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










Publication year range
1.
Int J Biol Macromol ; 260(Pt 2): 129562, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38246445

ABSTRACT

Dengue virus infection has significantly increased, with reported cases soaring from 505,430 in 2000 to 2,809,818 in 2022, emphasizing the need for effective treatments. Among the eleven structural and non-structural proteins of DENV, Non-structural protein 1 (NS1) has emerged as a promising target due to its diverse role in modulating the immune response, inducing vascular leakage, and facilitating viral replication and assembly. Monoclonal antibodies are the sole therapeutics to target NS1, but concerns about their cross-reactivity persist. Given these concerns, our study focuses on designing a novel Peptide Ligand Conjugate (PLC) as a potential alternative immunotherapeutic agent against NS1. This PLC aims to mediate the immune elimination of soluble NS1 and NS1-presenting DENV-infected host cells by pre-existing vaccine-induced immunity. By employing the High Throughput Virtual Screening (HTVS) method, QikProp analysis, and Molecular Dynamics studies, we identified three hits from Asinex Biodesigned Ligands out of 220,177 compounds that show strong binding affinity towards the monoclonal binding site of NS1 protein. After a rigorous analysis of physicochemical characteristics, antigenicity, allergenicity, and toxicity using various servers, we selected two peptides: the minimum epitopic region of the Diphtheria and Tetanus toxins as the peptide components of the PLCs. A non-cleavable, non-reactive oxime linker connected the ligand with the peptide through oxime and amide bonds. DPT vaccine is widely used in dengue-endemic countries, and it has been reported that antibodies titer against MER of Diphtheria toxin and Tetanus toxins persist lifelong in DPT-vaccinated people. Therefore, once the rationally designed PLCs bind to NS1 through the ligands, the peptide will induce an immune response against NS1 by triggering pre-existing DPT antibodies and activating memory cells. This orchestrated immune response will destroy soluble NS1 and NS1-expressing DENV-infected cells, thereby reducing the illness of severe dengue hemorrhagic fever and the DENV infection, respectively. Given the increasing demand for new therapeutics for DENV treatment, further investigation into this novel immune-therapeutic strategy may offer a new avenue for treating mild and severe dengue infections.


Subject(s)
Dengue Virus , Dengue , Severe Dengue , Humans , Dengue/therapy , Dengue/diagnosis , Ligands , Tetanus Toxin , Peptides , Immunotherapy , Oximes , Viral Nonstructural Proteins , Antibodies, Viral
2.
Eur J Med Chem ; 265: 116056, 2024 Feb 05.
Article in English | MEDLINE | ID: mdl-38171145

ABSTRACT

Different metabolic pathways like DNA replication, transcription, and recombination generate topological constrains in the genome. These topological constraints are resolved by essential molecular machines known as topoisomerases. To bring changes in DNA topology, the topoisomerases create a single or double-stranded nick in the template DNA, hold the nicked ends to let the tangled DNA pass through, and finally re-ligate the breaks. The DNA nicking and re-ligation activities as well as ATPase activities (when present) in topoisomerases are subjected to inhibition by several anticancer and antibacterial drugs, thus establishing these enzymes as successful targets in anticancer and antibacterial therapies. The anti-topoisomerase drugs interfere with the functioning of these enzymes and result in the accumulation of DNA tangles or lethal genomic breaks, thereby promoting host cell (or organism) death. The potential of topoisomerases in the human malarial parasite, Plasmodium falciparum in antimalarial drug development has received little attention so far. Interestingly, the parasite genome encodes orthologs of topoisomerases found in eukaryotes, prokaryotes, and archaea, thus, providing an enormous opportunity for investigating these enzymes for antimalarial therapeutics. This review focuses on the features of Plasmodium falciparum topoisomerases (PfTopos) with respect to their closer counterparts in other organisms. We will discuss overall advances and basic challenges with topoisomerase research in Plasmodium falciparum and our attempts to understand the interaction of PfTopos with classical and new-generation topoisomerase inhibitors using in silico molecular docking approach. The recent episodes of parasite resistance against artemisinin, the only effective antimalarial drug at present, further highlight the significance of investigating new drug targets including topoisomerases in antimalarial therapeutics.


