Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
J Innate Immun ; 16(1): 133-142, 2024.
Article in English | MEDLINE | ID: mdl-38325356

ABSTRACT

INTRODUCTION: Coronavirus disease 2019 caused by coronavirus-2 (SARS-CoV-2) has emerged as an aggressive viral pandemic. Health care providers confront a challenging task for rapid development of effective strategies to combat this and its long-term after effects. Virus entry into host cells involves interaction between receptor-binding domain (RBD) of spike (S) protein S1 subunit with angiotensin converting enzyme present on host cells. Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) is a moonlighting enzyme involved in cellular glycolytic energy metabolism and micronutrient homeostasis. It is deployed in various cellular compartments and the extra cellular milieu. Though it is known to moonlight as a component of mammalian innate immune defense machinery, till date its role in viral restriction remains unknown. METHOD: Recombinant S protein, the RBD, and human GAPDH protein were used for solid phase binding assays and biolayer interferometry. Pseudovirus particles expressing four different strain variants of S protein all harboring ZsGreen gene as marker of infection were used for flow cytometry-based infectivity assays. RESULTS: Pseudovirus entry into target cells in culture was significantly inhibited by addition of human GAPDH into the extracellular medium. Binding assays demonstrated that human GAPDH binds to S protein and RBD of SARS-CoV-2 with nanomolar affinity. CONCLUSIONS: Our investigations suggest that this interaction of GAPDH interferes in the viral docking with hACE2 receptors, thereby affecting viral ingress into mammalian cells.


Subject(s)
COVID-19 , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating) , Protein Binding , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Virus Internalization , Humans , Spike Glycoprotein, Coronavirus/metabolism , SARS-CoV-2/physiology , COVID-19/virology , HEK293 Cells , Betacoronavirus/physiology , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Pneumonia, Viral/virology , Pneumonia, Viral/immunology , Pandemics , Coronavirus Infections/virology , Angiotensin-Converting Enzyme 2/metabolism
2.
Immunol Res ; 2024 Feb 12.
Article in English | MEDLINE | ID: mdl-38347341

ABSTRACT

Diabetes mellitus (DM) is a major risk factor for tuberculosis (TB), though the underlying mechanisms linking DM and TB remain ambiguous. Macrophages are a key player in the innate immune response and their phagocytic ability is enhanced in response to microbial infections. Upon infection or inflammation, they also repel invading pathogens by generating; reactive oxygen species (ROS), reactive nitrogen species (RNS), pro-inflammatory cytokines (IL-1ß and IL-6), and anti-inflammatory cytokines (IL-10). However, the robustness of these innate defensive capabilities of macrophages when exposed to hyperglycemia remains unclear. In our current work, we explored the production of these host defense molecules in response to challenge with Mycobacterium tuberculosis (Mtb) infection and lipopolysaccharide (LPS) stimulation. Utilizing peritoneal macrophages from high-fat diet + streptozotocin induced diabetic mice and hyperglycemic THP-1-derived macrophages as model systems; we found that LPS stimulation and Mtb infection were ineffective in stimulating the production of ROS, RNS, and pro-inflammatory cytokines in cells exposed to hyperglycemia. On the contrary, an increase in production of anti-inflammatory cytokines was observed. To confirm the mechanism of decreased anti-bacterial activity of the diabetic macrophage, we explored activation status of these compromised macrophages and found decreased surface expression of activation (TLR-4) and differentiation markers (CD11b and CD11c). We postulate that this could be the cause for higher susceptibility for Mtb infection among diabetic individuals.

3.
Trends Biochem Sci ; 49(3): 195-198, 2024 03.
Article in English | MEDLINE | ID: mdl-38195289

ABSTRACT

Targeting translational factor proteins (TFPs) presents significant promise for the development of innovative antitubercular drugs. Previous insights from antibiotic binding mechanisms and recently solved 3D crystal structures of Mycobacterium tuberculosis (Mtb) elongation factor thermo unstable-GDP (EF-Tu-GDP), elongation factor thermo stable-EF-Tu (EF-Ts-EF-Tu), and elongation factor G-GDP (EF-G-GDP) have opened up new avenues for the design and development of potent antituberculosis (anti-TB) therapies.


