Dong Deng's research while affiliated with Sichuan University and other places

What is this page?


This page lists the scientific contributions of an author, who either does not have a ResearchGate profile, or has not yet added these contributions to their profile.

It was automatically created by ResearchGate to create a record of this author's body of work. We create such pages to advance our goal of creating and maintaining the most comprehensive scientific repository possible. In doing so, we process publicly available (personal) data relating to the author as a member of the scientific community.

If you're a ResearchGate member, you can follow this page to keep up with this author's work.

If you are this author, and you don't want us to display this page anymore, please let us know.

Publications (67)


Disordered Gln/Glu metabolism/conversion mediates the decrease in tubulin glutamylation levels in sperm flagella of Qrich2 KO mice. WT, wild-type; HOM or Qrich2 KO, Qrich2 knockout; NC, normal control; QRICH2-SH, QRICH2 knockdown; QRICH2-OE, QRICH2 overexpression. A Hierarchical cluster analysis of 20 amino acids in WT and Qrich2 KO mice. N = 6, the depth of red or blue represents the levels of the concentration. B Reduced concentration of Gln was observed in the testes of Qrich2 KO mice, while the concentration of Glu remained unchanged. N = 6, Student’s t test, ns, P > 0.05, ****P <  = 0.0001, error bars, s.e.m. C, D The increased Gln concentration and decreased Glu concentration were observed in the testes (C) and sperm (D) of Qrich2 KO mice. N = 3 or N = 4, Student’s t test, *P < 0.05, **P < 0.01, error bars, s.e.m. E, F Knockdown of QRICH2 increased the Gln concentration and decreased the Glu concentration (E); On the contrary, overexpression of QRICH2 decreased the Gln concentration and increased the Glu concentration (F). N = 3 or N = 4; Student’s t test, *P < 0.05, **P < 0.01, error bars, s.e.m. GQrich2 KO caused disordered expression of genes related to Gln/Glu metabolism/conversion in mouse testes and sperm (N = 3, Student’s t test, **P < 0.01, NS, P > 0.05, error bars, s.e.m. H Knockdown of QRICH2 in HeLa cells induced disordered expression of genes related to Gln/Glu metabolism/conversion, while overexpression of QRICH2 showed no significant effect on the expression of these genes. N = 3, Student’s t test, NS, P > 0.05, **P < 0.01, error bars, s.e.m. I, J Western blot (I) and immunofluorescence (J) showed increased expression of GLUL in the testes of Qrich2 KO mice. N = 3, Student’s t test, *P < 0.05, error bars, s.e.m. DAPI, blue; GLUL, red; scale bars, 125 µm
QRICH2 positively regulates the glutamylation levels of tubulin in flagella of mouse sperm. WT, wild-type; Qrich2 KO, Qrich2 knockout. (A, B) The western blot (A) and immunofluorescence (B) showed reduced expression of α-TUBULIN and decreased tubulin glutamylation levels in the testes of Qrich2 KO mice. The grayscale analysis of protein bands was shown in the bottom panel of A. N = 3, Student’s t test, *P < 0.05, error bars, s.e.m. DAPI, blue; GT335, red; scale bars, 125 µm. C, D The reduced expression of α-TUBULIN and decreased tubulin glutamylation levels in sperm of Qrich2 KO mice were observed by western blot (C) and immunofluorescence (D). The grayscale and fluorescence intensity analysis were shown in the bottom panel. N = 3, Student’s t test, *P < 0.05, **P < 0.01, error bars, s.e.m. DAPI, blue; GT335, green; α-TUB, α-TUBULIN, red; scale bars, 10 µm
QRICH2 maintains the glutamylation levels and stability of tubulin through interaction with tubulin. A, B Co-localization of QRICH2 and α-TUBULIN was observed in spermatogenic cells with different developmental stages of humans (A) and mice (B). DAPI, blue; α-TUBULIN, green; QRICH2, red; scale bars, 5 µm (A), 10 µm (B); Sa-d and 9–16 represent spermatogenic cells of different developmental stages in humans and mice. C, D Co-IP (C) and Duolink PLA (D) experiments indicated the interaction between QRICH2 and α-TUBULIN. E Motifs identified in QRICH2 across several species. F Structural model of the QRICH2/α-TUBULIN complex. Two regions on QRICH2 (in magenta helix including His107, Ser110, Glu114, Gly117, Asp118, Glu120, Lys121, Ile124, Thr125, Asn128, Leu129, Asp132, Lys136, Asp139, Leu143, Tyr144, Gly146, Ile147, Leu150, Asp151, Lys154-157Arg and loop including His570, Asp572, Pro575-Arg584) and the corresponding binding sites on α-TUBULIN (in green) were highlighted in opaque New Cartoon representations and indicated by close-up views. The other parts of QRICH2/α-TUBULIN were represented in transparent orange and ice-blue
Sperm motility is decreased and the microtubule structure of the flagella is abnormal in mice under a Gln/Glu-free diet. WT, wild-type; -Gln/Glu, mice under a Gln/Glu-free diet. A The computer-aided sperm analysis (CASA) showed decreased sperm motility in mice with absent Gln/Glu in diets. N = 3, Student’s t test, *P < 0.05, **P < 0.01, error bars, s.e.m. B No significant abnormality of spermatogenesis was observed in seminiferous tubules of mice under a Gln/Glu-free diet. 1–12 represent the different developmental stages of the spermatogenic epithelium. Scale bars, 125 µm. C Normal quantities of sperm were observed in the epididymis of mice under a Gln/Glu-free diet. Scale bars, 25 µm. D–G SEM showed morphologically normal head and developed flagella in the sperm of mice under a Gln/Glu-free diet. However, numerous sperm with bent or broken flagella were observed (D). Scale bars, 10 µm. TEM showed disorderly arranged microtubule structure in flagella of mice under a Gln/Glu-free diet (E). Scale bars, 300 nm. The proportion of sperm with abnormal flagella significantly increased in mice under a Gln/Glu-free diet (F, G). N = 3, Student’s t test, **P < 0.01, error bars, s.e.m
Glutamylation levels and expression of tubulin are decreased in sperm flagella of the mice with absent Gln/Glu in diets. WT, wild-type; -Gln/Glu, mice under a Gln/Glu-free diet; NC, normal control; -Gln, absence of Gln in cell culture medium. A, B Absence of Gln/Glu in the diets reduced the tubulin glutamylation levels and the α-TUBULIN expression in testes (A) and sperm flagella (B). The grayscale analysis of the protein bands was shown in the bottom panel. N = 3, Student’s t test, *P < 0.05, **P < 0.01, error bars, s.e.m. C Absence of Gln/Glu in the culture medium reduced the tubulin glutamylation levels. N = 3, Student’s t test, *P < 0.05, NS, P > 0.05, error bars, s.e.m. D Absence of Gln in the culture medium changed the distribution of α-TUBULIN in HeLa cells. DAPI, blue; GFP-α-TUBULIN, green; scale bars, 5 µm

