolism , Animals , Eicosapentaenoic Acid/pharmacology , Eicosapentaenoic Acid/therapeutic use , Humans , Insulin Resistance , Mice , Neuralgia/drug therapy , Neuralgia/genetics , Nociception , Nucleotide Transport Proteins/antagonists & inhibitors , Nucleotide Transport Proteins/genetics , Nucleotide Transport Proteins/metabolism
4.
J Biol Chem ; 299(12): 105406, 2023 12.
Article in English | MEDLINE | ID: mdl-38270391

ABSTRACT

Slc35c1 encodes an antiporter that transports GDP-fucose into the Golgi and returns GMP to the cytoplasm. The closely related gene Slc35c2 encodes a putative GDP-fucose transporter and promotes Notch fucosylation and Notch signaling in cultured cells. Here, we show that HEK293T cells lacking SLC35C1 transferred reduced amounts of O-fucose to secreted epidermal growth factor-like repeats from NOTCH1 or secreted thrombospondin type I repeats from thrombospondin 1. However, cells lacking SLC35C2 did not exhibit reduced fucosylation of these epidermal growth factor-like repeats or thrombospondin type I repeats. To investigate SLC35C2 functions in vivo, WW6 embryonic stem cells were targeted for Slc35c2. Slc35c2[-/-] mice were viable and fertile and exhibited no evidence of defective Notch signaling during skeletal or T cell development. By contrast, mice with inactivated Slc35c1 exhibited perinatal lethality and marked skeletal defects in late embryogenesis, typical of defective Notch signaling. Compound Slc35c1[-/-]Slc35c2[-/-] mutants were indistinguishable in skeletal phenotype from Slc35c1[-/-] embryos and neonates. Double mutants did not exhibit the exacerbated skeletal defects predicted if SLC35C2 was functionally important for Notch signaling in vivo. In addition, NOTCH1 immunoprecipitated from Slc35c1[-/-]Slc35c2[-/-] neonatal lung carried fucose detected by binding of Aleuria aurantia lectin. Given that the absence of both SLC35C1, a known GDP-fucose transporter, and SLC35C2, a putative GDP-fucose transporter, did not lead to afucosylated NOTCH1 nor to the severe Notch signaling defects and embryonic lethality expected if all GDP-fucose transport were abrogated, at least one more mechanism of GDP-fucose transport into the secretory pathway must exist in mammals.


Subject(s)
Fucose , Monosaccharide Transport Proteins , Nucleotide Transport Proteins , Animals , Female , Humans , Mice , Pregnancy , Epidermal Growth Factor , Fucose/metabolism , HEK293 Cells , Monosaccharide Transport Proteins/genetics , Neoplasm Proteins , Nucleotide Transport Proteins/genetics , Thrombospondins/metabolism , Mice, Knockout , Receptor, Notch1/metabolism , Signal Transduction
5.
J Virol ; 97(3): e0146322, 2023 03 30.
Article in English | MEDLINE | ID: mdl-36779754

ABSTRACT

Entry of influenza A viruses (IAVs) into host cells is initiated by binding to sialic acids (Sias), their primary host cell receptor, followed by endocytosis and membrane fusion to release the viral genome into the cytoplasm of the host cell. Host tropism is affected by these entry processes, with a primary factor being receptor specificity. Sias exist in several different chemical forms, including the hydroxylated N-glycolylneuraminic acid (Neu5Gc), which is found in many hosts; however, it has not been clear how modified Sias affect viral binding and entry. Neu5Gc is commonly found in many natural influenza hosts, including pigs and horses, but not in humans or ferrets. Here, we engineered HEK293 cells to express the hydoxylase gene (CMAH) that converts Neu5Ac to Neu5Gc, or knocked out the Sia-CMP transport gene (SLC35A1), resulting in cells that express 95% Neu5Gc or minimal level of Sias, respectively. H3N2 (X-31) showed significantly reduced infectivity in Neu5Gc-rich cells compared to wild-type HEK293 (>95% Neu5Ac). To determine the effects on binding and fusion, we generated supported lipid bilayers (SLBs) derived from the plasma membranes of these cells and carried out single particle microscopy. H3N2 (X-31) exhibited decreased binding to Neu5Gc-containing SLBs, but no significant difference in H3N2 (X-31)'s fusion kinetics to either SLB type, suggesting that reduced receptor binding does not affect subsequent membrane fusion. This finding suggests that for this virus to adapt to host cells rich in Neu5Gc, only receptor affinity changes are required without further adaptation of virus fusion machinery. IMPORTANCE Influenza A virus (IAV) infections continue to threaten human health, causing over 300,000 deaths yearly. IAV infection is initiated by the binding of influenza glycoprotein hemagglutinin (HA) to host cell sialic acids (Sias) and the subsequent viral-host membrane fusion. Generally, human IAVs preferentially bind to the Sia N-acetylneuraminic acid (Neu5Ac). Yet, other mammalian hosts, including pigs, express diverse nonhuman Sias, including N-glycolylneuraminic acid (Neu5Gc). The role of Neu5Gc in human IAV infections in those hosts is not well-understood, and the variant form may play a role in incidents of cross-species transmission and emergence of new epidemic variants. Therefore, it is important to investigate how human IAVs interact with Neu5Ac and Neu5Gc. Here, we use membrane platforms that mimic the host cell surface to examine receptor binding and membrane fusion events of human IAV H3N2. Our findings improve the understanding of viral entry mechanisms that can affect host tropism and virus evolution.


