Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 110.987
Filter
1.
Food Res Int ; 188: 114326, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38823825

ABSTRACT

Tormentilla erecta (L.) Raeusch is a widespread plant in Europe and Western Asia. Its rhizomes (Tormentilae rhizoma) are the main ingredient of herbal alcoholic beverages and can be used as a natural preservative in beer production. Apart from its unique taste qualities, therapeutic properties in gastrointestinal tract ailments are attributed to the tincture obtained from Tormentillae rhizoma. The presented research aimed to determine the mutual relationship between the components of Tormentillae tincture, present in popular alcoholic beverages, and intestinal epithelium (Caco-2 cell monolayers). A comprehensive qualitative and quantitative analysis of the tincture was performed, including the determination of condensed and hydrolyzable tannins as well as triterpenoids (UHPLC-DAD-MS/MS). Incubation of the tincture with Caco-2 monolayers has shown that only triterpenes pass through the monolayer, while condensed tannins are mainly bound to the monolayer surface. Ellagic acid derivatives were the only components of the Tormentillae tinctura being metabolized by cell monolayers to the compounds not previously described in the literature, which may be crucial in the treatment of intestinal diseases with inflammatory background.


Subject(s)
Intestinal Mucosa , Rhizome , Humans , Caco-2 Cells , Rhizome/chemistry , Intestinal Mucosa/metabolism , Triterpenes/metabolism , Plant Extracts/metabolism , Plant Extracts/pharmacology , Tandem Mass Spectrometry , Biological Transport , Chromatography, High Pressure Liquid , Alcoholic Beverages/analysis , Proanthocyanidins/metabolism , Hydrolyzable Tannins/metabolism , Ellagic Acid/metabolism
2.
Nat Commun ; 15(1): 4494, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38802368

ABSTRACT

Efflux pump antiporters confer drug resistance to bacteria by coupling proton import with the expulsion of antibiotics from the cytoplasm. Despite efforts there remains a lack of understanding as to how acid/base chemistry drives drug efflux. Here, we uncover the proton-coupling mechanism of the Staphylococcus aureus efflux pump NorA by elucidating structures in various protonation states of two essential acidic residues using cryo-EM. Protonation of Glu222 and Asp307 within the C-terminal domain stabilized the inward-occluded conformation by forming hydrogen bonds between the acidic residues and a single helix within the N-terminal domain responsible for occluding the substrate binding pocket. Remarkably, deprotonation of both Glu222 and Asp307 is needed to release interdomain tethering interactions, leading to opening of the pocket for antibiotic entry. Hence, the two acidic residues serve as a "belt and suspenders" protection mechanism to prevent simultaneous binding of protons and drug that enforce NorA coupling stoichiometry and confer antibiotic resistance.


Subject(s)
Bacterial Proteins , Cryoelectron Microscopy , Multidrug Resistance-Associated Proteins , Protons , Staphylococcus aureus , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Staphylococcus aureus/metabolism , Multidrug Resistance-Associated Proteins/metabolism , Multidrug Resistance-Associated Proteins/chemistry , Multidrug Resistance-Associated Proteins/genetics , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/chemistry , Models, Molecular , Biological Transport , Binding Sites , Hydrogen Bonding , Protein Conformation
3.
Life Sci ; 349: 122722, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38754814

ABSTRACT

AIMS: Steroidogenic acute regulatory (StAR)-related lipid transfer domain-3 (STARD3) is a sterol-binding protein that facilitates cholesterol transport between cellular organelles. Cholesterol accumulation in podocytes directly contributes to the pathogenesis of albuminuria and renal injury under the condition of diabetic kidney disease (DKD). The aim of this study is to determine the role of STARD3 on the intracellular distribution of cholesterol within podocytes. METHODS: In vivo and in vitro models of diabetes were performed. The protein levels of STARD3, Niemann-Pick disease type C1 (NPC1), and Niemann-Pick disease type C2 (NPC2) were respectively detected by western blot analysis, immunohistochemistry, and immunofluorescence. Filipin staining was used to evaluate the subcellular localization of cholesterol in podocytes. Mitochondrial damage was evaluated using JC-1 (CBIC2) and ROS (reactive oxygen species) assays. KEY FINDINGS: Upregulation of STARD3 under diabetes and hyperglycemia increases cholesterol transport from the late endosomal/lysosomal (LE/LY) to mitochondria, leading to mitochondrial cholesterol accumulation and cell injury in podocytes. Conversely, downregulating STARD3 expression attenuated mitochondrial cholesterol accumulation, and improved mitochondrial homeostasis. SIGNIFICANCE: STARD3 may govern intracellular cholesterol transport in podocytes, subsequently leading to regulation of mitochondrial metabolism. Therefore, targeting STARD3 emerges as a potential therapeutic strategy to mitigate diabetes-induced mitochondrial cholesterol accumulation and associated injury in podocytes.