+5

Metabolic profiling identifies Qrich2 as a novel glutamine sensor that regulates microtubule glutamylation and mitochondrial function in mouse sperm
  • Article
  • Full-text available

April 2024

·

23 Reads

Cellular and Molecular Life Sciences

Guohui Zhang

·

·

Haoxuan Yang

·

[...]

·

Wenming Xu

In our prior investigation, we discerned loss-of-function variants within the gene encoding glutamine-rich protein 2 (QRICH2) in two consanguineous families, leading to various morphological abnormalities in sperm flagella and male infertility. The Qrich2 knockout (KO) in mice also exhibits multiple morphological abnormalities of the flagella (MMAF) phenotype with a significantly decreased sperm motility. However, how ORICH2 regulates the formation of sperm flagella remains unclear. Abnormal glutamylation levels of tubulin cause dysplastic microtubules and flagella, eventually resulting in the decline of sperm motility and male infertility. In the current study, by further analyzing the Qrich2 KO mouse sperm, we found a reduced glutamylation level and instability of tubulin in Qrich2 KO mouse sperm flagella. In addition, we found that the amino acid metabolism was dysregulated in both testes and sperm, leading to the accumulated glutamine (Gln) and reduced glutamate (Glu) concentrations, and disorderly expressed genes responsible for Gln/Glu metabolism. Interestingly, mice fed with diets devoid of Gln/Glu phenocopied the Qrich2 KO mice. Furthermore, we identified several mitochondrial marker proteins that could not be correctly localized in sperm flagella, which might be responsible for the reduced mitochondrial function contributing to the reduced sperm motility in Qrich2 KO mice. Our study reveals a crucial role of a normal Gln/Glu metabolism in maintaining the structural stability of the microtubules in sperm flagella by regulating the glutamylation levels of the tubulin and identifies Qrich2 as a possible novel Gln sensor that regulates microtubule glutamylation and mitochondrial function in mouse sperm.