Subject(s)
Antitubercular Agents , Peptide Elongation Factor Tu , Guanosine Diphosphate/chemistry , Guanosine Diphosphate/metabolism , Peptide Elongation Factor Tu/chemistry , Peptide Elongation Factor Tu/metabolism , Antitubercular Agents/pharmacology , Antitubercular Agents/therapeutic use , Peptide Elongation Factors/chemistry , Peptide Elongation Factors/metabolism , Proteins/metabolism
4.
Free Radic Biol Med ; 208: 186-193, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37553026

ABSTRACT

Coronavirus disease-19 (COVID-19) can induce severe inflammation of the lungs and respiratory system. Severe COVID-19 is frequently associated with hyper inflammation and hyper-ferritinemia. High iron levels are known to trigger pro-inflammatory effects. Cumulative iron loading negatively impacts on a patients innate immune effector functions and increases the risk for infection related complications. Prognosis of severe acute respiratory SARS-CoV-2 patients may be impacted by iron excess. Iron is an essential co-factor for numerous essential cellular enzymes and vital cellular operations. Viruses hijack cells in order to replicate, and efficient replication requires an iron-replete host. Utilizing iron loaded cells in culture we evaluated their susceptibility to infection by pseudovirus expressing the SARS-CoV-2 spike protein and resultant cellular inflammatory response. We observed that, high levels of iron enhanced host cell ACE2 receptor expression contributing to higher infectivity of pseudovirus. In vitro Cellular iron overload also synergistically enhanced the levels of; reactive oxygen species, reactive nitrogen species, pro-inflammatory cytokines (IL-1ß, IL-6, IL-8 & TNF-α) and chemokine (CXCL-1&CCL-4) production in response to inflammatory stimulation of cells with spike protein. These results were confirmed using an in vivo mouse model. In future, limiting iron levels may be a promising adjuvant strategy in treating viral infection.


Subject(s)
COVID-19 , Iron Overload , Humans , Animals , Mice , SARS-CoV-2 , Inflammation , Iron
5.
Biochimie ; 214(Pt B): 102-113, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37385399

ABSTRACT

Mycobacterium tuberculosis enolase is an essential glycolytic enzyme that catalyzes the conversion of 2, phosphoglycerate (PGA) to phosphoenol pyruvate (PEP). It is also a crucial link between glycolysis and the tricarboxylic acid (TCA) pathway. The depletion of PEP has recently been associated with the emergence of non-replicating drug resistant bacteria. Enolase is also known to exhibit multiple alternate functions, such as promoting tissue invasion via its role as a plasminogen (Plg) receptor. In addition, proteomic studies have identified the presence of enolase in the Mtb degradosome and in biofilms. However, the precise role in these processes has not been elaborated. The enzyme was recently identified as a target for 2-amino thiazoles - a novel class of anti-mycobacterials. In vitro assays and characterization of this enzyme were unsuccessful due to the inability to obtain functional recombinant protein. In the present study, we report the expression and characterization of enolase using Mtb H37Ra as a host strain. Our study demonstrates that the enzyme activity and alternate functions of this protein are significantly impacted by the choice of expression host (Mtb H37Ra or E. coli). Detailed analysis of the protein from each source revealed subtle differences in the post-translational modifications. Lastly, our study confirms the role of enolase in Mtb biofilm formation and describes the potential for inhibiting this process.


Subject(s)
Mycobacterium tuberculosis , Phosphopyruvate Hydratase/genetics , Phosphopyruvate Hydratase/metabolism , Escherichia coli/metabolism , Proteomics , Plasminogen/metabolism
6.
J Innate Immun ; 15(1): 581-598, 2023.
Article in English | MEDLINE | ID: mdl-37080180

ABSTRACT

Mycobacterium tuberculosis (M.tb), the major causative agent of tuberculosis, has evolved mechanisms to evade host defenses and persist within host cells. Host-directed therapies against infected cells are emerging as an effective option. Cationic host defense peptide LL-37 is known to internalize into cells and induce autophagy resulting in intracellular killing of M.tb. This peptide also regulates the immune system and interacts with the multifunctional protein glyceraldehyde-3-phosphate dehydrogenase (GAPDH) inside macrophages. Our investigations revealed that GAPDH moonlights as a mononuclear cell surface receptor that internalizes LL-37. We confirmed that the surface levels of purinergic receptor 7, the receptor previously reported for this peptide, remained unaltered on M.tb infected macrophages. Upon infection or cellular activation with IFNγ, surface recruited GAPDH bound to and internalized LL-37 into endocytic compartments via a lipid raft-dependent process. We also discovered a role for GAPDH in LL-37-mediated autophagy induction and clearance of intracellular pathogens. In infected macrophages wherein GAPDH had been knocked down, we observed an inhibition of LL-37-mediated autophagy which was rescued by GAPDH overexpression. This process was dependent on intracellular calcium and p38 MAPK pathways. Our findings reveal a previously unknown process by which macrophages internalize an antimicrobial peptide via cell surface GAPDH and suggest a moonlighting role of GAPDH in regulating cellular phenotypic responses of LL-37 resulting in reduction of M.tb burden.