Subject(s)
Cholesterol , Diabetes Mellitus, Experimental , Diabetic Nephropathies , Mitochondria , Podocytes , Podocytes/metabolism , Podocytes/pathology , Animals , Cholesterol/metabolism , Mitochondria/metabolism , Mice , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/complications , Male , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Biological Transport , Mice, Inbred C57BL , Humans
4.
Nat Commun ; 15(1): 3985, 2024 May 11.
Article in English | MEDLINE | ID: mdl-38734677

ABSTRACT

Pentamidine and melarsoprol are primary drugs used to treat the lethal human sleeping sickness caused by the parasite Trypanosoma brucei. Cross-resistance to these two drugs has recently been linked to aquaglyceroporin 2 of the trypanosome (TbAQP2). TbAQP2 is the first member of the aquaporin family described as capable of drug transport; however, the underlying mechanism remains unclear. Here, we present cryo-electron microscopy structures of TbAQP2 bound to pentamidine or melarsoprol. Our structural studies, together with the molecular dynamic simulations, reveal the mechanisms shaping substrate specificity and drug permeation. Multiple amino acids in TbAQP2, near the extracellular entrance and inside the pore, create an expanded conducting tunnel, sterically and energetically allowing the permeation of pentamidine and melarsoprol. Our study elucidates the mechanism of drug transport by TbAQP2, providing valuable insights to inform the design of drugs against trypanosomiasis.


Subject(s)
Aquaglyceroporins , Cryoelectron Microscopy , Melarsoprol , Molecular Dynamics Simulation , Pentamidine , Trypanosoma brucei brucei , Trypanosoma brucei brucei/metabolism , Aquaglyceroporins/metabolism , Aquaglyceroporins/chemistry , Melarsoprol/metabolism , Melarsoprol/chemistry , Pentamidine/chemistry , Pentamidine/metabolism , Biological Transport , Trypanocidal Agents/chemistry , Trypanocidal Agents/metabolism , Trypanocidal Agents/pharmacology , Protozoan Proteins/metabolism , Protozoan Proteins/chemistry , Humans
6.
J Phys Chem B ; 128(20): 4911-4921, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38736363

ABSTRACT

To combat surging multidrug-resistant Gram-negative bacterial infections, better strategies to improve the efficacy of existing drugs are critical. Because the dual membrane cell envelope is the first line of defense for these bacteria, it is crucial to understand the permeation properties of the drugs through it. Our recent study shows that isosmotic conditions prevent drug permeation inside Gram-negative bacteria, Escherichia coli, while hypoosmotic stress enhances the process. Here, we unravel the reason behind such differential drug penetration. Specifically, we dissect the roles of electrostatic screening and low membrane permeability in the penetration failure of drugs under osmotically balanced conditions. We compare the transport of a quaternary ammonium compound malachite green in the presence of an electrolyte (NaCl) and a wide variety of commonly used organic osmolytes, e.g., sucrose, proline, glycerol, sorbitol, and urea. These osmolytes of different membrane permeability (i.e., nonpermeable sucrose and NaCl, freely permeable urea and glycerol, and partially permeable proline and sorbitol) clarify the role of osmotic stress in cell envelope permeability. The results showcase that under balanced osmotic conditions, drug molecules fail to penetrate inside E. coli cells because of low membrane permeabilities and not because of electrostatic screening imposed by the osmolytes. Contribution of the electrostatic interactions, however, cannot be completely overruled as at osmotically imbalanced conditions, drug transport across the bacterial subcellular compartments is found to be dependent on the osmolytes used.