Download
Share


Mechanism of activation of β-1,3-glucan synthase by Rho1

February 2024

·

126 Reads

The β-1,3 glucan synthase (GS) is essential for the biosynthesis of β-1,3 glucan, a well-conserved structural component of fungal cell wall. The GS holoenzyme is a multi-enzyme complex consisting of the glycosyltransferase FKS and the essential regulatory factor Rho1, a small GTPase. However, the precise mechanism by which Rho1 activates FKS1 activity in a GTP-dependent manner remains elusive. Here, we present two cryo-electron microscopy (cryo-EM) structures of FKS1 alone (resting state) and FKS1-Rho1 complex (activating state), respectively. Structural analysis reveals that FKS1 adopts a cellulase-like conformation, wherein two segments of the cytoplasmic domain tightly bound together to form a functional structural unit. Remarkably, we unveil that the interaction between Rho1 and FKS1 is enhanced in the presence of a nonhydrolyzable guanosine triphosphate analog (GTP-γ-S). Rho1 is positioned within a pocket between the cytoplasmic domain of FKS1 and the transmembrane helix spanning TM7-15, engaging with the highly conserved glycosyltransferase domain of FKS1 (GT domain). Comparative analysis between the unbound (resting state) and Rho1-bound structures of FKS1 reveals the extensive conformational changes within FKS1, specifically in the GT domain and TM7-15. These alterations suggest that Rho1's GTP/GDP cycling acts as a molecular pump, inducing a dynamic transition between the resting and activating states of FKS1. Notably, the activation of Rho1 triggering FKS1 conformation changes, an evolutionary conserved "finger helix" within the FKS1-Rho1 complex adopts an up-and-down movement, ultimately pushing the growing glucan chain into FKS1’s transmembrane channel, thereby facilitating β-1,3 glucan elongation. Collectively, our results provide a vivid ratchet and pawl model to describe the mechanism of fungal β-1,3-glucan biosynthesis.


Structural basis of the subcortical maternal complex and its implications in reproductive disorders

January 2024

·

83 Reads

·

1 Citation

Nature Structural & Molecular Biology

The subcortical maternal complex (SCMC) plays a crucial role in early embryonic development. Malfunction of SCMC leads to reproductive diseases in women. However, the molecular function and assembly basis for SCMC remain elusive. Here we reconstituted mouse SCMC and solved the structure at atomic resolution using single-particle cryo-electron microscopy. The core complex of SCMC was formed by MATER, TLE6 and FLOPED, and MATER embraced TLE6 and FLOPED via its NACHT and LRR domains. Two core complexes further dimerize through interactions between two LRR domains of MATERs in vitro. FILIA integrates into SCMC by interacting with the carboxyl-terminal region of FLOPED. Zygotes from mice with Floped C-terminus truncation showed delayed development and resembled the phenotype of zygotes from Filia knockout mice. More importantly, the assembly of mouse SCMC was affected by corresponding clinical variants associated with female reproductive diseases and corresponded with a prediction based on the mouse SCMC structure. Our study paves the way for further investigations on SCMC functions during mammalian preimplantation embryonic development and reveals underlying causes of female reproductive diseases related to SCMC mutations, providing a new strategy for the diagnosis of female reproductive disorders.