Subject(s)
Antimalarials , Humans , Antimalarials/pharmacology , Plasmodium falciparum , Molecular Docking Simulation , Isomerases , DNA/metabolism , Anti-Bacterial Agents/pharmacology
3.
Mol Divers ; 2023 Dec 21.
Article in English | MEDLINE | ID: mdl-38127294

ABSTRACT

The continuous emergence of resistance against most frontline antimalarial drugs has led to countless deaths in malaria-endemic countries, counting 619,000 deaths in 2021, with mutation in drug targets being the sole cause. As mutation is correlated frequently with fitness cost, the likelihood of mutation emergence in multiple targets at a time is extremely low. Hence, multitargeting compounds may seem promising to address drug resistance issues with additional benefits like increased efficacy, improved safety profile, and the requirement of fewer pills compared to traditional single and combinational drugs. In this study, we attempted to use the High Throughput Virtual Screening approach to predict multitarget inhibitors against six chemically validated Plasmodium falciparum (Pf) kinases (PfPKG, PfMAP2, PfCDPK4, PfTMK, PfPK5, PfPI4K), resulting in 21 multitargeting hits. The molecular dynamic simulation of the top six complexes (Myricetin-MAP2, Quercetin-CDPK4, Myricetin-TMK, Quercetin-PKG, Salidroside-PK5, and Salidroside-PI4K) showed stable interactions. Moreover, hierarchical clustering reveals the structural divergence of the compounds from the existing antimalarials, indicating less chance of cross-resistance. Additionally, the top three hits were validated through parasite growth inhibition assays, with quercetin and myricetin exhibiting an IC50 value of 1.84 and 3.93 µM, respectively.

4.
J Biomol Struct Dyn ; : 1-25, 2023 Aug 01.
Article in English | MEDLINE | ID: mdl-37528665

ABSTRACT

Compared to the previous year, there has been an increase of nearly 2 million malaria cases in 2021. The emergence of drug-resistant strains of Plasmodium falciparum, the most deadly malaria parasite, has led to a decline in the effectiveness of existing antimalarial drugs. To address this problem, the present study aimed to identify natural compounds with the potential to inhibit multiple validated antimalarial drug targets. The natural compounds from the Natural Product Activity and Species Source (NPASS) database were screened against ten validated drug targets of Plasmodium falciparum using a structure-based molecular docking method. Twenty compounds, with targets ranging from three to five, were determined as the top hits. The molecular dynamics simulations of the top six complexes (NPC246162 in complex with PfAdSS, PfGDH, and PfNMT; NPC271270 in complex with PfCK, PfGDH, and PfdUTPase) confirmed their stable binding affinity in the dynamic environment. The Tanimoto coefficient and distance matrix score analysis show the structural divergence of all the hit compounds from known antimalarials, indicating minimum chances of cross-resistance. Thus, we propose further investigating these compounds in biochemical and parasite inhibition studies to reveal the real therapeutic potential. If found successful, these compounds may be a new avenue for future drug discovery efforts to combat existing antimalarial drug resistance.Communicated by Ramaswamy H. Sarma.

5.
Mol Divers ; 2023 Apr 20.
Article in English | MEDLINE | ID: mdl-37079243

ABSTRACT

According to the Center for Disease Control and Prevention, as of August 23, 94 countries had confirmed 42,954 Monkeypox Virus cases. As specific monkeypox drugs are not yet developed, the treatment depends on repurposed FDA-approved drugs. According to a recent study, the Monkeypox outbreak is caused by a strain with a unique mutation, raising the likelihood that the virus will develop resistance to current drugs by acquiring mutations in the targets of currently used drugs. The probability of multiple mutations in two or more drug targets at a time is always low than mutation in a single drug target. Therefore, we identified 15 triple-targeting FDA-approved drugs that can inhibit three viral targets, including topoisomerase1, p37, and thymidylate kinase, using high throughput virtual screening approach. Further, the molecular dynamics simulation analysis of the top hits such as Naldemedine and Saquinavir with their respective targets reveals the formation of stable conformational changes of the ligand-protein complexes inside the dynamic biological environment. We suggest further research on these triple-targeting molecules to develop an effective therapy for the currently spreading Monkeypox.