Subject(s)
Cell Membrane Permeability , Escherichia coli , Osmotic Pressure , Static Electricity , Escherichia coli/drug effects , Escherichia coli/metabolism , Biological Transport , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism , Cell Membrane/metabolism , Cell Membrane/chemistry
7.
AAPS PharmSciTech ; 25(5): 113, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38750336

ABSTRACT

Transport of oral nanocarriers across the GI epithelium necessitates transport across hydrophilic mucus layer and the hydrophobic epithelium. Based on hydrophobic-hydrophilic balance, Curcumin-Lipomer (lipid-polymer hybrid nanoparticles) comprising hydrophobic stearic acid and hydrophilic Gantrez™ AN 119 (Gantrez) were developed, by a radical in-situ approach, to successfully traverse both barriers. A monophasic preconcentrate (Cur-Pre) comprising Cur (Curcumin), stearic acid, Gantrez and stabilizers, prepared by simple solution, was added to an aqueous phase to instantaneously generate Curcumin-Lipomer (Cur-Lipo) of nanosize and high entrapment efficiency (EE). Cur-Lipo size and EE was optimized by Box-Behnken Design. Cur-Lipomers of varying hydrophobic-hydrophilic property obtained by varying the stearic acid: Gantrez ratio exhibited size in the range 200-400 nm, EE > 95% and spherical morphology as seen in the TEM. A decrease in contact angle and in mucus interaction, evident with increase in Gantrez concentration, indicated an inverse corelation with hydrophilicity, while a linear corelation was observed for mucopenetration and hydrophilicity. Cur-SLN (solid lipid nanoparticles) which served as the hydrophobic reference revealed contact angle > 90°, maximum interaction with mucus and minimal mucopenetration. The ex-vivo permeation study through chicken ileum, revealed maximum permeation with Cur-Lipo1 and comparable and significantly lower permeation of Cur-Lipo1-D and Cur-SLN proposing the importance of balancing the hydrophobic-hydrophilic property of the nanoparticles. A 1.78-fold enhancement in flux of hydrophobic Cur-SLN, with no significant change in permeation of the hydrophilic Cur-Lipomers (p > 0.05) following stripping off the mucosal layer was observed. This reiterated the significance of hydrophobic-hydrophilic balance as a promising strategy to design nanoformulations with superior permeation across the GI barrier.


Subject(s)
Curcumin , Drug Carriers , Hydrophobic and Hydrophilic Interactions , Intestinal Mucosa , Nanoparticles , Stearic Acids , Nanoparticles/chemistry , Administration, Oral , Animals , Stearic Acids/chemistry , Curcumin/administration & dosage , Curcumin/pharmacokinetics , Curcumin/chemistry , Intestinal Mucosa/metabolism , Drug Carriers/chemistry , Particle Size , Lipids/chemistry , Polymers/chemistry , Biological Transport/physiology , Polyvinyls/chemistry
8.
Nat Commun ; 15(1): 4036, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38740750

ABSTRACT

Microbial Ni2+ homeostasis underpins the virulence of several clinical pathogens. Ni2+ is an essential cofactor in urease and [NiFe]-hydrogenases involved in colonization and persistence. Many microbes produce metallophores to sequester metals necessary for their metabolism and starve competing neighboring organisms. The fungal metallophore aspergillomarasmine A (AMA) shows narrow specificity for Zn2+, Ni2+, and Co2+. Here, we show that this specificity allows AMA to block the uptake of Ni2+ and attenuate bacterial Ni-dependent enzymes, offering a potential strategy for reducing virulence. Bacterial exposure to AMA perturbs H2 metabolism, ureolysis, struvite crystallization, and biofilm formation and shows efficacy in a Galleria mellonella animal infection model. The inhibition of Ni-dependent enzymes was aided by Zn2+, which complexes with AMA and competes with the native nickelophore for the uptake of Ni2+. Biochemical analyses demonstrated high-affinity binding of AMA-metal complexes to NikA, the periplasmic substrate-binding protein of the Ni2+ uptake system. Structural examination of NikA in complex with Ni-AMA revealed that the coordination geometry of Ni-AMA mimics the native ligand, Ni-(L-His)2, providing a structural basis for binding AMA-metal complexes. Structure-activity relationship studies of AMA identified regions of the molecule that improve NikA affinity and offer potential routes for further developing this compound as an anti-virulence agent.