Subject(s)
Host Microbial Interactions , Influenza A Virus, H3N2 Subtype , Sialic Acids , Virus Internalization , Animals , Humans , HEK293 Cells , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/metabolism , Membrane Fusion , Nucleotide Transport Proteins/genetics , Nucleotide Transport Proteins/metabolism , Sialic Acids/chemistry , Sialic Acids/pharmacology , Single Molecule Imaging , Virus Attachment/drug effects , Virus Internalization/drug effects , Host Microbial Interactions/genetics , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/virology
6.
Int J Mol Sci ; 25(12)2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38928424

ABSTRACT

The SLC35 (Solute Carrier 35) family members acting as nucleotide sugar transporters are typically localized in the endoplasmic reticulum or Golgi apparatus. It is, therefore, intriguing that some reports document the presence of orphan transporters SLC35F1 and SLC35F6 within the endosomal and lysosomal system. Here, we compared the subcellular distribution of these proteins and found that they are concentrated in separate compartments; i.e., recycling endosomes for SLC35F1 and lysosomes for SLC35F6. Swapping the C-terminal tail of these proteins resulted in a switch of localization, with SLC35F1 being trafficked to lysosomes while SLC35F6 remained in endosomes. This suggested the presence of specific sorting signals in these C-terminal regions. Using site-directed mutagenesis, fluorescence microscopy, and cell surface biotinylation assays, we found that the EQERLL360 signal located in the cytoplasmic tail of human SLC35F6 is involved in its lysosomal sorting (as previously shown for this conserved sequence in mouse SLC35F6), and that SLC35F1 localization in the recycling pathway depends on two YXXΦ-type signals: a Y367KQF sequence facilitates its internalization from the plasma membrane, while a Y392TSL motif prevents its transport to lysosomes, likely by promoting SLC35F1 recycling to the cell surface. Taken together, these results support that some SLC35 members may function at different levels of the endosomal and lysosomal system.


Subject(s)
Endosomes , Lysosomes , Humans , Amino Acid Sequence , Cell Membrane/metabolism , Endosomes/metabolism , Golgi Apparatus/metabolism , HEK293 Cells , HeLa Cells , Lysosomes/metabolism , Nucleotide Transport Proteins/metabolism , Nucleotide Transport Proteins/genetics , Protein Sorting Signals , Protein Transport
7.
J Neurochem ; 166(4): 705-719, 2023 08.
Article in English | MEDLINE | ID: mdl-37522158

ABSTRACT

The molecular mechanisms underlying posttraumatic stress disorder (PTSD) are yet to be fully elucidated, especially in underrepresented population groups. Expression quantitative trait loci (eQTLs) are DNA sequence variants that influence gene expression, in a local (cis-) or distal (trans-) manner, and subsequently impact cellular, tissue, and system physiology. This study aims to identify genetic loci associated with gene expression changes in a South African PTSD cohort. Genome-wide genotype and RNA-sequencing data were obtained from 32 trauma-exposed controls and 35 PTSD cases of mixed-ancestry, as part of the SHARED ROOTS project. The first approach utilised 108 937 single-nucleotide polymorphisms (SNPs) (MAF > 10%) and 11 312 genes with Matrix eQTL to map potential eQTLs, while controlling for covariates as appropriate. The second analysis was focused on 5638 SNPs related to a previously calculated PTSD polygenic risk score for this cohort. SNP-gene pairs were considered eQTLs if they surpassed Bonferroni correction and had a false discovery rate <0.05. We did not identify eQTLs that significantly influenced gene expression in a PTSD-dependent manner. However, several known cis-eQTLs, independent of PTSD diagnosis, were observed. rs8521 (C > T) was associated with TAGLN and SIDT2 expression, and rs11085906 (C > T) was associated with ZNF333 expression. This exploratory study provides insight into the molecular mechanisms associated with PTSD in a non-European, admixed sample population. This study was limited by the cross-sectional design and insufficient statistical power. Overall, this study should encourage further multi-omics approaches towards investigating PTSD in diverse populations.