Characterization of PfENT1
a Nucleoside binding by PfENT1 as measured by ITC. A representative ITC experiment is presented. The binding affinity (Kd) is presented as the value of the mean ± SD (n = 3), n means independent experiment. N.D., not detectable. Supplementary Fig. 2 shows the original ITC data and analyses. b The inosine transport activity of PfENT1. Time course for uptake of inosine in the knock-in yeast strain. The yeast strain without PfENT1 was tested as a control. Three independent experiments are performed for each point. Data are presented as mean ± SD. c Nucleoside competition and compound inhibition of PfENT1. The transport of [³H]-inosine was examined in uptake assays at 30 min in the presence of the indicated nucleosides/nucleobase (5 mM) and inhibitor GSK4 (10 μM). The yeast strain without PfENT1 was tested as a negative control. The PfENT1 knock-in yeast strain in the absence of non-radioactive nucleosides and inhibitor was test as positive control. Three independent experiments are performed for each substrate or inhibitor. Data are presented as mean ± SD. Source data are provided as a Source Data file.
The overall structure of PfENT1
a Density map and structure of the PfENT1Y190A-Nb19 complex. The map and structure of PfENT1Y190A are colored cyan, and the map and structure of Nb19 are shown in gray. b Density map and structure of the inosine-bound PfENT1Y190A-Nb48 complex. The map and structure of PfENT1Y190A are colored green, and the map and overall structure of Nb48 are shown as light gray. The inosine molecule is shown in spheres. c The overall structure of PfENT1Y190A. The 11 TMs of PfENT1Y190A are represented as rainbow-colored cylinders and viewed at two angles.
Substrate binding site of PfENT1
a Density map of inosine in the cavity of PfENT1 (6 σ, blue mesh). The density is fitted to the inosine molecule (sticks). b Inosine binding site of PfENT1. Dashed lines denote the inosine coordination in PfENT1. c Inosine binding affinity of the PfENT1 mutant (S49A, W53A, Q135, D287A, R291A) measured by ITC. The binding affinity (Kd) is presented as the value of the mean ± SD (n = 3), n means independent experiment. N.D., not detectable. Supplementary Fig. 8 shows further ITC data and analyses. d Inosine uptake assay of PfENT1 mutants. Three independent experiments are performed for each construct. Data are presented as mean ± SD (n = 3). e Expression of the codon-optimized PfENT1-HA proteins in an fui1Δ S. cerevisiae strain were detected by western blot probed with anti-HA antibodies. Source data are provided as a Source Data file.
Alternating access cycle of ENTs
a Superposition of the apo PfENT1 and inosine-bound PfENT1 complex. The apo- and inosine-bound PfENT1 is shown as a cartoon with the colors of light gray and blue, respectively. Inosine is represented as sticks. b Superposition of the N and C domains of PfENT1 and hENT1. PfENT1 and hENT1 is shown as cartoon with the colors of blue and green, respectively. c Superposition of the PfENT1, and hENT1 overall structure. PfENT1 and hENT1 is shown as cartoon with the colors of blue and green, respectively. d Critical residues in the alternating access cycle. The key residues are shown as sticks, interaction between key residues is shown by dashed lines.
Inhibition mechanism of GSK4
a Density map and structure of PfENT1GFP. The map and structure of PfENT1 are colored blue. The map and structure of GSK4 are shown in yellow. b Superposition of the PfENT1-Apo, inosine-bound PfENT1 complex, and GSK4-bound PfENT1 complex. PfENT1-Apo, inosine-bound PfENT1 complex, and GSK4-bound PfENT1 complex are shown as cartoons with colors of gray, blue, and pink, respectively. c Density map of GSK4 in the cavity of PfENT1 (6 σ, blue mesh). The density is fitted to the GSK4 molecule (sticks). d GSK4 binding site of PfENT1. Dashed lines denote GSK4 coordination in PfENT1. e Inosine binding affinity of the PfENT1 mutant (W53A, Q135, F139, R291A) measured by ITC. The binding affinity (Kd) is presented as the value of the mean ± SD (n = 3), n means independent experiment. N.D., not detectable. Supplementary Fig. 13 shows the original ITC data and analyses. Source data are provided as a Source Data file. f Superposition of the PfENT1-Apo, inosine-bound PfENT1 complex, and GSK4-bound PfENT1 complex, PfENT1-Apo, inosine-bound PfENT1 complex, and GSK4-bound PfENT1 complex are shown as cartoons with colors of gray, blue, and pink, respectively. Key residues are shown as sticks. Inosine and GSK4 are shown as sticks with blue and pink colors, respectively.
Structural basis of the substrate recognition and inhibition mechanism of Plasmodium falciparum nucleoside transporter PfENT1