Subject(s)
Bacterial Proteins , Nickel , Nickel/metabolism , Nickel/chemistry , Animals , Virulence/drug effects , Bacterial Proteins/metabolism , Biofilms/drug effects , Zinc/metabolism , Zinc/chemistry , Moths/microbiology , Urease/metabolism , Urease/antagonists & inhibitors , Biological Transport
9.
Nature ; 629(8012): 704-709, 2024 May.
Article in English | MEDLINE | ID: mdl-38693257

ABSTRACT

Choline is an essential nutrient that the human body needs in vast quantities for cell membrane synthesis, epigenetic modification and neurotransmission. The brain has a particularly high demand for choline, but how it enters the brain remains unknown1-3. The major facilitator superfamily transporter FLVCR1 (also known as MFSD7B or SLC49A1) was recently determined to be a choline transporter but is not highly expressed at the blood-brain barrier, whereas the related protein FLVCR2 (also known as MFSD7C or SLC49A2) is expressed in endothelial cells at the blood-brain barrier4-7. Previous studies have shown that mutations in human Flvcr2 cause cerebral vascular abnormalities, hydrocephalus and embryonic lethality, but the physiological role of FLVCR2 is unknown4,5. Here we demonstrate both in vivo and in vitro that FLVCR2 is a BBB choline transporter and is responsible for the majority of choline uptake into the brain. We also determine the structures of choline-bound FLVCR2 in both inward-facing and outward-facing states using cryo-electron microscopy. These results reveal how the brain obtains choline and provide molecular-level insights into how FLVCR2 binds choline in an aromatic cage and mediates its uptake. Our work could provide a novel framework for the targeted delivery of therapeutic agents into the brain.


Subject(s)
Blood-Brain Barrier , Brain , Choline , Cryoelectron Microscopy , Membrane Transport Proteins , Models, Molecular , Choline/metabolism , Animals , Humans , Brain/metabolism , Mice , Blood-Brain Barrier/metabolism , Membrane Transport Proteins/metabolism , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/genetics , Female , Male , Biological Transport
10.
Nature ; 629(8012): 710-716, 2024 May.
Article in English | MEDLINE | ID: mdl-38693265

ABSTRACT

Phosphatidylcholine and phosphatidylethanolamine, the two most abundant phospholipids in mammalian cells, are synthesized de novo by the Kennedy pathway from choline and ethanolamine, respectively1-6. Despite the essential roles of these lipids, the mechanisms that enable the cellular uptake of choline and ethanolamine remain unknown. Here we show that the protein encoded by FLVCR1, whose mutation leads to the neurodegenerative syndrome posterior column ataxia and retinitis pigmentosa7-9, transports extracellular choline and ethanolamine into cells for phosphorylation by downstream kinases to initiate the Kennedy pathway. Structures of FLVCR1 in the presence of choline and ethanolamine reveal that both metabolites bind to a common binding site comprising aromatic and polar residues. Despite binding to a common site, FLVCR1 interacts in different ways with the larger quaternary amine of choline in and with the primary amine of ethanolamine. Structure-guided mutagenesis identified residues that are crucial for the transport of ethanolamine, but dispensable for choline transport, enabling functional separation of the entry points into the two branches of the Kennedy pathway. Altogether, these studies reveal how FLVCR1 is a high-affinity metabolite transporter that serves as the common origin for phospholipid biosynthesis by two branches of the Kennedy pathway.


Subject(s)
Choline , Ethanolamine , Membrane Transport Proteins , Models, Molecular , Humans , Choline/metabolism , Binding Sites , Ethanolamine/metabolism , Ethanolamine/chemistry , Membrane Transport Proteins/metabolism , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/genetics , Phosphatidylethanolamines/metabolism , Biological Transport , Animals , Phosphatidylcholines/metabolism , Phosphatidylcholines/chemistry , Phosphorylation
12.
FASEB J ; 38(10): e23703, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38805156