6.
Life Sci ; 311(Pt A): 121121, 2022 Dec 15.
Article in English | MEDLINE | ID: mdl-36283456

ABSTRACT

AIMS: Malaria deaths occur primarily due to complicated malaria associated with the sequestration of Plasmodium falciparum-infected erythrocyte (PfIE) in the capillary microvasculature. This study aims to design peptide ligand conjugates (PLCs) for treating complicated malaria using various in silico techniques. The PLC includes a natural ligand for the Plasmodium falciparum erythrocyte membrane protein 1 (PfEMP1): expressed explicitly on the surface of PfIE, and a highly immunogenic peptide derived from the commonly used peptide vaccines in malaria-endemic countries. The ligand is predicted to prevent the sequestration of PfIE, and the peptide is predicted to eliminate PfIE from circulation by the pre-existing vaccine-induced immunity. MAIN METHODS: The epitope identification from the vaccines and the analysis of physicochemical properties, antigenicity, allergenicity, and toxicity were performed using SVMTriP, ProtParam, VaxiJen, AllerTop, and ToxinPred servers, respectively. The high throughput virtual screening (HTVS) and drug-like properties analysis of natural compound ligands were carried out by Schrodinger-2021 software. The molecular dynamics simulations were performed through the WebGro server. KEY FINDINGS: HTVS revealed three bioactive natural ligands for PfEMP1 from (NPASS) database. Three super immunogenic peptides were identified from malaria-endemic countries' commonly used peptide vaccines. Finally, Nine PLCs were designed with different combinations of peptides and ligands with the suitable non-cleavable triazole linker. SIGNIFICANCE: Antimalarials have been losing efficacy in a time when malaria deaths in 2020 significantly increased than in 2019. In this scenario, further research on the designed PLCs may offer some innovative immune therapeutics for complicated malaria with minimum possibilities of drug resistance.


Subject(s)
Malaria Vaccines , Malaria, Falciparum , Malaria , Humans , Malaria, Falciparum/drug therapy , Malaria, Falciparum/prevention & control , Ligands , Malaria Vaccines/therapeutic use , Plasmodium falciparum , Malaria/drug therapy , Erythrocytes , Peptides/therapeutic use , Immunotherapy
7.
3 Biotech ; 12(9): 198, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35923684

ABSTRACT

Omicron, a variant of concern (VOC) of SARS-CoV-2, emerged in South Africa in November 2021. Omicron has been continuously acquiring a series of new mutations, especially in the spike (S) protein that led to high infectivity and transmissibility. Peptides targeting the receptor-binding domain (RBD) of the spike protein by which omicron and its variants attach to the host receptor, angiotensin-converting enzyme (ACE2) can block the viral infection at the first step. This study aims to identify antiviral peptides from the Antiviral peptide database (AVPdb) and HIV-inhibitory peptide database (HIPdb) against the RBD of omicron by using a molecular docking approach. The lead RBD binder peptides obtained through molecular docking were screened for allergenicity and physicochemical criteria (isoelectric point (pI) and net charge) required for peptide-based drugs. The binding affinity of the best five peptide inhibitors with the RBD of omicron was validated further by molecular dynamics (MD) simulation. Our result introduces five antiviral peptides, including AVP1056, AVP1059, AVP1225, AVP1801, and HIP755, that may effectively hinder omicron-host interactions. It is worth mentioning that all the three major sub-variants of omicron, BA.1 (B.1.1.529.1), BA.2 (B.1.1.529.2), and BA.3 (B.1.1.529.3), exhibits conserved ACE-2 interacting residues. Hence, the screened antiviral peptides with similar affinity can also interrupt the RBD-mediated invasion of different major sub-variants of omicron. Altogether, these peptides can be considered in the peptide-based therapeutics development for omicron treatment after further experimentation. Supplementary Information: The online version contains supplementary material available at 10.1007/s13205-022-03258-4.