March 2023

·

126 Reads

·

5 Citations

Nature Communications

By lacking de novo purine biosynthesis enzymes, Plasmodium falciparum requires purine nucleoside uptake from host cells. The indispensable nucleoside transporter ENT1 of P. falciparum facilitates nucleoside uptake in the asexual blood stage. Specific inhibitors of PfENT1 prevent the proliferation of P. falciparum at submicromolar concentrations. However, the substrate recognition and inhibitory mechanism of PfENT1 are still elusive. Here, we report cryo-EM structures of PfENT1 in apo, inosine-bound, and inhibitor-bound states. Together with in vitro binding and uptake assays, we identify that inosine is the primary substrate of PfENT1 and that the inosine-binding site is located in the central cavity of PfENT1. The endofacial inhibitor GSK4 occupies the orthosteric site of PfENT1 and explores the allosteric site to block the conformational change of PfENT1. Furthermore, we propose a general “rocker switch” alternating access cycle for ENT transporters. Understanding the substrate recognition and inhibitory mechanisms of PfENT1 will greatly facilitate future efforts in the rational design of antimalarial drugs.


Structural aspects of the glucose and monocarboxylate transporters involved in the Warburg effect

September 2022

·

72 Reads

·

5 Citations

International Union of Biochemistry and Molecular Biology Life

Cancer cells shift their glucose catabolism from aerobic respiration to lactic fermentation even in the presence of oxygen, and this is known as the “Warburg effect”. To accommodate the high glucose demands and to avoid lactate accumulation, the expression levels of human glucose transporters (GLUTs) and human monocarboxylate transporters (MCTs) are elevated to maintain metabolic homeostasis. Therefore, inhibition of GLUTs and/or MCTs provides potential therapeutic strategies for cancer treatment. Here, we summarize recent advances in the structural characterization of GLUTs and MCTs, providing a comprehensive understanding of their transport and inhibition mechanisms to facilitate further development of anticancer therapies.



High-fidelity biosensing of dNTPs and nucleic acids by controllable subnanometer channel PaMscS

December 2021

·

100 Reads

·

7 Citations

Biosensors and Bioelectronics

Current tools for dNTP analysis mainly rely on expensive fluorescent labeling, mass spectrometry or electrochemistry. Single-molecule assay by protein nanopores with an internal diameter of ca. 1–3.6 nm provides a useful tool for dNTP sensing. However, the most commonly used protein nanopores require additional modifications to enable dNTP detection. In this study, the PaMscS channel (mechanosensitive channel of small conductance from Pseudomonas aeruginosa) embedded in the bilayer lipid membrane (BLM) of E. coli polar lipid extract was applied as a nanopore for single molecular sensing. Two mutants of PaMscS nanopores on the side portal region (PaMscS W130A and PaMscS K180R) were selected for direct dNTP or pyrophosphoric acid (PPi) detection without aptamer or protein modification. Notably, the PaMscS mutant pore can be adjusted by regulation of osmolarity differences, which is crucial for the optimal detection of specific molecules. In addition, we established a PaMscS-based diagnosis method for the rapid sensing of disease-associated nucleic acids by monitoring the consumption of dNTPs, with 86% specificity and 100% sensitivity among 22 clinical samples. This protein nanopore, without aptamer or modification, paves a new way for dNTPs, PPi direct sensing and nucleic acid detection with low cost but high versatility.