Subject(s)
Mycobacterium tuberculosis , Tuberculosis , Humans , Macrophages , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Mycobacterium tuberculosis/physiology , Antimicrobial Cationic Peptides/metabolism
7.
Chempluschem ; 88(4): e202300125, 2023 04.
Article in English | MEDLINE | ID: mdl-36970973

ABSTRACT

Photo-oxidase nanozymes are emerging enzyme-mimicking materials that produce reactive oxygen species (ROS) upon light illumination and subsequently catalyze the oxidation of the substrate. Carbon dots are promising photo-oxidase nanozymes due to their biocompatibility and straightforward synthesis. Carbon dot-based photo-oxidase nanozymes become active for ROS generation under UV or blue light illumination. In this work, sulfur and nitrogen doped carbon dots (S,N-CDs) were synthesized by solvent-free, microwave assisted technique. We demonstrated that sulfur, nitrogen doping of carbon dots (band gap of 2.11 eV) has enabled photo-oxidation of 3,3,5,5'-tetramethylbenzidine (TMB) with extended visible light (up to 525 nm) excitation at pH 4. The photo-oxidase activities by S,N-CDs produce Michaelis-Menten constant (Km ) of 1.18 mM and the maximum initial velocity (Vmax ) as 4.66×10-8  Ms-1 , under 525 nm illumination. Furthermore, visible light illumination can also induce bactericidal activities with growth inhibition of Escherichia coli (E. coli). These results demonstrate that S,N-CDs can increase intracellular ROS in the presence of LED light illumination.


Subject(s)
Anti-Infective Agents , Oxidoreductases , Oxidoreductases/chemistry , Reactive Oxygen Species , Escherichia coli , Light , Carbon/chemistry , Nitrogen/chemistry , Sulfur/chemistry
8.
Cell Mol Life Sci ; 79(1): 62, 2022 Jan 09.
Article in English | MEDLINE | ID: mdl-35001155

ABSTRACT

Availability of iron is a key factor in the survival and multiplication of Mycobacterium tuberculosis (M.tb) within host macrophage phagosomes. Despite host cell iron regulatory machineries attempts to deny supply of this essential micronutrient, intraphagosomal M.tb continues to access extracellular iron. In the current study, we report that intracellular M.tb exploits mammalian secreted Glyceraldehyde 3-phosphate dehydrogenase (sGAPDH) for the delivery of host iron carrier proteins lactoferrin (Lf) and transferrin (Tf). Studying the trafficking of iron carriers in infected cells we observed that sGAPDH along with the iron carrier proteins are preferentially internalized into infected cells and trafficked to M.tb containing phagosomes where they are internalized by resident mycobacteria resulting in iron delivery. Collectively our findings provide a new mechanism of iron acquisition by M.tb involving the hijack of host sGAPDH. This may contribute to its successful pathogenesis and provide an option for targeted therapeutic intervention.


Subject(s)
Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Iron/metabolism , Lactoferrin/metabolism , Mycobacterium tuberculosis/metabolism , Transferrin/metabolism , Animals , Biological Transport/physiology , Cell Line, Tumor , Humans , L Cells , Mice , Mice, Inbred C57BL , Phagosomes/metabolism , THP-1 Cells , Tuberculosis/pathology
9.
Mol Neurobiol ; 58(11): 5790-5798, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34406601