8.
J Biomol Struct Dyn ; 40(19): 9543-9567, 2022.
Article in English | MEDLINE | ID: mdl-34062110

ABSTRACT

In view of many European countries and the USA leading to the second wave of COVID-19 pandemic, winter season, the evolution of new mutations in the spike protein, and no registered drugs and vaccines for COVID-19 treatment, the discovery of effective and novel therapeutic agents is urgently required. The degrees and frequencies of COVID-19 clinical complications are related to uncontrolled immune responses, secondary bacterial infections, diabetes, cardiovascular disease, hypertension, and chronic pulmonary diseases. It is essential to recognize that the drug repurposing strategy so far remains the only means to manage the disease burden of COVID-19. Despite some success of using single-target drugs in treating the disease, it is beyond suspicion that the virus will acquire drug resistance by acquiring mutations in the drug target. The possible synergistic inhibition of drug efficacy due to drug-drug interaction cannot be avoided while treating COVID-19 and allied clinical complications. Hence, to avoid the unintended development drug resistance and loss of efficacy due to drug-drug interaction, multi-target drugs can be promising tools for the most challenging disease. In the present work, we have carried out molecular docking studies of compounds from the FDA approved drug library, and the FDA approved and passed phase -1 drug libraries with ten therapeutic targets of COVID-19. Results showed that known drugs, including nine anti-inflammatory compounds, four antibiotics, six antidiabetic compounds, and one cardioprotective compound, could effectively inhibit multiple therapeutic targets of COVID-19. Further in-vitro, in vivo, and clinical studies will guide these drugs' proper allocation to treat COVID-19.Communicated by Ramaswamy H. Sarma.


Subject(s)
COVID-19 , Humans , Pandemics , Molecular Docking Simulation , COVID-19 Vaccines , Drug Repositioning/methods
9.
Infect Genet Evol ; 93: 104987, 2021 09.
Article in English | MEDLINE | ID: mdl-34216796

ABSTRACT

There has been a consistent rise in malaria cases in the last few years. The existing malaria control measures are challenged by insecticide resistance in the mosquito vector, drug résistance in parasite populations, and asymptomatic malaria (ASM) in healthy individuals. The absence of apparent malaria symptoms and the presence of low parasitemia makes ASM a hidden reservoir for malaria transmission and an impediment in malaria elimination efforts. This review focuses on ASM in malaria-endemic countries and the past and present research trends from those geographical locations. The harmful impacts of asymptomatic malaria on human health and its contribution to disease transmission are highlighted. We discuss certain crucial genetic changes in the parasite and host immune response necessary for maintaining low parasitemia leading to long-term parasite survival in the host. Since the chronic health effects and the potential roles for disease transmission of ASM remain mostly unknown to significant populations, we offer proposals for developing general awareness. We also suggest advanced technology-based diagnostic methods, and treatment strategies to eliminate ASM.


Subject(s)
Asymptomatic Infections , Disease Eradication , Disease Reservoirs/parasitology , Host-Parasite Interactions , Immunity , Malaria/parasitology , Humans , Malaria/prevention & control , Malaria/transmission , Parasitemia/parasitology , Risk Assessment
10.
Eur J Pharm Sci ; 151: 105375, 2020 Aug 01.
Article in English | MEDLINE | ID: mdl-32417398