ABSTRACT

Renal tubules are featured with copious mitochondria and robust transport activity. Mutations in mitochondrial genes cause congenital renal tubulopathies, and changes in transport activity affect mitochondrial morphology, suggesting mitochondrial function and transport activity are tightly coupled. Current methods of using bulk kidney tissues or cultured cells to study mitochondrial bioenergetics are limited. Here, we optimized an extracellular flux analysis (EFA) to study mitochondrial respiration and energy metabolism using microdissected mouse renal tubule segments. EFA detects mitochondrial respiration and glycolysis by measuring oxygen consumption and extracellular acidification rates, respectively. We show that both measurements positively correlate with sample sizes of a few centimeter-length renal tubules. The thick ascending limbs (TALs) and distal convoluted tubules (DCTs) critically utilize glucose/pyruvate as energy substrates, whereas proximal tubules (PTs) are significantly much less so. Acute inhibition of TALs' transport activity by ouabain treatment reduces basal and ATP-linked mitochondrial respiration. Chronic inhibition of transport activity by 2-week furosemide treatment or deletion of with-no-lysine kinase 4 (Wnk4) decreases maximal mitochondrial capacity. In addition, chronic inhibition downregulates mitochondrial DNA mass and mitochondrial length/density in TALs and DCTs. Conversely, gain-of-function Wnk4 mutation increases maximal mitochondrial capacity and mitochondrial length/density without increasing mitochondrial DNA mass. In conclusion, EFA is a sensitive and reliable method to investigate mitochondrial functions in isolated renal tubules. Transport activity tightly regulates mitochondrial bioenergetics and biogenesis to meet the energy demand in renal tubules. The system allows future investigation into whether and how mitochondria contribute to tubular remodeling adapted to changes in transport activity.


Subject(s)
Energy Metabolism , Kidney Tubules , Mitochondria , Animals , Mice , Mitochondria/metabolism , Kidney Tubules/metabolism , Male , Mice, Inbred C57BL , Oxygen Consumption , Organelle Biogenesis , Biological Transport , Glycolysis/physiology , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics
13.
Planta ; 260(1): 7, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38789841

ABSTRACT

MAIN CONCLUSION: The study thoroughly investigates nanosilver production, properties, and interactions, shedding light on its multifaceted applications. It underscores the importance of characterizing nanosilver for predicting its behavior in complex environments. Particularly, it highlights the agricultural and environmental ramifications of nanosilver uptake by plants. Nowadays, silver nanoparticles (AgNPs) are a very adaptable nanomaterial with many uses, particularly in antibacterial treatments and agricultural operations. Clarification of key elements of nanosilver, such as its synthesis and characterization procedures, antibacterial activity, and intricate interactions with plants, particularly those pertaining to uptake and translocation mechanisms, is the aim of this in-depth investigation. Nanosilver synthesis is a multifaceted process that includes a range of methodologies, including chemical, biological, and sustainable approaches that are also environmentally benign. This section provides a critical evaluation of these methods, considering their impacts on repeatability, scalability, and environmental impact. The physicochemical properties of nanosilver were determined by means of characterization procedures. This review highlights the significance of analytical approaches such as spectroscopy, microscopy, and other state-of the-art methods for fully characterizing nanosilver particles. Although grasp of these properties is necessary in order to predict the behavior and potential impacts of nanosilver in complex biological and environmental systems. The second half of this article delves into the intricate interactions that plants have with nanosilver, emphasizing the mechanisms of absorption and translocation. There are significant ramifications for agricultural and environmental problems from the uptake of nanosilver by plants and its subsequent passage through their tissues. In summary, by summarizing the state-of-the-art information in this field, this study offers a comprehensive overview of the production, characterization, antibacterial capabilities, and interactions of nanosilver with plants. This paper contributes to the ongoing conversation in nanotechnology.