Subject(s)
Nucleotide Transport Proteins , Stress Disorders, Post-Traumatic , Humans , Stress Disorders, Post-Traumatic/genetics , Cross-Sectional Studies , South Africa , Quantitative Trait Loci/genetics , Gene Expression , Polymorphism, Single Nucleotide/genetics , Genome-Wide Association Study , Gene Expression Regulation , Nucleotide Transport Proteins/genetics
8.
J Virol ; 96(24): e0162622, 2022 12 21.
Article in English | MEDLINE | ID: mdl-36453883

ABSTRACT

Porcine deltacoronavirus (PDCoV), an emerging enteropathogenic coronavirus, not only causes diarrhea in piglets but also possesses the potential to infect humans. To better understand host-virus genetic dependencies and find potential therapeutic targets for PDCoV, we used a porcine single-guide RNA (sgRNA) lentivirus library to screen host factors related to PDCoV infection in LLC-PK1 cells. The solute carrier family 35 member A1 (SLC35A1), a key molecule in the sialic acid (SA) synthesis pathway, was identified as a host factor required for PDCoV infection. A knockout of SLC35A1 caused decreases in the amounts of cell surface sialic acid (SA) and viral adsorption; meanwhile, trypsin promoted the use of SA in PDCoV infection. By constructing and assessing a series of recombinant PDCoV strains with the deletion or mutation of possible critical domain or amino acid residues for SA binding in the S1 N-terminal domain, we found that S T182 might be a PDCoV SA-binding site. However, the double knockout of SLC35A1 and amino peptidase N (APN) could not block PDCoV infection completely. Additionally, we found that different swine enteric coronaviruses, including transmissible gastroenteritis coronavirus, porcine epidemic diarrhea virus, and swine acute diarrhea syndrome coronavirus, are differentially dependent on SA. Overall, our study uncovered a collection of host factors that can be exploited as drug targets against PDCoV infection and deepened our understanding of the relationship between PDCoV and SA. IMPORTANCE Identifying the host factors required for replication will be helpful to uncover the pathogenesis mechanisms and develop antivirals against the emerging coronavirus porcine deltacoronavirus (PDCoV). Herein, we performed a genome-wide clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9 knockout screen, the results of which revealed that the solute carrier family 35 member A1 (SLC35A1) is a host factor required for PDCoV infection that acts by regulating cell surface sialic acid (SA). We also identified the T182 site in the N-terminal domain of PDCoV S1 subunit as being associated with the SA-binding site and found that trypsin promotes the use of cell surface SA by PDCoV. Furthermore, different swine enteric coronaviruses use SLC35A1 differently for infection. This is the first study to screen host factors required for PDCoV replication using a genome-wide CRISPR-Cas9 functional knockout, thereby providing clues for developing antiviral drugs against PDCoV infection.


Subject(s)
Coronavirus Infections , Host Microbial Interactions , Nucleotide Transport Proteins , Swine Diseases , Animals , Humans , Adsorption , Coronavirus , Coronavirus Infections/physiopathology , CRISPR-Cas Systems , N-Acetylneuraminic Acid/metabolism , Nucleotide Transport Proteins/genetics , Nucleotide Transport Proteins/metabolism , Swine , Swine Diseases/physiopathology , Trypsin , Host Microbial Interactions/genetics , Protein Domains , Binding Sites
9.
J Biol Chem ; 297(3): 101069, 2021 09.
Article in English | MEDLINE | ID: mdl-34384782

ABSTRACT

The CMP-sialic acid transporter SLC35A1 and UDP-galactose transporter SLC35A2 are two well-characterized nucleotide sugar transporters with distinctive substrate specificities. Mutations in either induce congenital disorders of glycosylation. Despite the biomedical relevance, mechanisms of substrate specificity are unclear. To address this critical issue, we utilized a structure-guided mutagenesis strategy and assayed a series of SLC35A2 and SLC35A1 mutants using a rescue approach. Our results suggest that three pockets in the central cavity of each transporter provide substrate specificity. The pockets comprise (1) nucleobase (residues E52, K55, and Y214 of SLC35A1; E75, K78, N235, and G239 of SLC35A2); (2) middle (residues Q101, N102, and T260 of SLC35A1; Q125, N126, Q129, Y130, and Q278 of SLC35A2); and (3) sugar (residues K124, T128, S188, and K272 of SLC35A1; K148, T152, S213, and K297 of SLC35A2) pockets. Within these pockets, two components appear to be especially critical for substrate specificity. Y214 (for SLC35A1) and G239 (for SLC35A2) in the nucleobase pocket appear to discriminate cytosine from uracil. Furthermore, Q129 and Q278 of SLC35A2 in the middle pocket appear to interact specifically with the ß-phosphate of UDP while the corresponding A105 and A253 residues in SLC35A1 do not interact with CMP, which lacks a ß-phosphate. Overall, our findings contribute to a molecular understanding of substrate specificity and coordination in SLC35A1 and SLC35A2 and have important implications for the understanding and treatment of diseases associated with mutations or dysregulations of these two transporters.