ABSTRACT

The ongoing enigmatic COVID-19 outbreak, first reported from Wuhan, China, on last day of the year 2019, which has spread to 213 countries, territories/areas till 28th April 2020, threatens hundreds of thousands human souls. This devastating viral infection has stimulated the urgent development of viable vaccine against COVID-19 across the research institutes around the globe. The World Health Organization (WHO) has also confirmed that the recent pandemic is causing Public Health Emergency of International apprehension. Moreover, the earlier two pathogenic SARS-CoV and MERS-CoV and many others yet to be identified pose a universal menace. Here, in this piece of work, we have utilized an in silico structural biology and advanced immunoinformatic strategies to devise a multi-epitope subunit vaccine against ongoing COVID-19 infection. The engineered vaccine sequence is adjuvanted with ß-3 defensin and comprised of B-cell epitopes, HTL epitopes and CTL epitopes. This is very likely that the vaccine will be able to elicit the strong immune response. Further, specific binding of the engineered vaccine and immune cell receptor TLR3 was estimated by molecular interaction studies. Strong interaction in the binding groove as well as good docking scores affirmed the stringency of engineered vaccine. The interaction is stable with minimal deviation in root-mean square deviation and root-mean-square fluctuation was confirmed by the molecular dynamics simulation experiment. The immune-simulation by C-ImmSim server, which mimics the natural immune environment, yielded more potent immune response data of B-cells, Th cells, Tc cells and IgG for vaccine. The encouraging data obtained from the various in-silico works indicated this vaccine as an effective therapeutic against COVID-19.


Subject(s)
Betacoronavirus/immunology , Coronavirus Infections/immunology , Epitopes/immunology , High-Throughput Screening Assays/methods , Pneumonia, Viral/immunology , Viral Vaccines/immunology , Amino Acid Sequence , Antigens, Viral/chemistry , Antigens, Viral/genetics , Antigens, Viral/immunology , B-Lymphocytes/immunology , Binding Sites , Biomedical Engineering , COVID-19 , COVID-19 Vaccines , Computational Biology , Computer Simulation , Coronavirus Infections/prevention & control , Drug Design , Epitopes, B-Lymphocyte , Humans , Immunoglobulin G/immunology , Pandemics/prevention & control , Pneumonia, Viral/prevention & control , Proteome , SARS-CoV-2 , T-Lymphocytes/immunology , Toll-Like Receptor 3/drug effects
11.
Int J Biol Macromol ; 160: 1-17, 2020 Oct 01.
Article in English | MEDLINE | ID: mdl-32470577

ABSTRACT

The present-day world is severely suffering from the recently emerged SARS-CoV-2. The lack of prescribed drugs for the deadly virus has stressed the likely need to identify novel inhibitors to alleviate and stop the pandemic. In the present high throughput virtual screening study, we used in silico techniques like receptor-ligand docking, Molecular dynamic (MD), and ADME properties to screen natural compounds. It has been documented that many natural compounds display antiviral activities, including anti-SARS-CoV effect. The present study deals with compounds of Natural Product Activity and Species Source (NPASS) database with known biological activity that probably impedes the activity of six essential enzymes of the virus. Promising drug-like compounds were identified, demonstrating better docking score and binding energy for each druggable targets. After an extensive screening analysis, three novel multi-target natural compounds were predicted to subdue the activity of three/more major drug targets simultaneously. Concerning the utility of natural compounds in the formulation of many therapies, we propose these compounds as excellent lead candidates for the development of therapeutic drugs against SARS-CoV-2.


Subject(s)
Betacoronavirus/drug effects , Betacoronavirus/metabolism , Biological Products/metabolism , Biological Products/pharmacology , Molecular Targeted Therapy , Betacoronavirus/enzymology , Betacoronavirus/physiology , Biological Products/therapeutic use , Coronavirus Infections/drug therapy , Coronavirus Infections/metabolism , Drug Evaluation, Preclinical , High-Throughput Screening Assays , Humans , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Conformation , SARS-CoV-2 , Time Factors , User-Computer Interface , COVID-19 Drug Treatment
12.
Int J Biol Macromol ; 152: 535-545, 2020 Jun 01.
Article in English | MEDLINE | ID: mdl-32112848

ABSTRACT

Human immune cell toll-like receptors (TLRs) provide a novel chance for the development of the vaccine adjuvant engaging TLR signaling. A library of peptides was developed and peptides structure was generated through homology modeling and refinement. Further, these peptides were subjected to receptor-ligand interaction study against human immune cell TLRs using Schrödinger-suite software. Here, we identified the most potent ligands for each human immune cell receptor and identified it as a potent adjuvant. This work portrays the ability of binding of different known protein adjuvants with human TLRs 1--10. The significance of the study deals with the identification of adjuvant (ligand) for human TLRs individually which assist in the development of the optimal highly immunogenic vaccine.