ABSTRACT

Protein aggregate accumulation is a pathological hallmark of several neurodegenerative disorders. Autophagy is critical for clearance of aggregate-prone proteins. In this study, we identify a novel role of the multifunctional glycolytic enzyme glyceraldehyde-3-phosphate dehydrogenase (GAPDH) in clearance of intracellular protein aggregates. Previously, it has been reported that though clearance of wild-type huntingtin protein is mediated by chaperone-mediated autophagy (CMA), however, degradation of mutant huntingtin (mHtt with numerous poly Q repeats) remains impaired by this route as mutant Htt binds with high affinity to Hsc70 and LAMP-2A. This delays delivery of misfolded protein to lysosomes and results in accumulation of intracellular aggregates which are degraded only by macroautophagy. Earlier investigations also suggest that mHtt causes inactivation of mTOR signaling, causing upregulation of autophagy. GAPDH had earlier been reported to interact with mHtt resulting in cellular toxicity. Utilizing a cell culture model of mHtt aggregates coupled with modulation of GAPDH expression, we analyzed the formation of intracellular aggregates and correlated this with autophagy induction. We observed that GAPDH knockdown cells transfected with N-terminal mutant huntingtin (103 poly Q residues) aggregate-prone protein exhibit diminished autophagy. GAPDH was found to regulate autophagy via the mTOR pathway. Significantly more and larger-sized huntingtin protein aggregates were observed in GAPDH knockdown cells compared to empty vector-transfected control cells. This correlated with the observed decrease in autophagy. Overexpression of GAPDH had a protective effect on cells resulting in a decreased load of aggregates. Our results demonstrate that GAPDH assists in the clearance of protein aggregates by autophagy induction. These findings provide a new insight in understanding the mechanism of mutant huntingtin aggregate clearance. By studying the molecular mechanism of protein aggregate clearance via GAPDH, we hope to provide a new approach in targeting and understanding several neurodegenerative disorders.


Subject(s)
Autophagy/physiology , Glyceraldehyde-3-Phosphate Dehydrogenases/physiology , Huntingtin Protein/metabolism , Protein Aggregates , Cell Line, Tumor , Gene Knockdown Techniques , Glyceraldehyde-3-Phosphate Dehydrogenases/antagonists & inhibitors , Glyceraldehyde-3-Phosphate Dehydrogenases/genetics , HEK293 Cells , Humans , Huntingtin Protein/genetics , Neuroblastoma , Peptides/genetics , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Ras Homolog Enriched in Brain Protein/metabolism , TOR Serine-Threonine Kinases/metabolism
10.
Biochim Biophys Acta Mol Basis Dis ; 1867(10): 166202, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34144092

ABSTRACT

Onset of protein aggregation reflects failure of the cellular folding machinery to keep aggregation-prone protein from misfolding and accumulating into a non-degradable state. FRET based analysis and biochemical data reveal that cytosolic prion (cyPrP) and httQ-103 interact with the multifunctional protein glyceraldehyde-3-phosphate dehydrogenase (GAPDH) leading to few detectable aggregates in GAPDH-over expressing cells.The preventive effect of GAPDH suggests that this abundant and long-lived cytoplasmic protein has an active role in the shielding and maintenance, in soluble form of proteins as heterogeneous as huntingtin and cyPrP.


Subject(s)
Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Protein Aggregates/physiology , Animals , COS Cells , Cell Line , Cell Line, Tumor , Chlorocebus aethiops , HeLa Cells , Humans
11.
Cell Physiol Biochem ; 52(3): 517-531, 2019.
Article in English | MEDLINE | ID: mdl-30897319

ABSTRACT

BACKGROUND/AIMS: Hypoxia triggers a rapid increase in iron demand to meet the requirements of enhanced erythropoiesis. The mobilization of iron stores from macrophage to plasma as holo-transferrin (Tf) from where it is accessible to erythroid precursor cells impacts iron homeostasis. Despite the immediate need for enhanced iron uptake by bone marrow cells, numerous studies have shown that transferrin receptor levels do not rise until more than 24 hours after the onset of hypoxia, suggesting the existence of heretofore unknown rapid response cellular machinery for iron acquisition in the early stages of cellular hypoxia. METHODS: We performed flow cytometry to measure cell surface levels of TfR1, GAPDH, and Tf binding after hypoxia treatment. We utilized FRET analysis and co-immunoprecipitation methods to establish the interaction between Tf and GAPDH. RESULTS: In the current study, we demonstrated that hypoxia induces K562 cells to translocate the cytosolic moonlighting protein glyceraldehyde-3-phosphate dehydrogenase (GAPDH) onto cell surfaces and into the extracellular milieu to acquire transferrin-bound iron, even while levels of the classical transferrin receptor TfR1 (CD71) remain suppressed. GAPDH knockdown confirmed this protein's role in transferrin acquisition. Interestingly, macrophages did not show enhanced levels of extracellular GAPDH under hypoxia. CONCLUSION: Our results suggest the role of GAPDH-mediated Tf uptake as a rapid response mechanism by which cells acquire iron during the early stages of hypoxia. This is a tissue-specific phenomenon for the distinct requirements of cells that are consumers of iron versus cells that play a role in iron storage and recycling. This rapid deployment of an abundantly available multipurpose molecule allows hypoxic cells to internalize more Tf and maintain enhanced iron supplies in the early stages of hypoxia before specialized receptors can be synthesized and deployed to the cell membrane.