Subject(s)
Metal Nanoparticles , Plants , Silver , Silver/pharmacology , Silver/chemistry , Metal Nanoparticles/chemistry , Plants/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Infective Agents/metabolism , Anti-Infective Agents/pharmacology , Biological Transport
14.
Int J Mol Sci ; 25(10)2024 May 09.
Article in English | MEDLINE | ID: mdl-38791173

ABSTRACT

Astrocytes actively participate in neurotransmitter homeostasis by bidirectional communication with neuronal cells, a concept named the tripartite synapse, yet their role in dopamine (DA) homeostasis remains understudied. In the present study, we investigated the kinetic and molecular mechanisms of DA transport in cultured striatal astrocytes of adult rats. Kinetic uptake experiments were performed using radiolabeled [3H]-DA, whereas mRNA expression of the dopamine, norepinephrine, organic cation and plasma membrane monoamine transporters (DAT, NET, OCTs and PMAT) and DA receptors D1 and D2 was determined by qPCR. Additionally, astrocyte cultures were subjected to a 24 h treatment with the DA receptor agonist apomorphine, the DA receptor antagonist haloperidol and the DA precursor L-DOPA. [3H]-DA uptake exhibited temperature, concentration and sodium dependence, with potent inhibition by desipramine, nortriptyline and decynium-22, suggesting the involvement of multiple transporters. qPCR revealed prominent mRNA expression of the NET, the PMAT and OCT1, alongside lower levels of mRNA for OCT2, OCT3 and the DAT. Notably, apomorphine significantly altered NET, PMAT and D1 mRNA expression, while haloperidol and L-DOPA had a modest impact. Our findings demonstrate that striatal astrocytes aid in DA clearance by multiple transporters, which are influenced by dopaminergic drugs. Our study enhances the understanding of regional DA uptake, paving the way for targeted therapeutic interventions in dopaminergic disorders.


Subject(s)
Astrocytes , Corpus Striatum , Dopamine , Animals , Astrocytes/metabolism , Astrocytes/drug effects , Dopamine/metabolism , Rats , Corpus Striatum/metabolism , Corpus Striatum/drug effects , Haloperidol/pharmacology , Kinetics , Dopamine Plasma Membrane Transport Proteins/metabolism , Dopamine Plasma Membrane Transport Proteins/genetics , Apomorphine/pharmacology , Cells, Cultured , Male , Receptors, Dopamine D1/metabolism , Biological Transport/drug effects , Levodopa/pharmacology
15.
Int J Mol Sci ; 25(10)2024 May 17.
Article in English | MEDLINE | ID: mdl-38791523

ABSTRACT

Glucose transporters GLUT1 belong to the major facilitator superfamily and are essential to human glucose uptake. The overexpression of GLUT1 in tumor cells designates it as a pivotal target for glycoconjugate anticancer drugs. However, the interaction mechanism of glycoconjugate drugs with GLUT1 remains largely unknown. Here, we employed all-atom molecular dynamics simulations, coupled to steered and umbrella sampling techniques, to examine the thermodynamics governing the transport of glucose and two glycoconjugate drugs (i.e., 6-D-glucose-conjugated methane sulfonate and 6-D-glucose chlorambucil) by GLUT1. We characterized the specific interactions between GLUT1 and substrates at different transport stages, including substrate recognition, transport, and releasing, and identified the key residues involved in these procedures. Importantly, our results described, for the first time, the free energy profiles of GLUT1-transporting glycoconjugate drugs, and demonstrated that H160 and W388 served as important gates to regulate their transport via GLUT1. These findings provide novel atomic-scale insights for understanding the transport mechanism of GLUT1, facilitating the discovery and rational design of GLUT1-targeted anticancer drugs.


Subject(s)
Glucose Transporter Type 1 , Glycoconjugates , Molecular Dynamics Simulation , Glucose Transporter Type 1/metabolism , Glucose Transporter Type 1/chemistry , Glycoconjugates/metabolism , Glycoconjugates/chemistry , Humans , Glucose/metabolism , Biological Transport , Thermodynamics
16.
Int J Mol Sci ; 25(10)2024 May 18.
Article in English | MEDLINE | ID: mdl-38791559

ABSTRACT

This editorial summarizes the seven scientific papers published in the Special Issue "Overcoming Biological Barriers: Importance of Membrane Transporters in Homeostasis, Disease, and Disease Treatment 2 [...].