Subject(s)
Monosaccharide Transport Proteins/genetics , Nucleotide Transport Proteins/genetics , Animals , CHO Cells , Cricetulus , Glycosylation , HeLa Cells , Humans , Membrane Transport Proteins/metabolism , Monosaccharide Transport Proteins/metabolism , Monosaccharide Transport Proteins/ultrastructure , Mutation , Nucleotide Transport Proteins/metabolism , Nucleotide Transport Proteins/ultrastructure , Nucleotides/metabolism , Substrate Specificity
10.
Mol Pain ; 18: 17448069221089592, 2022.
Article in English | MEDLINE | ID: mdl-35266813

ABSTRACT

Phantom tooth pain (PTP) is a rare and specific neuropathic pain that occurs after pulpectomy and tooth extraction, but its cause is not understood. We hypothesized that there is a genetic contribution to PTP. We focused on solute carrier family 17 member 9 (SLC17A9)/vesicular nucleotide transporter (VNUT) and purinergic receptor P2Y12 (P2RY12), both of which have been associated with neuropathic pain and pain transduction signaling in the trigeminal ganglion in rodents. We sought to corroborate these associations in humans. We investigated gene polymorphisms that contribute to PTP. We statistically examined the association between genetic polymorphisms and PTP vulnerability in 150 patients with orofacial pain, including PTP, and 500 healthy subjects. We found that the rs735055 polymorphism of the SLC17A9 gene and rs3732759 polymorphism of the P2RY12 gene were associated with the development of PTP. Carriers of the minor allele of rs735055 and individuals who were homozygous for the major allele of rs3732759 had a higher rate of PTP. Carriers of the minor allele of rs735055 reportedly had high SLC17A9 mRNA expression in the spinal cord, which may increase the storage and release of adenosine triphosphate. Individuals who were homozygous for the major allele of rs3732759 may have higher P2RY12 expression that is more active in microglia. Therefore, these carriers may be more susceptible to PTP. These results suggest that specific genetic polymorphisms of the SLC17A9 and P2RY12 genes are involved in PTP. This is the first report on genes that are associated with PTP in humans.


Subject(s)
Neuralgia , Nucleotide Transport Proteins , Humans , Adenosine Triphosphate/metabolism , Nucleotide Transport Proteins/genetics , Nucleotide Transport Proteins/metabolism , Polymorphism, Single Nucleotide/genetics , Receptors, Purinergic P2Y12/genetics , Receptors, Purinergic P2Y12/metabolism
11.
FASEB J ; 35(4): e21223, 2021 04.
Article in English | MEDLINE | ID: mdl-33715196

ABSTRACT

The role of Sidt2 in the process of glucose and lipid metabolism has been recently reported. However, whether Sidt2 is involved in the metabolic regulation in skeletal muscle remains unknown. In this study, for the first time, using skeletal muscle-selective Sidt2 knockout mice, we found that Sidt2 was vital for the quality control of mitochondria in mouse skeletal muscle. These mice showed significantly reduced muscle tolerance and structurally abnormal mitochondria. Deletion of the Sidt2 gene resulted in decreased expression of mitochondrial fusion protein 2 (Mfn2) and Dynamin-related protein 1 (Drp1), as well as peroxisome proliferator-activated receptor γ coactivator-1 (PGC1-α). In addition, the clearance of damaged mitochondria in skeletal muscle was inhibited upon Sidt2 deletion, which was caused by blockade of autophagy flow. Mechanistically, the fusion of autophagosomes and lysosomes was compromised in Sidt2 knockout skeletal muscle cells. In summary, the deletion of the Sidt2 gene not only interfered with the quality control of mitochondria, but also inhibited the clearance of mitochondria and caused the accumulation of a large number of damaged mitochondria, ultimately leading to the abnormal structure and function of skeletal muscle.


Subject(s)
Cell Membrane , Lysosomes , Muscle, Skeletal/metabolism , Nucleotide Transport Proteins/metabolism , Animals , Autophagy/physiology , Cell Line , Gene Expression Regulation , Genetic Predisposition to Disease , Mice , Mice, Knockout , Microscopy, Electron, Transmission , Mitochondria, Muscle/metabolism , Muscle, Skeletal/cytology , Muscular Diseases/genetics , Nucleotide Transport Proteins/genetics
12.
Arterioscler Thromb Vasc Biol ; 41(9): 2494-2508, 2021 09.
Article in English | MEDLINE | ID: mdl-34233476