Subject(s)
Cell Hypoxia , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Iron/metabolism , Antigens, CD/genetics , Antigens, CD/metabolism , Cell Membrane/metabolism , Cytosol/metabolism , Glyceraldehyde-3-Phosphate Dehydrogenases/antagonists & inhibitors , Glyceraldehyde-3-Phosphate Dehydrogenases/genetics , Humans , K562 Cells , Macrophages/cytology , Macrophages/metabolism , Protein Binding , RNA Interference , RNA, Small Interfering/metabolism , Receptors, Transferrin/genetics , Receptors, Transferrin/metabolism , Transferrin/metabolism
12.
J Antimicrob Chemother ; 74(4): 912-920, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30689890

ABSTRACT

BACKGROUND: The emergence of drug-resistant bacteria is a major hurdle for effective treatment of infections caused by Mycobacterium tuberculosis and ESKAPE pathogens. In comparison with conventional drug discovery, drug repurposing offers an effective yet rapid approach to identifying novel antibiotics. METHODS: Ethyl bromopyruvate was evaluated for its ability to inhibit M. tuberculosis and ESKAPE pathogens using growth inhibition assays. The selectivity index of ethyl bromopyruvate was determined, followed by time-kill kinetics against M. tuberculosis and Staphylococcus aureus. We first tested its ability to synergize with approved drugs and then tested its ability to decimate bacterial biofilm. Intracellular killing of M. tuberculosis was determined and in vivo potential was determined in a neutropenic murine model of S. aureus infection. RESULTS: We identified ethyl bromopyruvate as an equipotent broad-spectrum antibacterial agent targeting drug-susceptible and -resistant M. tuberculosis and ESKAPE pathogens. Ethyl bromopyruvate exhibited concentration-dependent bactericidal activity. In M. tuberculosis, ethyl bromopyruvate inhibited GAPDH with a concomitant reduction in ATP levels and transferrin-mediated iron uptake. Apart from GAPDH, this compound inhibited pyruvate kinase, isocitrate lyase and malate synthase to varying extents. Ethyl bromopyruvate did not negatively interact with any drug and significantly reduced biofilm at a 64-fold lower concentration than vancomycin. When tested in an S. aureus neutropenic thigh infection model, ethyl bromopyruvate exhibited efficacy equal to that of vancomycin in reducing bacterial counts in thigh, and at 1/25th of the dosage. CONCLUSIONS: Ethyl bromopyruvate exhibits all the characteristics required to be positioned as a potential broad-spectrum antibacterial agent.


Subject(s)
Anti-Bacterial Agents/pharmacology , Enzyme Inhibitors/pharmacology , Microbial Viability/drug effects , Mycobacterium tuberculosis/drug effects , Pyruvates/pharmacology , Staphylococcus aureus/drug effects , Animals , Anti-Bacterial Agents/administration & dosage , Disease Models, Animal , Drug Repositioning , Enzyme Inhibitors/administration & dosage , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/antagonists & inhibitors , Mice, Inbred BALB C , Pyruvates/administration & dosage , Staphylococcal Infections/drug therapy , Transferrin/antagonists & inhibitors , Treatment Outcome
13.
FASEB J ; 33(4): 5626-5640, 2019 04.
Article in English | MEDLINE | ID: mdl-30640524

ABSTRACT

During physiologic stresses, like micronutrient starvation, infection, and cancer, the cytosolic moonlighting protein glyceraldehyde-3-phosphate dehydrogenase (GAPDH) is trafficked to the plasma membrane (PM) and extracellular milieu (ECM). Our work demonstrates that GAPDH mobilized to the PM, and the ECM does not utilize the classic endoplasmic reticulum-Golgi route of secretion; instead, it is first selectively translocated into early and late endosomes from the cytosol via microautophagy. GAPDH recruited to this common entry point is subsequently delivered into multivesicular bodies, leading to its membrane trafficking through secretion via exosomes and secretory lysosomes. We present evidence that both pathways of GAPDH membrane trafficking are up-regulated upon iron starvation, potentially by mobilization of intracellular calcium. These pathways also play a role in clearance of misfolded intracellular polypeptide aggregates. Our findings suggest that cells build in redundancy for vital cellular pathways to maintain micronutrient homeostasis and prevent buildup of toxic intracellular misfolded protein refuse.-Chauhan, A. S., Kumar, M., Chaudhary, S., Dhiman, A., Patidar, A., Jakhar, P., Jaswal, P., Sharma, K., Sheokand, N., Malhotra, H., Raje, C. I., Raje. M. Trafficking of a multifunctional protein by endosomal microautophagy: linking two independent unconventional secretory pathways.