Subject(s)
Immune System/immunology , Ligands , Peptides/chemistry , Toll-Like Receptors/immunology , Vaccines , Adjuvants, Immunologic , Algorithms , Humans , Molecular Docking Simulation , Protein Binding , Receptors, Immunologic , Signal Transduction , Software
13.
ACS Omega ; 4(8): 13069-13079, 2019 Aug 20.
Article in English | MEDLINE | ID: mdl-31460434

ABSTRACT

Nipah virus (NiV) is an emerging zoonotic pathogen, reported for the recent severe outbreaks of encephalitis and respiratory illness in humans and animals, respectively. Many antiviral drugs have been discovered to inhibit this pathogen, but none of them were that much efficient. To overcome the complications associated with this severe pathogenic virus, we have designed a multi-epitope subunit vaccine using computational immunology strategies. Identification of structural and nonstructural proteins of Nipah virus assisted in the vaccine designing. The selected proteins are known to be involved in the survival of the virus. The antigenic binders (B-cell, HTL, and CTL) from the selected proteins were prognosticated. These antigenic binders will be able to generate the humoral as well as cell-mediated immunity. All the epitopes were united with the help of suitable linkers and with an adjuvant at the N-terminal of the vaccine, for the enhancement of immunogenicity. The physiological characterization, along with antigenicity and allergenicity of the designed vaccine candidates, was estimated. The 3D structure prediction and its validation were performed. The validated vaccine model was then docked and simulated with the TLR-3 receptor to check the stability of the docked complex. This next-generation approach will provide a new vision for the development of a high immunogenic vaccine against the NiV.

14.
J Biol Chem ; 290(3): 1712-28, 2015 Jan 16.
Article in English | MEDLINE | ID: mdl-25425642

ABSTRACT

To survive and persist within its human host, the malaria parasite Plasmodium falciparum utilizes a battery of lineage-specific innovations to invade and multiply in human erythrocytes. With central roles in invasion and cytokinesis, the inner membrane complex, a Golgi-derived double membrane structure underlying the plasma membrane of the parasite, represents a unique and unifying structure characteristic to all organisms belonging to a large phylogenetic group called Alveolata. More than 30 structurally and phylogenetically distinct proteins are embedded in the IMC, where a portion of these proteins displays N-terminal acylation motifs. Although N-terminal myristoylation is catalyzed co-translationally within the cytoplasm of the parasite, palmitoylation takes place at membranes and is mediated by palmitoyl acyltransferases (PATs). Here, we identify a PAT (PfDHHC1) that is exclusively localized to the IMC. Systematic phylogenetic analysis of the alveolate PAT family reveals PfDHHC1 to be a member of a highly conserved, apicomplexan-specific clade of PATs. We show that during schizogony this enzyme has an identical distribution like two dual-acylated, IMC-localized proteins (PfISP1 and PfISP3). We used these proteins to probe into specific sequence requirements for IMC-specific membrane recruitment and their interaction with differentially localized PATs of the parasite.


Subject(s)
Acyltransferases/metabolism , Cell Membrane/metabolism , Membrane Proteins/metabolism , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Actins/chemistry , Biotin/chemistry , Catalysis , DNA Mutational Analysis , Green Fluorescent Proteins/metabolism , Humans , Malaria/parasitology , Phylogeny , Protein Structure, Tertiary , Protein Transport
15.
ACS Chem Biol ; 9(10): 2366-73, 2014 Oct 17.
Article in English | MEDLINE | ID: mdl-25089658

ABSTRACT

Malaria continues to be a major health problem globally. There is an urgent need to find new antimalarials. Acriflavine (ACF) is known as an antibacterial agent and more recently as an anticancer agent. Here, we report that ACF inhibits the growth of asexual stages of both chloroquine (CQ) sensitive and resistant strains of human malarial parasite, Plasmodium falciparum in vitro at nanomolar concentration. ACF clears the malaria infection in vivo from the bloodstreams of mice infected with Plasmodium berghei. Interestingly, ACF is accumulated only in the parasitized red blood cells (RBCs) and parasite specific transporters may have role in this specific drug accumulation. We further show that ACF impairs DNA replication foci formation in the parasites and affects the enzymatic activities of apicoplast specific Gyrase protein. We thus establish ACF as a potential antimalarial amidst the widespread incidences of drug resistant Plasmodium strains.