Fig 1. Overall structure of PfFNT. (A) Hexose-monocarboxylate transport system of the P. falciparum-infected erythrocyte. Glucose and lactate are represented by orange hexagons and yellow triangles, respectively. Protons are presented by the blue circles. The magenta pentagon represents the PfFNT protein. Other reported structures, including human glucose transporter GLUT1 (PDB code: 4PYP), human monocarboxylate transporter MCT1 (PDB code: 6LZ0), and P. falciparum hexose transporter PfHT1 (PDB code: 6M2L), are presented as surface representations. Inhibitors of PfFNT (MMV007839) and PfHT1 (C3361) are displayed as sphere models. (B) Overall structure of pentameric PfFNT. The central tunnel of the pentamer and substrate translocation path in the protomer are indicated by pink circle and ellipse in the top view, respectively. (C) Topology diagram of a PfFNT protomer. The N-terminal and carboxyl-terminal TM segments are colored blue and green, respectively. Soluble helices, including the EH, NTH, and CTH, are colored gray. The NTL and O loops are colored sandy brown and salmon, respectively. Thr106 and His230 are presented as side chain models. (D) Cartoon representation of a PfFNT protomer. Components of the protomer are labeled in C. CTH, carboxyl-terminal helix; EH, extracellular helix; GLUT1, glucose transporter 1; MCT1, monocarboxylate transporter 1; NTH, N-terminal helix; NTL, N-terminal loop; PDB, Protein Data Bank; PfFNT, P. falciparum formate-nitrite transporter; PfHT1, P. falciparum hexose transporter 1; PVM, parasitophorous vacuole membrane; TM, transmembrane. https://doi.org/10.1371/journal.pbio.3001386.g001
Fig 2. Inhibition of PfFNT by MMV007839. (A) Binding affinity between PfFNT and MMV007839. (B) Density map of PfFNT in complex with MMV007839. The extra density for MMV007839 is highlighted in pink. The 5 protomers of PfFNT are distinguished by different colors. (C) The ligand density fits with the vinylogous acid form of MMV007839. The density for MMV007839, shown as the pink mesh, is contoured at 7.5 σ. (D) Coordination between PfFNT and MMV007839. MMV007839 is represented by sphere model. The polar contact between PfFNT and MMV007839 is shown. Inhibitor binding residues are shown as sticks and colored yellow. (E) The hydrophobic interactions between PfFNT and MMV007839. The hydrophobic residues in the cavity are shown as sticks and colored yellow. (F) Binding affinity between PfFNT T106A and MMV007839. (G) Binding affinity between PfFNT H230A and MMV007839. (H) Binding affinity between PfFNT G107S and MMV007839. The binding assay was performed via ITC and repeated 3 times. A representative titration is presented. The binding affinity (Kd) and N are presented as the value of mean ± SD (S2 Table). The raw data can be found in S1 Data. ITC, isothermal titration calorimetry; PfFNT, P. falciparum formate-nitrite transporter.
Fig 3. Molecular docking of MMV007839 and BH267.meta in the wild-type PfFNT or G107S resistant mutant. (A) The docking model of MMV007839 in the wild-type PfFNT. (B) The docking model of MMV007839 in PfFNT G107S . (C) The docking model of BH267.meta in the wild-type PfFNT. (D) The docking model of MMV007839 in PfFNT G107S . (E) Summary of binding energy of docking results. The transparent stick model of MMV007839 represents the real location in the inhibitor-bound structure. The structure of PfFNT is presented as cartoon representation and Gly107 or S107 are presented as sticks. The docking compounds are represented as sticks and colored gray. PfFNT, P. falciparum formate-nitrite transporter; WT, wild type. https://doi.org/10.1371/journal.pbio.3001386.g003
Fig 4. Structural comparison between apo form and inhibitor-bound PfFNT. (A) The constrictive site of a PfFNT protomer. The surface potential models of PfFNTs are presented. The residues near the constrictive site are shown as sticks. (B) The channel passages of PfFNTs and ecFocA were calculated using HOLE. The region containing the constriction sites is overlapped by transparent gray rectangle. The raw data can be found in S3 Data. (C) The superposition of pentameric PfFNT in 2 states. The apo PfFNT and inhibitor-bound PfFNT are colored blue and pink, respectively. (D) A comparison of the binding pocket for MMV007839. The inhibitor molecule is shown as grey stick. The Phe94 and Ile98 are represented as sticks. The apo PfFNT and inhibitor-bound PfFNT are colored the same as panel C. ecFocA, E. coli FocA; PfFNT, P. falciparum formate-nitrite transporter. https://doi.org/10.1371/journal.pbio.3001386.g004
Fig 5. Intracellular region of PfFNT. (A) A unique intracellular region is presented in a side view of PfFNT. The overall structure of PfFNT is shown as a cylinder cartoon. The region containing the intracellular region overlaps with a transparent light blue rectangle. (B) Overlap of NTLs in a bottom view of PfFNT. The overall structure of PfFNT is shown as a surface, and the 5 protomers are distinguished by different colors. (C) The interactions between NTLs. Each protomer is colored the same as in patterns A and B. Hydrogen bonds are shown as the gray dashed lines. (D) Coordination between the cytosolic CTH and the rest of the PfFNT protomer. The structure of the protomer of PfFNT is shown as cartoon and fitted to the cryo-EM density. The residues involved in the interactions are shown as pink sticks. Hydrogen bonds are shown as gray dashed lines. cryo-EM, cryo-electron microscopy; CTH, carboxyl-terminal helix; NTL, N-terminal loop; PfFNT, P. falciparum formate-nitrite transporter. https://doi.org/10.1371/journal.pbio.3001386.g005
Structural characterization of the Plasmodium falciparum lactate transporter PfFNT alone and in complex with antimalarial compound MMV007839 reveals its inhibition mechanism