Subject(s)
Endosomes/metabolism , Microautophagy/physiology , Protein Transport/physiology , Secretory Pathway/physiology , Animals , Autophagy/physiology , Cell Line , Cell Membrane/metabolism , Cell Movement/physiology , Cytosol/metabolism , Endoplasmic Reticulum/metabolism , Exosomes/metabolism , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Golgi Apparatus/metabolism , Lysosomes/metabolism , Mice , Multivesicular Bodies/metabolism , Up-Regulation/physiology
14.
FASEB J ; 31(6): 2638-2648, 2017 06.
Article in English | MEDLINE | ID: mdl-28298336

ABSTRACT

Prokaryotic pathogens establish infection in mammals by capturing the proteolytic enzyme plasminogen (Plg) onto their surface to digest host extracellular matrix (ECM). One of the bacterial surface Plg receptors is the multifunctional glycolytic enzyme glyceraldehyde-3-phosphate dehydrogenase (GAPDH). In a defensive response, the host mounts an inflammatory response, which involves infiltration of leukocytes to sites of inflammation. This requires macrophage exit from the blood and migration across basement membranes, a phenomenon dependent on proteolytic remodeling of the ECM utilizing Plg. The ability of Plg to facilitate inflammatory cell recruitment critically depends on receptors on the surface of phagocyte cells. Utilizing a combination of biochemical, cellular, knockdown, and in vivo approaches, we demonstrated that upon inflammation, macrophages recruit GAPDH onto their surface to carry out the same task of capturing Plg to digest ECM to aid rapid phagocyte migration and combat the invading pathogens. We propose that GAPDH is an ancient, evolutionarily conserved receptor that plays a key role in the Plg-dependent regulation of macrophage recruitment in the inflammatory response to microbial aggression, thus pitting prokaryotic GAPDH against mammalian GAPDH, with both involved in a conserved role of Plg activation on the surface of their respective cells, to conflicting ends.-Chauhan, A. S., Kumar, M., Chaudhary, S., Patidar, A., Dhiman, A., Sheokand, N., Malhotra, H., Raje, C. I., Raje, M. Moonlighting glycolytic protein glyceraldehyde-3-phosphate dehydrogenase (GAPDH): an evolutionarily conserved plasminogen receptor on mammalian cells.


Subject(s)
Evolution, Molecular , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/metabolism , Macrophages/metabolism , Plasminogen/metabolism , Animals , Cell Line , Cell Movement , Gene Expression Regulation, Enzymologic/physiology , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/genetics , Mice , Receptors, Cell Surface , Receptors, Urokinase Plasminogen Activator/genetics , Receptors, Urokinase Plasminogen Activator/metabolism , Up-Regulation
15.
Microb Cell Fact ; 15(1): 140, 2016 Aug 11.
Article in English | MEDLINE | ID: mdl-27514669

ABSTRACT

BACKGROUND: Obtaining sufficient quantities of recombinant M.tb proteins using traditional approaches is often unsuccessful. Several enzymes of the glycolytic cycle are known to be multifunctional, however relatively few enzymes from M.tb H37Rv have been characterized in the context of their enzymatic and pleiotropic roles. One of the primary reasons is the difficulty in obtaining sufficient amounts of functionally active protein. RESULTS: In the current study, using M.tb glyceraldehyde-3-phosphate dehydrogenase (GAPDH) we demonstrate that expression in E. coli or M. smegmatis results in insolubility and improper subcellular localization. In addition, expression of such conserved multisubunit proteins poses the problem of heteromerization with host homologues. Importantly the expression host dramatically affected the yield and functionality of GAPDH in terms of both enzymatic activity and moonlighting function (transferrin binding). The applicability of this system was further confirmed using two additional enzymes i.e. M.tb Pyruvate kinase and Enolase. CONCLUSIONS: Our studies establish that the attenuated strain M.tb H37Ra is a suitable host for the expression of highly hydrophobic, conserved, multimeric proteins of M.tb H37Rv. Significantly, this expression host overcomes the limitations of E. coli and M. smegmatis expression and yields recombinant protein that is qualitatively superior to that obtained by traditional methods. The current study highlights the fact that protein functionality (which is an an essential requirement for all in vitro assays and drug development) may be altered by the choice of expression host.