Subject(s)
Acriflavine/pharmacology , Antimalarials/pharmacology , Erythrocytes/drug effects , Malaria/drug therapy , Plasmodium berghei/drug effects , Plasmodium falciparum/drug effects , Animals , DNA Replication/drug effects , Erythrocytes/parasitology , Humans , In Vitro Techniques , Intercalating Agents/pharmacology , Malaria/parasitology , Mice , Topoisomerase II Inhibitors/pharmacology
16.
Front Biosci (Landmark Ed) ; 18(3): 982-92, 2013 06 01.
Article in English | MEDLINE | ID: mdl-23747861

ABSTRACT

Dinoflagellates, apicomplexans and ciliates are members of the monophyletic supergroup of Alveolata. The protists of this phylogenetic cluster have adapted to various ecological niches and lifestyles. Dinoflagellates and cilates can be found in any aquatic environment, whereas the phylum Apicomplexa solely comprises intracellular parasites. Despite their diversity all alveolates are united by the presence of membranous vesicles, so called alveoli, located beneath the plasma membrane. In addition to strengthening the cytoskeleton, these vesicles appear to possess taxon-specific functionality. In dinoflagellates and ciliates the alveoli predominantly play a structural role and can function as calcium stores. However, for the Apicomplexa, the alveolar vesicles -here jointly called the inner membrane complex (IMC)- are additionally involved in invasion of the host cell and are important scaffold elements during cytokinesis. Recent studies shed light on the architecture of the apicomplexan IMC and the number and diversity of its constituent proteins. This plethora of proteins and their varying evolutionary origin underlines the versatility of the IMC as a result of the adaption to a parasitic lifestyle.


Subject(s)
Apicomplexa/physiology , Intracellular Membranes/physiology , Protozoan Proteins/physiology
17.
Nucleic Acids Res ; 38(20): 7037-53, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20571080

ABSTRACT

Apicoplast, an essential organelle of human malaria parasite Plasmodium falciparum contains a ∼35 kb circular genome and is a possible target for therapy. Proteins required for the replication and maintenance of the apicoplast DNA are not clearly known. Here we report the presence of single-stranded DNA binding protein (SSB) in P falciparum. PfSSB is targeted to the apicoplast and it binds to apicoplast DNA. A strong ssDNA binding activity specific to SSB was also detected in P. falciparum lysate. Both the recombinant and endogenous proteins form tetramers and the homology modelling shows the presence of an oligosaccharide/oligonucleotide-binding fold responsible for ssDNA binding. Additionally, we used SSB as a tool to track the mechanism of delayed death phenomena shown by apicoplast targeted drugs ciprofloxacin and tetracycline. We find that the transport of PfSSB is severely affected during the second life cycle following drug treatment. Moreover, the translation of PfSSB protein and not the transcription of PfSSB seem to be down-regulated specifically during second life cycle although there is no considerable change in protein expression profile between drug-treated and untreated parasites. These results suggest dual control of translocation and translation of apicoplast targeted proteins behind the delayed death phenomena.


Subject(s)
DNA-Binding Proteins/metabolism , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Antiprotozoal Agents/pharmacology , Cell Nucleus/metabolism , Ciprofloxacin/pharmacology , DNA, Protozoan/metabolism , DNA, Single-Stranded/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Erythrocytes/parasitology , Genetic Complementation Test , Organelles/metabolism , Plasmodium falciparum/genetics , Plasmodium falciparum/growth & development , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Structural Homology, Protein
18.
Eukaryot Cell ; 8(11): 1759-69, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19700639