Subject(s)
Homeostasis , Membrane Transport Proteins , Humans , Membrane Transport Proteins/metabolism , Animals , Biological Transport
17.
Plant Physiol Biochem ; 211: 108712, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38733940

ABSTRACT

Phosphorus (P), a macronutrient, plays key roles in plant growth, development, and yield. Phosphate (Pi) transporters (PHTs) and PHOSPHATE1 (PHO1) are central to Pi acquisition and distribution. Potentially, PHO1 is also involved in signal transduction under low P. The current study was designed to identify and functionally characterize the PHO1 gene family in chickpea (CaPHO1s). Five CaPHO1 genes were identified through a comprehensive genome-wide search. Phylogenetically, CaPHO1s formed two clades, and protein sequence analyses confirmed the presence of conserved domains. CaPHO1s are expressed in different plant organs including root nodules and are induced by Pi-limiting conditions. Functional complementation of atpho1 mutant with three CaPHO1 members, CaPHO1, CaPHO1;like, and CaPHO1;H1, independently demonstrated their role in root to shoot Pi transport, and their redundant functions. To further validate this, we raised independent RNA-interference (RNAi) lines of CaPHO1, CaPHO1;like, and CaPHO1;H1 along with triple mutant line in chickpea. While single gene RNAi lines behaved just like WT, triple knock-down RNAi lines (capho1/like/h1) showed reduced shoot growth and shoot Pi content. Lastly, we showed that CaPHO1s are involved in root nodule development and Pi content. Our findings suggest that CaPHO1 members function redundantly in root to shoot Pi export and root nodule development in chickpea.


Subject(s)
Cicer , Plant Proteins , Plant Root Nodulation , Cicer/genetics , Cicer/metabolism , Cicer/growth & development , Plant Proteins/genetics , Plant Proteins/metabolism , Plant Root Nodulation/genetics , Gene Expression Regulation, Plant , Phosphates/metabolism , Phosphate Transport Proteins/metabolism , Phosphate Transport Proteins/genetics , Plant Roots/metabolism , Plant Roots/genetics , Plant Roots/growth & development , Phylogeny , Biological Transport/genetics , Multigene Family
18.
Elife ; 132024 May 23.
Article in English | MEDLINE | ID: mdl-38780415

ABSTRACT

Stramenopiles form a clade of diverse eukaryotic organisms, including multicellular algae, the fish and plant pathogenic oomycetes, such as the potato blight Phytophthora, and the human intestinal protozoan Blastocystis. In most eukaryotes, glycolysis is a strictly cytosolic metabolic pathway that converts glucose to pyruvate, resulting in the production of NADH and ATP (Adenosine triphosphate). In contrast, stramenopiles have a branched glycolysis in which the enzymes of the pay-off phase are located in both the cytosol and the mitochondrial matrix. Here, we identify a mitochondrial carrier in Blastocystis that can transport glycolytic intermediates, such as dihydroxyacetone phosphate and glyceraldehyde-3-phosphate, across the mitochondrial inner membrane, linking the cytosolic and mitochondrial branches of glycolysis. Comparative analyses with the phylogenetically related human mitochondrial oxoglutarate carrier (SLC25A11) and dicarboxylate carrier (SLC25A10) show that the glycolytic intermediate carrier has lost its ability to transport the canonical substrates malate and oxoglutarate. Blastocystis lacks several key components of oxidative phosphorylation required for the generation of mitochondrial ATP, such as complexes III and IV, ATP synthase, and ADP/ATP carriers. The presence of the glycolytic pay-off phase in the mitochondrial matrix generates ATP, which powers energy-requiring processes, such as macromolecular synthesis, as well as NADH, used by mitochondrial complex I to generate a proton motive force to drive the import of proteins and molecules. Given its unique substrate specificity and central role in carbon and energy metabolism, the carrier for glycolytic intermediates identified here represents a specific drug and pesticide target against stramenopile pathogens, which are of great economic importance.