ABSTRACT

Objective: Low HDL-C (high-density lipoprotein cholesterol) is the most frequent dyslipidemia in Mexicans, but few studies have examined the underlying genetic basis. Our purpose was to identify genetic variants associated with HDL-C levels and cardiovascular risk in the Mexican population. Approach and Results: A genome-wide association studies for HDL-C levels in 2335 Mexicans, identified four loci associated with genome-wide significance: CETP, ABCA1, LIPC, and SIDT2. The SIDT2 missense Val636Ile variant was associated with HDL-C levels and was replicated in 3 independent cohorts (P=5.9×10−18 in the conjoint analysis). The SIDT2/Val636Ile variant is more frequent in Native American and derived populations than in other ethnic groups. This variant was also associated with increased ApoA1 and glycerophospholipid serum levels, decreased LDL-C (low-density lipoprotein cholesterol) and ApoB levels, and a lower risk of premature CAD. Because SIDT2 was previously identified as a protein involved in sterol transport, we tested whether the SIDT2/Ile636 protein affected this function using an in vitro site-directed mutagenesis approach. The SIDT2/Ile636 protein showed increased uptake of the cholesterol analog dehydroergosterol, suggesting this variant affects function. Finally, liver transcriptome data from humans and the Hybrid Mouse Diversity Panel are consistent with the involvement of SIDT2 in lipid and lipoprotein metabolism. Conclusions: This is the first genome-wide association study for HDL-C levels seeking associations with coronary artery disease in the Mexican population. Our findings provide new insight into the genetic architecture of HDL-C and highlight SIDT2 as a new player in cholesterol and lipoprotein metabolism in humans.


Subject(s)
Cholesterol, HDL/blood , Coronary Artery Disease/genetics , Hyperlipoproteinemia Type II/genetics , Nucleotide Transport Proteins/genetics , Polymorphism, Single Nucleotide , Adult , Age of Onset , Animals , Biomarkers/blood , Case-Control Studies , Child , Coronary Artery Disease/blood , Coronary Artery Disease/diagnosis , Coronary Artery Disease/epidemiology , Disease Models, Animal , Female , Genetic Predisposition to Disease , Genome-Wide Association Study , HEK293 Cells , Heart Disease Risk Factors , Humans , Hyperlipoproteinemia Type II/blood , Hyperlipoproteinemia Type II/diagnosis , Hyperlipoproteinemia Type II/epidemiology , Male , Mendelian Randomization Analysis , Mexico/epidemiology , Mice , Middle Aged , Nucleotide Transport Proteins/metabolism , Phenotype , Risk Assessment
13.
J Biol Chem ; 295(48): 16445-16463, 2020 11 27.
Article in English | MEDLINE | ID: mdl-32938718

ABSTRACT

Nucleotide sugar transporters, encoded by the SLC35 gene family, deliver nucleotide sugars throughout the cell for various glycosyltransferase-catalyzed glycosylation reactions. GlcNAc, in the form of UDP-GlcNAc, and galactose, as UDP-Gal, are delivered into the Golgi apparatus by SLC35A3 and SLC35A2 transporters, respectively. However, although the UDP-Gal transporting activity of SLC35A2 has been clearly demonstrated, UDP-GlcNAc delivery by SLC35A3 is not fully understood. Therefore, we analyzed a panel of CHO, HEK293T, and HepG2 cell lines including WT cells, SLC35A2 knockouts, SLC35A3 knockouts, and double-knockout cells. Cells lacking SLC35A2 displayed significant changes in N- and O-glycan synthesis. However, in SLC35A3-knockout CHO cells, only limited changes were observed; GlcNAc was still incorporated into N-glycans, but complex type N-glycan branching was impaired, although UDP-GlcNAc transport into Golgi vesicles was not decreased. In SLC35A3-knockout HEK293T cells, UDP-GlcNAc transport was significantly decreased but not completely abolished. However, N-glycan branching was not impaired in these cells. In CHO and HEK293T cells, the effect of SLC35A3 deficiency on N-glycan branching was potentiated in the absence of SLC35A2. Moreover, in SLC35A3-knockout HEK293T and HepG2 cells, GlcNAc was still incorporated into O-glycans. However, in the case of HepG2 cells, no qualitative changes in N-glycans between WT and SLC35A3 knockout cells nor between SLC35A2 knockout and double-knockout cells were observed. These findings suggest that SLC35A3 may not be the primary UDP-GlcNAc transporter and/or different mechanisms of UDP-GlcNAc transport into the Golgi apparatus may exist.


Subject(s)
Glycosyltransferases/metabolism , Golgi Apparatus/metabolism , Nucleotide Transport Proteins/metabolism , Polysaccharides/biosynthesis , Animals , CHO Cells , Cricetulus , Gene Knockdown Techniques , Glycosyltransferases/genetics , Golgi Apparatus/genetics , HEK293 Cells , Hep G2 Cells , Humans , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Monosaccharide Transport Proteins/genetics , Monosaccharide Transport Proteins/metabolism , Nucleotide Transport Proteins/genetics , Polysaccharides/genetics
14.
J Virol ; 95(2)2020 12 22.
Article in English | MEDLINE | ID: mdl-33087464