Subject(s)
Bacterial Proteins/genetics , Glyceraldehyde-3-Phosphate Dehydrogenases/genetics , Multienzyme Complexes/genetics , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/genetics , Protein Multimerization , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Escherichia coli/genetics , Glyceraldehyde-3-Phosphate Dehydrogenases/chemistry , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Multienzyme Complexes/metabolism , Mycobacterium smegmatis/genetics , Phosphopyruvate Hydratase/genetics , Pyruvate Kinase/genetics , Recombinant Proteins/metabolism , Solubility , Transferrin/metabolism
16.
Protein Expr Purif ; 127: 22-27, 2016 11.
Article in English | MEDLINE | ID: mdl-27389468

ABSTRACT

Glyceraldehyde-3-phosphate dehydrogenase [GAPDH, NAD + oxidoreductase (phosphorylating) 1.2.1.12] catalyzes the conversion of glyceraldehyde-3-phosphate to 1,3-bisphosphoglycerate coupled with the reduction of NAD(+) to NADH. In addition to its role in glycolysis, this enzyme has numerous alternate functions, in both prokaryotes and eukaryotes. In plants, additional functions have been reported from multiple species including Pisum sativum. A recent study has identified that GAPDH may play an important role in seed ageing and programmed cell death. Despite this the existing purification protocols are almost 40 years old, and only partial characterization of the enzyme has been reported. In the current study, we report a modified method for purification of enzymatically active pea seed GAPDH along with the characterization of the enzyme. Using 2D gel electrophoresis our study also demonstrates that pea seeds contain four isoforms of NAD(+) dependent GAPDH.


Subject(s)
Glyceraldehyde-3-Phosphate Dehydrogenases/chemistry , Glyceraldehyde-3-Phosphate Dehydrogenases/isolation & purification , Pisum sativum/enzymology , Plant Proteins/chemistry , Plant Proteins/isolation & purification , Animals , Chickens , Rabbits
17.
J Cell Sci ; 129(4): 843-53, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26743084

ABSTRACT

Iron (Fe), a vital micronutrient for all organisms, must be managed judiciously because both deficiency or excess can trigger severe pathology. Although cellular Fe import is well understood, its export is thought to be limited to transmembrane extrusion through ferroportin (also known as Slc40a1), the only known mammalian Fe exporter. Utilizing primary cells and cell lines (including those with no discernible expression of ferroportin on their surface), we demonstrate that upon Fe loading, the multifunctional enzyme glyceraldehyde-3-phosphate dehydrogenase (GAPDH), which is recruited to the cell surface, 'treadmills' apotransferrin in and out of the cell. Kinetic analysis utilizing labeled ligand, GAPDH-knockdown cells, (55)Fe-labeled cells and pharmacological inhibitors of endocytosis confirmed GAPDH-dependent apotransferrin internalization as a prerequisite for cellular Fe export. These studies define an unusual rapid recycling process of retroendocytosis for cellular Fe extrusion, a process mirroring receptor mediated internalization that has never before been considered for maintenance of cellular cationic homeostasis. Modulation of this unusual pathway could provide insights for management of Fe overload disorders.


Subject(s)
Apoproteins/metabolism , Endocytosis , Iron/metabolism , Transferrin/metabolism , Animals , Cell Line , Mice , Protein Transport
18.
Biochem Soc Trans ; 42(6): 1796-801, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25399609

ABSTRACT

Iron is essential for the survival of both prokaryotic and eukaryotic organisms. It functions as a cofactor for several vital enzymes and iron deprivation is fatal to cells. However, at the same time, excess amounts of iron are also toxic to cells due to the formation of free radicals via the Fenton reaction. As a consequence of its double-edged behaviour, the uptake and regulation of iron involves an intricate balance of acquisition, trafficking, recycling and shuffling between various tissues and organs. This is accomplished by differential regulation of genes involving numerous proteins and enzymes. Several of the proteins identified in these processes, such as glyceraldehyde-3-phosphate dehydrogenase (GAPDH), aconitase and lactoferrin (Lf), possess multiple functions within the cell. Such proteins are referred to as moonlighting or multifunctional proteins, whereby proteins initially thought to possess a single well-established function have subsequently been discovered to exhibit alternative functions. In many cases, these multiple functions are conserved across species.