September 2021

·

361 Reads

·

13 Citations

PLOS Biology

PLOS Biology

Plasmodium falciparum , the deadliest causal agent of malaria, caused more than half of the 229 million malaria cases worldwide in 2019. The emergence and spreading of frontline drug-resistant Plasmodium strains are challenging to overcome in the battle against malaria and raise urgent demands for novel antimalarial agents. The P . falciparum formate–nitrite transporter (PfFNT) is a potential drug target due to its housekeeping role in lactate efflux during the intraerythrocytic stage. Targeting PfFNT, MMV007839 was identified as a lead compound that kills parasites at submicromolar concentrations. Here, we present 2 cryogenic-electron microscopy (cryo-EM) structures of PfFNT, one with the protein in its apo form and one with it in complex with MMV007839, both at 2.3 Å resolution. Benefiting from the high-resolution structures, our study provides the molecular basis for both the lactate transport of PfFNT and the inhibition mechanism of MMV007839, which facilitates further antimalarial drug design.


Citations (58)


... This suggests that the interactions of these proteins detected in this study could occur at a high incidence in oocytes and preimplantation embryos. A more recent study revealed that murine NLRP5, possessing more than 10 LRRs, forms a homodimer via its two of the multiple LRRs, and interacts with OOEP and TLE6 via its NACHT domain and/or limited LRRs [24]. We hypothesize that NLRP5 could also interact with UHRF1 in vivo where NLRP5 is a part of CTLs formed by the SCMC through a different region(s) of the NACHT domain and/or a different LRR(s), distinct from those involved in the homodimerization and interaction with OOEP and TLE6. ...

Reference:

The maternal protein NLRP5 stabilizes UHRF1 in the cytoplasm: implication for the pathogenesis of multilocus imprinting disturbance
Structural basis of the subcortical maternal complex and its implications in reproductive disorders

Nature Structural & Molecular Biology

... The experimental support for this structure confirms it as a useful tool for the characterization of the binding site(s) for the PfENT1 inhibitory compounds and may facilitate further medicinal chemistry efforts to improve the GSK compounds' potency and efficacy. While this manuscript was in preparation, a paper was published that reported high resolution cryo-electron microscopy structures of PfENT1 with inosine and with GSK-4 bound in the transport pathway [17]. They mutated to alanine five of the 16 residues mutated to cysteine in this work and functionally characterized the mutants by inosine binding isothermal calorimetry and transport. ...

Structural basis of the substrate recognition and inhibition mechanism of Plasmodium falciparum nucleoside transporter PfENT1

Nature Communications

... The MCT1-4 isoforms exhibit different affinities for respective substrates. Moreover, the direction of transport (i.e., influx or efflux) is determined by concentration gradients of protons and monocarboxylate across the membrane that depend on their metabolic state, the prevailing substrate, and the current microenvironment (Halestrap and Price, 1999;Jiang et al., 2022). In the present study, we observed a higher amount of MCT transporters in the presence of L-lactate. ...