ABSTRACT

Engagement of cell surface receptors by viruses is a critical determinant of viral tropism and disease. The reovirus attachment protein σ1 binds sialylated glycans and proteinaceous receptors to mediate infection, but the specific requirements for different cell types are not entirely known. To identify host factors required for reovirus-induced cell death, we conducted a CRISPR-knockout screen targeting over 20,000 genes in murine microglial BV2 cells. Candidate genes required for reovirus to cause cell death were highly enriched for sialic acid synthesis and transport. Two of the top candidates identified, CMP N-acetylneuraminic acid synthetase (Cmas) and solute carrier family 35 member A1 (Slc35a1), promote sialic acid expression on the cell surface. Two reovirus strains that differ in the capacity to bind sialic acid, T3SA+ and T3SA-, were used to evaluate Cmas and Slc35a1 as potential host genes required for reovirus infection. Following CRISPR-Cas9 disruption of either gene, cell surface expression of sialic acid was diminished. These results correlated with decreased binding of strain T3SA+, which is capable of engaging sialic acid. Disruption of either gene did not alter the low-level binding of T3SA-, which does not engage sialic acid. Furthermore, infectivity of T3SA+ was diminished to levels similar to those of T3SA- in cells lacking Cmas and Slc35a1 by CRISPR ablation. However, exogenous expression of Cmas and Slc35a1 into the respective null cells restored sialic acid expression and T3SA+ binding and infectivity. These results demonstrate that Cmas and Slc35a1, which mediate cell surface expression of sialic acid, are required in murine microglial cells for efficient reovirus binding and infection.IMPORTANCE Attachment factors and receptors are important determinants of dissemination and tropism during reovirus-induced disease. In a CRISPR cell survival screen, we discovered two genes, Cmas and Slc35a1, which encode proteins required for sialic acid expression on the cell surface and mediate reovirus infection of microglial cells. This work elucidates host genes that render microglial cells susceptible to reovirus infection and expands current understanding of the receptors on microglial cells that are engaged by reovirus. Such knowledge may lead to new strategies to selectively target microglial cells for oncolytic applications.


Subject(s)
N-Acylneuraminate Cytidylyltransferase/metabolism , Nucleotide Transport Proteins/metabolism , Reoviridae Infections/virology , Reoviridae/physiology , Animals , Capsid Proteins/genetics , Capsid Proteins/metabolism , Cell Line , Cell Membrane/metabolism , Cell Survival , Mice , N-Acetylneuraminic Acid/metabolism , N-Acylneuraminate Cytidylyltransferase/genetics , Nucleotide Transport Proteins/genetics , Receptors, Virus/metabolism , Reoviridae/genetics , Reoviridae/metabolism , Reoviridae Infections/metabolism , Virus Attachment , Virus Replication
15.
Haematologica ; 106(3): 759-769, 2021 03 01.
Article in English | MEDLINE | ID: mdl-32303557

ABSTRACT

Sialic acid is a common terminal residue of glycans on proteins and acidic sphingolipids such as gangliosides and has important biological functions. The sialylation process is controlled by more than 20 different sialyltransferases, many of which exhibit overlapping functions. Thus, it is difficult to determine the overall biological function of sialylation by targeted deletion of individual sialyltransferases. To address this issue, we established a mouse line with the Slc35a1 gene flanked by loxP sites. Slc35a1 encodes the cytidine-5'-monophosphate (CMP)-sialic acid transporter that transports CMP-sialic acid from the cytoplasm into the Golgi apparatus for sialylation. Here we report our study regarding the role of sialylation on megakaryocytes and platelets using a mouse line with significantly reduced sialylation in megakaryocytes and platelets (Plt Slc35a1­ /­). The major phenotype of Plt Slc35a1­/­ mice was thrombocytopenia. The number of bone marrow megakaryocytes in Plt Slc35a1­/­ mice was reduced, and megakaryocyte maturation was also impaired. In addition, an increased number of desialylated platelets was cleared by Küpffer cells in the liver of Plt Slc35a1­/­ mice. This study provides new insights into the role of sialylation in platelet homeostasis and the mechanisms of thrombocytopenia in diseases associated with platelet desialylation, such as immune thrombocytopenia and a rare congenital disorder of glycosylation (CDG), SLC35A1-CDG, which is caused by SLC35A1 mutations.


Subject(s)
Nucleotide Transport Proteins , Thrombocytopenia , Blood Platelets , Humans , Liver , N-Acetylneuraminic Acid , Nucleotide Transport Proteins/genetics , Thrombocytopenia/genetics , Thrombopoiesis
16.
J Immunol ; 202(12): 3483-3492, 2019 06 15.
Article in English | MEDLINE | ID: mdl-31061008

ABSTRACT

dsRNA is a common by-product of viral replication and acts as a potent trigger of antiviral immunity. SIDT1 and SIDT2 are closely related members of the SID-1 transmembrane family. SIDT2 functions as a dsRNA transporter and is required to traffic internalized dsRNA from endocytic compartments into the cytosol for innate immune activation, but the role of SIDT1 in dsRNA transport and in the innate immune response to viral infection is unclear. In this study, we show that Sidt1 expression is upregulated in response to dsRNA and type I IFN exposure and that SIDT1 interacts with SIDT2. Moreover, similar to SIDT2, SIDT1 localizes to the endolysosomal compartment, interacts with the long dsRNA analog poly(I:C), and, when overexpressed, enhances endosomal escape of poly(I:C) in vitro. To elucidate the role of SIDT1 in vivo, we generated SIDT1-deficient mice. Similar to Sidt2-/- mice, SIDT1-deficient mice produced significantly less type I IFN following infection with HSV type 1. In contrast to Sidt2-/- mice, however, SIDT1-deficient animals showed no impairment in survival postinfection with either HSV type 1 or encephalomyocarditis virus. Consistent with this, we observed that, unlike SIDT2, tissue expression of SIDT1 was relatively restricted, suggesting that, whereas SIDT1 can transport extracellular dsRNA into the cytoplasm following endocytosis in vitro, the transport activity of SIDT2 is likely to be functionally dominant in vivo.