Subject(s)
Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Iron/metabolism , Animals , Bacteria/metabolism , Lactoferrin/metabolism , Receptors, Transferrin/metabolism
19.
Nat Commun ; 5: 4730, 2014 Aug 28.
Article in English | MEDLINE | ID: mdl-25163484

ABSTRACT

Mycobacterium tuberculosis (M.tb), which requires iron for survival, acquires this element by synthesizing iron-binding molecules known as siderophores and by recruiting a host iron-transport protein, transferrin, to the phagosome. The siderophores extract iron from transferrin and transport it into the bacterium. Here we describe an additional mechanism for iron acquisition, consisting of an M.tb protein that drives transport of human holo-transferrin into M.tb cells. The pathogenic strain M.tb H37Rv expresses several proteins that can bind human holo-transferrin. One of these proteins is the glycolytic enzyme glyceraldehyde-3-phosphate dehydrogenase (GAPDH, Rv1436), which is present on the surface of M.tb and its relative Mycobacterium smegmatis. Overexpression of GAPDH results in increased transferrin binding to M.tb cells and iron uptake. Human transferrin is internalized across the mycobacterial cell wall in a GAPDH-dependent manner within infected macrophages.


Subject(s)
Bacterial Proteins/metabolism , Host-Pathogen Interactions , Iron/metabolism , Mycobacterium tuberculosis/metabolism , Transferrin/metabolism , Amino Acid Sequence , Animals , Bacterial Proteins/genetics , Cell Wall/metabolism , Female , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/genetics , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/immunology , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/metabolism , Humans , Immunoprecipitation , Macrophages/metabolism , Macrophages/microbiology , Mice, Inbred BALB C , Molecular Sequence Data , Mycobacterium smegmatis/genetics , Mycobacterium smegmatis/metabolism , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/pathogenicity , Rabbits , Siderophores/metabolism , Transferrin/genetics
20.
Biochim Biophys Acta ; 1830(6): 3816-27, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23541988

ABSTRACT

BACKGROUND: The long held view is that mammalian cells obtain transferrin (Tf) bound iron utilizing specialized membrane anchored receptors. Here we report that, during increased iron demand, cells secrete the glycolytic enzyme glyceraldehyde-3-phosphate dehydrogenase (GAPDH) which enhances cellular uptake of Tf and iron. METHODS: These observations could be mimicked by utilizing purified GAPDH injected into mice as well as when supplemented in culture medium of model cell lines and primary cell types that play a key role in iron metabolism. Transferrin and iron delivery was evaluated by biochemical, biophysical and imaging based assays. RESULTS: This mode of iron uptake is a saturable, energy dependent pathway, utilizing raft as well as non-raft domains of the cell membrane and also involves the membrane protein CD87 (uPAR). Tf internalized by this mode is also catabolized. CONCLUSIONS: Our research demonstrates that, even in cell types that express the known surface receptor based mechanism for transferrin uptake, more transferrin is delivered by this route which represents a hidden dimension of iron homeostasis. GENERAL SIGNIFICANCE: Iron is an essential trace metal for practically all living organisms however its acquisition presents major challenges. The current paradigm is that living organisms have developed well orchestrated and evolved mechanisms involving iron carrier molecules and their specific receptors to regulate its absorption, transport, storage and mobilization. Our research uncovers a hidden and primitive pathway of bulk iron trafficking involving a secreted receptor that is a multifunctional glycolytic enzyme that has implications in pathological conditions such as infectious diseases and cancer.


Subject(s)
Autocrine Communication/physiology , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Iron/metabolism , Receptors, Transferrin/metabolism , Transferrin/metabolism , Animals , Biological Transport, Active/physiology , CHO Cells , Cricetinae , Cricetulus , Glyceraldehyde-3-Phosphate Dehydrogenases/genetics , HeLa Cells , Humans , Jurkat Cells , K562 Cells , Mice , Receptors, Transferrin/genetics , Receptors, Urokinase Plasminogen Activator/genetics , Receptors, Urokinase Plasminogen Activator/metabolism , Transferrin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...