All living organisms breakdown food molecules to generate energy for processes, such as growing, reproducing and movement. The series of chemical reactions that breakdown sugars into smaller molecules ­ known as glycolysis ­ is so important that it occurs in all life forms, from bacteria to humans. In higher organisms, such as fungi and animals, these reactions take place in the cytosol, the space surrounding the cell's various compartments. A transport protein then shuttles the end-product of glycolysis ­ pyruvate ­ into specialised compartments, known as the mitochondria, where most energy is produced. However, recently it was discovered that a group of living organisms, called the stramenopiles, have a branched glycolysis in which the enzymes involved in the second half of this process are located in both the cytosol and mitochondrial matrix. But it was not known how the intermediate molecules produced after the first half of glycolysis enter the mitochondria. To answer this question, Pyrihová et al. searched for transport protein(s) that could link the two halves of the glycolysis pathway. Computational analyses, comparing the genetic sequences of many transport proteins from several different species, revealed a new group found only in stramenopiles. Pyrihová et al. then used microscopy to visualise these new transport proteins ­ called GIC-1 and GIC-2 ­ in the parasite Blastocystis, which infects the human gut, and observed that they localise to mitochondria. Further biochemical experiments showed that GIC-1 and GIC-2 can physically bind these intermediate molecules, but only GIC-2 can transport them across membranes. Taken together, these observations suggest that GIC-2 links the two halves of glycolysis in Blastocystis. Further analyses could reveal corresponding transport proteins in other stramenopiles, many of which have devastating effects on agriculture, such as Phytophthora, which causes potato blight, or Saprolegnia, which causes skin infections in farmed salmon. Since human cells do not have equivalent transporters, they could be new drug targets not only for Blastocystis, but for these harmful pathogens as well.


Subject(s)
Blastocystis , Cytosol , Glycolysis , Mitochondria , Blastocystis/metabolism , Blastocystis/genetics , Humans , Mitochondria/metabolism , Cytosol/metabolism , Biological Transport , Protozoan Proteins/metabolism , Protozoan Proteins/genetics
19.
Cells ; 13(10)2024 May 07.
Article in English | MEDLINE | ID: mdl-38786013

ABSTRACT

Modern medicine has allowed for many advances in neurological and neurodegenerative disease (ND). However, the number of patients suffering from brain diseases is ever increasing and the treatment of brain diseases remains an issue, as drug efficacy is dramatically reduced due to the existence of the unique vascular structure, namely the blood-brain barrier (BBB). Several approaches to enhance drug delivery to the brain have been investigated but many have proven to be unsuccessful due to limited transport or damage induced in the BBB. Alternative approaches to enhance molecular delivery to the brain have been revealed in recent studies through the existence of molecular delivery pathways that regulate the passage of peripheral molecules. In this review, we present recent advancements of the basic research for these delivery pathways as well as examples of promising ventures to overcome the molecular hurdles that will enhance therapeutic interventions in the brain and potentially save the lives of millions of patients.


Subject(s)
Blood-Brain Barrier , Drug Delivery Systems , Blood-Brain Barrier/metabolism , Humans , Drug Delivery Systems/methods , Animals , Brain/metabolism , Biological Transport
20.
Planta ; 259(6): 141, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38695915

ABSTRACT

MAIN CONCLUSION: This review highlights the roles of phloem in the long-distance transport and accumulation of As in rice plants, facilitating the formulation of new strategies to reduce the grain As content. Rice is a staple diet for a significant proportion of the global population. As toxicity is a major issue affecting the rice productivity and quality worldwide. Phloem tissues of rice plants play vital roles in As speciation, long-distance transport, and unloading, thereby controlling the As accumulation in rice grains. Phloem transport accounts for a significant proportion of As transport to grains, ranging from 54 to 100% depending on the species [inorganic arsenate (As(V)), arsenite (As(III)), or organic dimethylarsinic acid (DMA(V)]. However, the specific mechanism of As transport through phloem leading to its accumulation in grains remains unknown. Therefore, understanding the molecular mechanism of phloem-mediated As transport is necessary to determine the roles of phloem in long-distance As transport and subsequently reduce the grain As content via biotechnological interventions. This review discusses the roles of phloem tissues in the long-distance transport and accumulation of As in rice grains. This review also highlights the biotechnological approaches using critical genetic factors involved in nodal accumulation, vacuolar sequestration, and cellular efflux of As in phloem- or phloem-associated tissues. Furthermore, the limitations of existing transgenic techniques are outlined to facilitate the formulation of novel strategies for the development of rice with reduced grain As content.


Subject(s)
Arsenic , Oryza , Phloem , Oryza/metabolism , Oryza/growth & development , Oryza/genetics , Phloem/metabolism , Arsenic/metabolism , Biological Transport , Edible Grain/metabolism , Edible Grain/growth & development
SELECTION OF CITATIONS
SEARCH DETAIL
...