Subject(s)
Cardiovirus Infections/immunology , Cytoplasm/metabolism , Encephalomyocarditis virus/physiology , Endosomes/metabolism , Herpes Simplex/immunology , Herpesvirus 1, Human/physiology , Lysosomes/metabolism , Membrane Transport Proteins/metabolism , Nucleotide Transport Proteins/metabolism , Animals , Cells, Cultured , DNA/immunology , Membrane Transport Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Nucleotide Transport Proteins/genetics , Poly I-C/immunology , RNA Transport/genetics
17.
Proc Natl Acad Sci U S A ; 115(45): E10778-E10787, 2018 11 06.
Article in English | MEDLINE | ID: mdl-30352850

ABSTRACT

Matching ATP:NADPH provision and consumption in the chloroplast is a prerequisite for efficient photosynthesis. In terms of ATP:NADPH ratio, the amount of ATP generated from the linear electron flow does not meet the demand of the Calvin-Benson-Bassham (CBB) cycle. Several different mechanisms to increase ATP availability have evolved, including cyclic electron flow in higher plants and the direct import of mitochondrial-derived ATP in diatoms. By imaging a fluorescent ATP sensor protein expressed in living Arabidopsis thaliana seedlings, we found that MgATP2- concentrations were lower in the stroma of mature chloroplasts than in the cytosol, and exogenous ATP was able to enter chloroplasts isolated from 4- and 5-day-old seedlings, but not chloroplasts isolated from 10- or 20-day-old photosynthetic tissues. This observation is in line with the previous finding that the expression of chloroplast nucleotide transporters (NTTs) in Arabidopsis mesophyll is limited to very young seedlings. Employing a combination of photosynthetic and respiratory inhibitors with compartment-specific imaging of ATP, we corroborate the dependency of stromal ATP production on mitochondrial dissipation of photosynthetic reductant. Our data suggest that, during illumination, the provision and consumption of ATP:NADPH in chloroplasts can be balanced by exporting excess reductants rather than importing ATP from the cytosol.


Subject(s)
Adenosine Triphosphate/metabolism , Arabidopsis/metabolism , Chloroplasts/metabolism , Gene Expression Regulation, Plant , Mitochondria/metabolism , Photosynthesis/genetics , Plant Leaves/metabolism , Arabidopsis/genetics , Arabidopsis/growth & development , Biological Transport , Biosensing Techniques/methods , Chloroplasts/genetics , Cytosol/metabolism , Fluorescence Resonance Energy Transfer , Gene Expression Regulation, Developmental , Genes, Reporter , Light , NADP/metabolism , Nucleotide Transport Proteins/genetics , Nucleotide Transport Proteins/metabolism , Oxidation-Reduction , Plant Leaves/genetics , Plant Leaves/growth & development , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Seedlings/genetics , Seedlings/growth & development , Seedlings/metabolism , Signal Transduction
18.
Plant Cell Physiol ; 61(5): 897-908, 2020 May 01.
Article in English | MEDLINE | ID: mdl-32065636

ABSTRACT

Despite the fundamental importance of nicotinamide adenine dinucleotide (NAD+) for metabolism, the physiological roles of NAD+ carriers in plants remain unclear. We previously characterized the Arabidopsis thaliana gene (At1g25380), named AtNDT2, encoding a protein located in the mitochondrial inner membrane, which imports NAD+ from the cytosol using ADP and AMP as counter-exchange substrates for NAD+. Here, we further investigated the physiological roles of NDT2, by isolating a T-DNA insertion line, generating an antisense line and characterizing these genotypes in detail. Reduced NDT2 expression affected reproductive phase by reducing total seed yield. In addition, reduced seed germination and retardation in seedling establishment were observed in the mutant lines. Moreover, remarkable changes in primary metabolism were observed in dry and germinated seeds and an increase in fatty acid levels was verified during seedling establishment. Furthermore, flowers and seedlings of NDT2 mutants displayed upregulation of de novo and salvage pathway genes encoding NAD+ biosynthesis enzymes, demonstrating the transcriptional control mediated by NDT2 activity over these genes. Taken together, our results suggest that NDT2 expression is fundamental for maintaining NAD+ balance amongst organelles that modulate metabolism, physiology and developmental processes of heterotrophic tissues.