Structural aspects of the glucose and monocarboxylate transporters involved in the Warburg effect

International Union of Biochemistry and Molecular Biology Life

... Channel proteins are embedded in the lipid bilayer of P. aeruginosa, which is a non-specific, water-soluble diffusion channel that spans the cell membrane. Among them, the outer membrane proteins OprC, OprD2 and OprE show strong pore activity [17]. Scoffield et al[18] and Wang et al [19] reported that OprD2 gene deletion is the main mechanism of P. aeruginosa resistance to imipenem. ...

High-fidelity biosensing of dNTPs and nucleic acids by controllable subnanometer channel PaMscS
  • Citing Article
  • December 2021

Biosensors and Bioelectronics

... Through the systematic chemical degradation of Halofuginol and by comparison with its unequivocal antimalarial activity, researchers link specific functional groups (which play a decisive role in the potency regarding its antimalarial activity). The knowledge obtained in this structural activity relationship analysis forms the basis for the constructive design of compounds with higher potency and improved selectivity for antimalarial agents [4,[20][21][22]. ...

Structural characterization of the Plasmodium falciparum lactate transporter PfFNT alone and in complex with antimalarial compound MMV007839 reveals its inhibition mechanism
PLOS Biology

PLOS Biology

... Transmembrane 120B (TMEM120B) localizes on chromosome 12q24.31 and is composed of six transmembrane domains and a coil-coil domain [36]. Its homolog TMEM120A, also known as NET29, is crucial during adipogenesis and is expressed in both white and brown adipose tissues [37]. ...

Cryo-EM structures of human TMEM120A and TMEM120B

Cell Discovery

... By conclusively showing that TMEM120A specifically interacts with CoASH, the work of Rong et al. suggests that this membrane protein might in fact work as a membrane-embedded enzyme. It is also notable that Rong et al., along with several other groups (Ke et al., 2021;Niu et al., 2021;Parpaite et al., 2021;Rong et al., 2021;Xue et al., 2021), could not reproduce the mechanosensitive currents reported previously. Moreover, there are several lines of evidence to suggest that TMEM120A has a greater role in lipid metabolism. ...

Reference:

Pain or gain?
Cryo-EM structures of human TMEM120A and TMEM120B
  • Citing Preprint
  • June 2021

... As a real-world test of paDSF and Aurora-concise performance, we then attempted to create paDSF assays for ten proteins from the SARS-CoV-2 virus. These proteins have many properties that typically complicate standard techniques, including disorder 24,25 , phase separation 26,27 , protein-protein interactions 28,29 and protein-nucleic-acid interactions 30 . They also play diverse roles in SARS-CoV-2 replication and pathogenesis, including entry, translation, proteolysis, RNA binding and genome packing (Fig. 2a) 30 . ...

Crystal structure of SARS-CoV-2 nsp10 bound to nsp14-ExoN domain reveals an exoribonuclease with both structural and functional integrity

Nucleic Acids Research

... The nupG gene from E. coli K-12 was originally cloned on plasmid pKY2592 ( In a preliminary investigation of substrate recognition by NupG following the crystal structure, the same study used ITC to measure the binding of uridine to purified NupG (in MES buffer, pH 6.0, with 0.02% DDM), with the wild-type protein producing a binding affinity (Kd value) of 199.67 ± 15.01 μM (Wang et al., 2021). Several single-site specific alanine mutants of NupG were constructed informed by superimposition of the NupG structure with those of LacY (Abramson et al., 2003) and the E. coli xylose transporter XylE (Sun et al., 2012), and by molecular docking of NupG with uridine ( Figure 1). ...

Molecular basis for substrate recognition by the bacterial nucleoside transporter NupG

Journal of Biological Chemistry

... Circulating testosterone levels gradually decrease with age, while prostate cancer incidence increases with age. The activity of SRD5A2, which converts testosterone to DHT, declines with disease progression (9,45). These facts indicate that testosterone is not the sole determinant of prostate cancer. ...

Crystal structure of steroid reductase SRD5A reveals conserved steroid reduction mechanism

Nature Communications