ABSTRACT

DNA gyrase is the only topoisomerase that can introduce negative supercoils into the DNA at the cost of ATP hydrolysis. Some but not all the steps of the topoisomerization reaction are understood clearly for both eukaryotic topoII and DNA gyrase. This study is an attempt to understand whether the B subunit of DNA gyrase binds to DNA directly, which may be central to the stimulation of its ATPase activity essential for gyrase function. We have dissected the Plasmodium falciparum gyrase B (PfGyrB) subunit to identify a 45-amino-acid region in the toprim domain that is responsible for its intrinsic DNA binding activity, DNA-stimulated ATPase activity, and DNA cleavage. We find that DNA has to enter through the ATP-operated clamp of PfGyrB to gain access to the DNA binding region. Furthermore, the rate of ATP hydrolysis of PfGyrB increases significantly with increasing DNA length, suggesting a possible communication between the ATPase domain and the DNA binding region that can account for its optimal ATPase activity. These results not only highlight the mechanism of GyrB action in the deadly human parasite P. falciparum but also provide meaningful insights into the current mechanistic model of DNA transport by gyrase during the topoisomerization reaction.


Subject(s)
DNA Gyrase/chemistry , DNA Gyrase/metabolism , Plasmodium falciparum/enzymology , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Amino Acid Sequence , DNA Gyrase/genetics , Molecular Sequence Data , Plasmodium falciparum/chemistry , Plasmodium falciparum/genetics , Protein Structure, Tertiary , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Protozoan Proteins/genetics , Sequence Alignment
19.
FEMS Microbiol Lett ; 282(2): 266-72, 2008 May.
Article in English | MEDLINE | ID: mdl-18397290

ABSTRACT

Plasmodium falciparum sirtuin, PfSir2, contains histone deacetylase (HDAC) activity that may be central to the regulation of virulence gene expression in the parasites. Although a few reports have been published recently regarding in vitro and in vivo function of PfSir2, expression of the endogenous protein (c. 30 kDa) has not been shown yet. Here we report the presence of PfSir2 in the parasite at the protein level by specific antibodies. HDAC activity of PfSir2 can be inhibited by nicotinamide, a product of sirtuin reaction. Surprisingly, we find that nicotinamide also delays parasite growth significantly in culture. These findings further our knowledge on PfSir2 and raise the possibility of using an inexpensive agent like nicotinamide as an antimalarial in combination with other antiparasitic drugs.


Subject(s)
Antimalarials , Histone Deacetylase Inhibitors , Niacinamide/pharmacology , Plasmodium falciparum/drug effects , Sirtuins/metabolism , Animals , Antibodies, Protozoan , DNA, Protozoan , Plasmodium falciparum/enzymology , Plasmodium falciparum/growth & development
20.
Nucleic Acids Res ; 35(9): 2861-74, 2007.
Article in English | MEDLINE | ID: mdl-17430964

ABSTRACT

Hexameric DnaB type replicative helicases are essential for DNA strand unwinding along with the direction of replication fork movement. These helicases in general contain an amino terminal domain and a carboxy terminal domain separated by a linker region. Due to the lack of crystal structure of a full-length DnaB like helicase, the domain structure and function of these types of helicases are not clear. We have reported recently that Helicobacter pylori DnaB helicase is a replicative helicase in vitro and it can bypass Escherichia coli DnaC activity in vivo. Using biochemical, biophysical and genetic complementation assays, here we show that though the N-terminal region of HpDnaB is required for conformational changes between C6 and C3 rotational symmetry, it is not essential for in vitro helicase activity and in vivo function of the protein. Instead, an extreme carboxy terminal region and an adjacent unique 34 amino acid insertion region were found to be essential for HpDnaB activity suggesting that these regions are important for proper folding and oligomerization of this protein. These results confer great potential in understanding the domain structures of DnaB type helicases and their related function.


Subject(s)
Bacterial Proteins/chemistry , DnaB Helicases/chemistry , Helicobacter pylori/enzymology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , DnaB Helicases/genetics , DnaB Helicases/metabolism , Genetic Complementation Test , Protein Structure, Tertiary , Sequence Deletion , Structural Homology, Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...