Subject(s)
Arabidopsis Proteins/genetics , Down-Regulation/genetics , Gene Expression Regulation, Plant , Germination/genetics , Mitochondria/metabolism , Mitochondrial Proteins/genetics , NAD/metabolism , Nucleotide Transport Proteins/genetics , Seeds/growth & development , Seeds/genetics , Arabidopsis Proteins/metabolism , Biological Transport , Flowers/physiology , Genotype , Heterotrophic Processes , Mitochondrial Proteins/metabolism , Nucleotide Transport Proteins/metabolism , Nucleotides/metabolism , Pyridines/metabolism , Reproduction/physiology
19.
PLoS Pathog ; 14(1): e1006765, 2018 01.
Article in English | MEDLINE | ID: mdl-29346417

ABSTRACT

Cryptococcus neoformans, an AIDS-defining opportunistic pathogen, is the leading cause of fungal meningitis worldwide and is responsible for hundreds of thousands of deaths annually. Cryptococcal glycans are required for fungal survival in the host and for pathogenesis. Most glycans are made in the secretory pathway, although the activated precursors for their synthesis, nucleotide sugars, are made primarily in the cytosol. Nucleotide sugar transporters are membrane proteins that solve this topological problem, by exchanging nucleotide sugars for the corresponding nucleoside phosphates. The major virulence factor of C. neoformans is an anti-phagocytic polysaccharide capsule that is displayed on the cell surface; capsule polysaccharides are also shed from the cell and impede the host immune response. Xylose, a neutral monosaccharide that is absent from model yeast, is a significant capsule component. Here we show that Uxt1 and Uxt2 are both transporters specific for the xylose donor, UDP-xylose, although they exhibit distinct subcellular localization, expression patterns, and kinetic parameters. Both proteins also transport the galactofuranose donor, UDP-galactofuranose. We further show that Uxt1 and Uxt2 are required for xylose incorporation into capsule and protein; they are also necessary for C. neoformans to cause disease in mice, although surprisingly not for fungal viability in the context of infection. These findings provide a starting point for deciphering the substrate specificity of an important class of transporters, elucidate a synthetic pathway that may be productively targeted for therapy, and contribute to our understanding of fundamental glycobiology.


Subject(s)
Cryptococcus neoformans/metabolism , Fungal Proteins/metabolism , Glycoproteins/metabolism , Nucleotide Transport Proteins/metabolism , Uridine Diphosphate Xylose/metabolism , Animals , Biological Transport , Cryptococcosis/microbiology , Cryptococcosis/pathology , Cryptococcus neoformans/pathogenicity , Cryptococcus neoformans/ultrastructure , Female , Fungal Capsules/metabolism , Fungal Capsules/ultrastructure , Fungal Proteins/genetics , Galactose/analogs & derivatives , Galactose/metabolism , Gene Deletion , Gene Expression Regulation, Fungal , Glycoproteins/genetics , Kinetics , Mice , Microscopy, Electron, Transmission , Mutation , Nucleotide Transport Proteins/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Transport , Uridine Diphosphate/analogs & derivatives , Uridine Diphosphate/metabolism , Virulence
20.
Plant Cell ; 29(1): 129-143, 2017 01.
Article in English | MEDLINE | ID: mdl-28062750

ABSTRACT

UDP-glucuronic acid (UDP-GlcA) is the precursor of many plant cell wall polysaccharides and is required for production of seed mucilage. Following synthesis in the cytosol, it is transported into the lumen of the Golgi apparatus, where it is converted to UDP-galacturonic acid (UDP-GalA), UDP-arabinose, and UDP-xylose. To identify the Golgi-localized UDP-GlcA transporter, we screened Arabidopsis thaliana mutants in genes coding for putative nucleotide sugar transporters for altered seed mucilage, a structure rich in the GalA-containing polysaccharide rhamnogalacturonan I. As a result, we identified UUAT1, which encodes a Golgi-localized protein that transports UDP-GlcA and UDP-GalA in vitro. The seed coat of uuat1 mutants had less GalA, rhamnose, and xylose in the soluble mucilage, and the distal cell walls had decreased arabinan content. Cell walls of other organs and cells had lower arabinose levels in roots and pollen tubes, but no differences were observed in GalA or xylose contents. Furthermore, the GlcA content of glucuronoxylan in the stem was not affected in the mutant. Interestingly, the degree of homogalacturonan methylation increased in uuat1 These results suggest that this UDP-GlcA transporter plays a key role defining the seed mucilage sugar composition and that its absence produces pleiotropic effects in this component of the plant extracellular matrix.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/metabolism , Golgi Apparatus/metabolism , Nucleotide Transport Proteins/metabolism , Polysaccharides/metabolism , Seeds/metabolism , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Cell Wall/genetics , Cell Wall/metabolism , Gene Expression Regulation, Plant , Immunoblotting , Microscopy, Confocal , Mutation , Nucleotide Transport Proteins/genetics , Pectins/metabolism , Plants, Genetically Modified , Seeds/genetics , Uridine Diphosphate Sugars/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL