Manisha M Dias’s research while affiliated with Monash University (Australia) and other places

What is this page?


This page lists works of an author who doesn't have a ResearchGate profile or hasn't added the works to their profile yet. It is automatically generated from public (personal) data to further our legitimate goal of comprehensive and accurate scientific recordkeeping. If you are this author and want this page removed, please let us know.

Publications (10)


High-Throughput Screening to Identify Inhibitors of Plasmodium falciparum Importin α
  • Article
  • Full-text available

April 2022

·

139 Reads

·

8 Citations

Cells

·

Manisha M. Dias

·

·

[...]

·

The global burden of malaria and toxoplasmosis has been limited by the use of efficacious anti-parasitic agents, however, emerging resistance in Plasmodium species and Toxoplasma gondii threatens disease control worldwide, implying that new agents/therapeutic targets are urgently needed. Nuclear localization signal (NLS)-dependent transport into the nucleus, mediated by members of the importin (IMP) superfamily of nuclear transporters, has shown potential as a target for intervention to limit viral infection. Here, we show for the first time that IMPα from P. falciparum and T. gondii have promise as targets for small molecule inhibitors. We use high-throughput screening to identify agents able to inhibit P. falciparum IMPα binding to a P. falciparum NLS, identifying a number of compounds that inhibit binding in the µM-nM range, through direct binding to P. falciparum IMPα, as shown in thermostability assays. Of these, BAY 11-7085 is shown to be a specific inhibitor of P. falciparum IMPα-NLS recognition. Importantly, a number of the inhibitors limited growth by both P. falciparum and T. gondii. The results strengthen the hypothesis that apicomplexan IMPα proteins have potential as therapeutic targets to aid in identifying novel agents for two important, yet neglected, parasitic diseases.

Download

Overlapping binding sites for importin β1 and Suppressor of Fused on Glioma-associated oncogene homolog 1 (Gli1) regulate its nuclear localisation.

May 2014

·

57 Reads

·

21 Citations

Biochemical Journal

A key factor in oncogenesis is the transport into the nucleus of oncogenic signalling molecules, such as Gli1 (Glioma-associated oncogene homolog 1), the central transcriptional activator in the Hedgehog signalling pathway. Little is known, however, how factors such as Gli are transported into the nucleus, and how this may be regulated by interaction with other cellular factors, such as the negative regulator Suppressor of Fused (SuFu). Here we show for the first time that nuclear entry of Gli1 is regulated by a unique mechanism through mutually exclusive binding by its nuclear import factor, importin (Imp) β1, and SuFu. Using quantitative live mammalian cell imaging, we show that nuclear accumulation of GFP-Gli1 fusion proteins, but not of a control protein, is specifically inhibited by coexpression of SuFu. Using a direct binding assay, we show that Impβ1 exhibits high nM affinity to Gli1, with specific knockdown of Impβ1 expression able to inhibit Gli1 nuclear accumulation, thus implicating Impβ1 as the nuclear transporter for Gli1 for the first time. SuFu also binds to Gli1 with high nM affinity, intriguingly being able to compete with Impβ1 for binding to Gli1, through the fact that the sites for SuFu and Impβ1 binding site overlap in the Gli1 N-terminus. The results indicate for the first time that the relative intracellular concentrations of SuFu and Imp β1 are likely to determine the localisation of Gli1, with implications for its action in cancer, as well as in developmental systems.


FIGURE 3. The RbBS causes cytoplasmic retention of T-ag in vivo. A, CLSM images showing the return of nuclear fluorescence after photobleaching (see " Experimental Procedures " ) in COS-7 cells expressing the indicated GFP-T-ag constructs. B, quantification of the recovery over time of nuclear fluorescence after photobleaching , shown as Fn/c. C, pooled data (mean, S.E., n 7) for the fractional recovery (% recovery of Fn/c relative to GFP-T-ag-(110 –135); left) and the initial rate (Fn/c s 1 ; right) into the nucleus for the indicated GFP-T-ag constructs. p values indicate significant differences.  
FIGURE 4. p110 Rb expression reduces nuclear import of T-ag constructs containing a functional RbBS. A, CLSM images are shown for the indicated GFP-T-ag proteins 14 h post-transfection in either SAOS-2 and/or SR-40 cell lines. B, results for image analysis, performed as described in the legend to Fig. 2, where n 38. p values are indicated for significant differences in Fn/c between the 2 cell lines or between constructs. C, CLSM images are shown for the indicated GFP-T-ag proteins 14-h post-transfection in either SAOS-2 and/or SR-40 cell lines. D, results for image analysis, performed as described in the legend to Fig. 2, where n 17. p values denote significant differences in Fn/c between the two cell lines or between constructs.  
FIGURE 1. Schematic representation of SV40 T-ag fusion protein constructs used in this study. The position of the NLS (PKKKRKV 132 , single letter amino acid code) is shown, together with the RbBS and mutants thereof; substituted residues are denoted in bold type and underlined.  
FIGURE 2. The Rb binding site (RbBS-T-ag amino acids 102–110) reduces T-ag maximal nuclear accumulation . A, CLSM images of COS-7 cells transfected to express the indicated GFP-T-ag proteins 8 h post-transfection . B, results of image analysis, performed using the Image J software to determine the nuclear to cytoplasmic ratio (Fn/c), determined from measurements of nuclear (Fn) and cytoplasmic (Fc) fluorescence, subsequent to the subtraction of background fluorescence (autofluorescence). Results represent the mean S.E. (n 52). p values indicate significant differences between Fn/c values in the absence or presence of the RbBS.  
FIGURE 8. Nuclear accumulation of p110 Rb is reduced in the presence of overexpressed T-ag dependent on negative charge at the Ser 106 phosphorylation site. A, CLSM images are shown for CV-1 cells transfected to express GFP-T-ag-(102–135) constructs as indicated, followed by fixation 16-h posttransfection , and immunostaining using a specific anti-p110 Rb antibody followed by an Alexa568-labeled secondary antibody. B, results for image analysis of CLSM files such as those in A, performed as per Fig. 2, showing the average Fn (left), Fc (middle), and Fn/c ratio (right) (n 20) of p110 Rb in the presence of the indicated GFP-T-ag derivatives; Fn and Fc values are expressed in percent relative to those for p110 Rb in the absence of exogenous T-ag expression. Significant differences in the absence or presence of the T-ag constructs are indicated by the p values. C, results for image analysis of CLSM files such as those in A, performed as per Fig. 2, showing the average Fn/c ratio (n 20) of the GFP-T-ag derivatives. Significant differences (p 0.0001) are evident between the values for both the WT and Asp 106 derivative, compared with that for GFP-T-ag-(102–135:A 106 ).  
Binding of p110 Retinoblastoma Protein Inhibits Nuclear Import of Simian Virus SV40 Large Tumor Antigen

March 2010

·

80 Reads

·

13 Citations

Journal of Biological Chemistry

Nuclear import of the simian virus 40 large tumor antigen (T-ag) is dependent on its nuclear localization signal (NLS) within amino acids 126-132 that is recognized by the importin alpha/beta1 heterodimer, as well as a protein kinase CK2 site at serine 112 upstream of the NLS, which enhances the interaction approximately 50-fold. Here we show for the first time that T-ag nuclear import is negatively regulated by N-terminal sequences (amino acids 102-110), which represent the binding site (BS) for the retinoblastoma (Rb) tumor suppressor protein (p110(Rb)). Quantitative confocal laser scanning microscopic analysis of the transport properties of T-ag constructs with or without Rb binding site mutations in living transfected cells or in a reconstituted nuclear transport system indicates that the presence of the RbBS significantly reduces nuclear accumulation of T-ag. A number of approaches, including the analysis of T-ag nuclear import in an isogenic cell pair with and without functional p110(Rb) implicate p110(Rb) binding as being responsible for the reduced nuclear accumulation, with the Ser(106) phosphorylation site within the RbBS appearing to enhance the inhibitory effect. Immunoprecipitation experiments confirmed association of T-ag and p110(Rb) and dependence thereof on negative charge at Ser(106). The involvement of p110(Rb) in modulating T-ag nuclear transport has implications for the regulation of nuclear import of other proteins from viruses of medical significance that interact with p110(Rb), and how this may relate to transformation.


Crystal Structure of a Legionella pneumophila Ecto -Triphosphate Diphosphohydrolase, A Structural and Functional Homolog of the Eukaryotic NTPDases

February 2010

·

62 Reads

·

43 Citations

Structure

Many pathogenic bacteria have sophisticated mechanisms to interfere with the mammalian immune response. These include the disruption of host extracellular ATP levels that, in humans, is tightly regulated by the nucleoside triphosphate diphosphohydrolase family (NTPDases). NTPDases are found almost exclusively in eukaryotes, the notable exception being their presence in some pathogenic prokaryotes. To address the function of bacterial NTPDases, we describe the structures of an NTPDase from the pathogen Legionella pneumophila (Lpg1905/Lp1NTPDase) in its apo state and in complex with the ATP analog AMPPNP and the subtype-specific NTPDase inhibitor ARL 67156. Lp1NTPDase is structurally and catalytically related to eukaryotic NTPDases and the structure provides a basis for NTPDase-specific inhibition. Furthermore, we demonstrate that the activity of Lp1NTPDase correlates directly with intracellular replication of Legionella within macrophages. Collectively, these findings provide insight into the mechanism of this enzyme and highlight its role in host-pathogen interactions.


FIG. 1. Dynamic localization of M protein in the nucleus and cytoplasm in RSV-infected cells. (A) Vero cells were infected with RSV, fixed, and permeabilized at the times indicated, and M localization was visualized using MAbM and Alexa Fluor 488-coupled secondary antibodies. Fluorescence imaging was performed in the Yokogawa spinning-disk confocal with Andor Ixon EMCCD camera using a 100 oil immersion objective lens. Scale bar, 10 m. (B) RSV-infected Vero cells were collected at 6 and 24 h p.i., separated into nuclear and cytoplasmic fractions, and electrophoresed on a 4 to 20% polyacrylamide gel. After Western transfer, the blot was probed with RabbitM, followed by alkaline phosphatase-conjugated secondary antibody with bound antibodies detected using Western Blue stabilized substrate for alkaline phosphatase (Promega). Molecular mass markers are indicated on the right; the position of M is shown on the left. N, nuclear fraction; C, cytoplasmic fraction.  
FIG. 3. Subcellular localization of RSV M truncated derivatives in living, transfected Vero cells. (A) Schematic diagram of the M sequence and truncated GFP-M constructs highlighting the nuclear targeting sequences, including the binding site for IMP1 (14) within amino acids 110 to 183, and the putative nNES and cNES sequences shown in the single-letter code. The numbers refer to the M amino acid sequence. (B) Subcellular localization of full-length and truncated M proteins in transfected Vero cells in the absence or presence of LMB. Vero cells were transfected to express the GFP-M or GFP-Rev(2-116) fusion proteins as indicated and treated either without (LMB) or with 2.8 ng of LMB/ml (LMB) for 6 h prior to live CLSM imaging at 24 h posttransfection using a 100 oil immersion objective lens as described previously (41). Scale bar, 10 m. (C) CLSM files such as those in panel B were analyzed as described in the legend to Fig. 2B. The results shown are for the means the SEM (n 20) determined from a single experiment, representative of two similar experiments, with significant differences between the Fn/c values in the absence or presence of LMB as indicated.  
FIG. 4. RSV M contains a Crm1-recognized NES as shown by analysis of point mutant derivatives. (A) Schematic diagram of the mutational substitutions within the nNES, cNES, and NLS sequences, with mutated residues shown in lowercase letters (see also Fig. 3A). (B) Subcellular localization of NES and NLS mutants of M protein in transfected Vero cells in the absence or presence of LMB. Vero cells were transfected to express either GFP alone or GFP-M fusion proteins as indicated and treated either without or with 2.8 ng of LMB/ml as indicated 6 h prior to live cell CLSM imaging as described for Fig. 3. Scale bar, 10 m. (C) CLSM files such as those in panel B, were analyzed as described in the legend to Fig. 2B. The results shown are for the means the SEM (n 30) determined from a single experiment, representative of two similar experiments, with significant differences in the Fn/c values between the absence and presence of LMB as indicated.
FIG. 5. Mutation of the cNES or NLS of M leads to loss in replication fitness. (A) Vero cells were transfected with plasmids encoding the RSV genome coding for wild-type M (rA2), nuclear-import-impaired M (rA2NLSm), or nuclear-export-deficient M (rA2cNESm). Virus was rescued from the supernatant and used to infect A549 cells; samples were collected every day for 5 days and titers of the infectious virus were determined as described in the text. rA2NLSm was recovered from all three rescued attempts, whereas rA2cNESm could not be recovered in seven attempts (data not shown). The data are shown as the logPFU/ml over time (means the SEM of triplicate samples). (B) Vero cells were infected with rA2 or rA2NLSm virus at an MOI of 3; samples were collected at indicated times, and titers of infectious virus were determined as described in the text. (C) Vero cells infected as in panel B were fixed, permeabilized, and analyzed for subcellular localization of M, as described in the legend to Fig. 1, at 6 h and 24 h p.i. (D) CLSM files such as those in panel C were analyzed as described in the legend to Fig. 2B. The results shown are for the means the SEM (n 15) determined from two similar experiments, with significant differences in the Fn/c values between the two virus infections as indicated.
The Respiratory Syncytial Virus Matrix Protein Possesses a Crm1-Mediated Nuclear Export Mechanism

January 2009

·

184 Reads

·

98 Citations

The respiratory syncytial virus (RSV) matrix (M) protein is localized in the nucleus of infected cells early in infection but is mostly cytoplasmic late in infection. We have previously shown that M localizes in the nucleus through the action of the importin beta1 nuclear import receptor. Here, we establish for the first time that M's ability to shuttle to the cytoplasm is due to the action of the nuclear export receptor Crm1, as shown in infected cells, and in cells transfected to express green fluorescent protein (GFP)-M fusion proteins. Specific inhibition of Crm1-mediated nuclear export by leptomycin B increased M nuclear accumulation. Analysis of truncated and point-mutated M derivatives indicated that Crm1-dependent nuclear export of M is attributable to a nuclear export signal (NES) within residues 194 to 206. Importantly, inhibition of M nuclear export resulted in reduced virus production, and a recombinant RSV carrying a mutated NES could not be rescued by reverse genetics. That this is likely to be due to the inability of a nuclear export deficient M to localize to regions of virus assembly is indicated by the fact that a nuclear-export-deficient GFP-M fails to localize to regions of virus assembly when expressed in cells infected with wild-type RSV. Together, our data suggest that Crm1-dependent nuclear export of M is central to RSV infection, representing the first report of such a mechanism for a paramyxovirus M protein and with important implications for related paramyxoviruses.


Human Cytomegalovirus DNA Polymerase Catalytic Subunit pUL54 Possesses Independently Acting Nuclear Localization and ppUL44 Binding Motifs

November 2006

·

93 Reads

·

34 Citations

Traffic

The catalytic subunit of human cytomegalovirus (HCMV) DNA polymerase pUL54 is a 1242-amino-acid protein, whose function, stimulated by the processivity factor, phosphoprotein UL44 (ppUL44), is essential for viral replication. The C-terminal residues (amino acids 1220-1242) of pUL54 have been reported to be sufficient for ppUL44 binding in vitro. Although believed to be important for functioning in the nuclei of infected cells, no data are available on either the interaction of pUL54 with ppUL44 in living mammalian cells or the mechanism of pUL54 nuclear transport and its relationship with that of ppUL44. The present study examines for the first time the nuclear import pathway of pUL54 and its interaction with ppUL44 using dual color, quantitative confocal laser scanning microscopy on live transfected cells and quantitative gel mobility shift assays. We showed that of two nuclear localization signals (NLSs) located at amino acids 1153-1159 (NLSA) and 1222-1227 (NLSB), NLSA is sufficient to confer nuclear localization on green fluorescent protein (GFP) by mediating interaction with importin alpha/beta. We also showed that pUL54 residues 1213-1242 are sufficient to confer ppUL44 binding abilities on GFP and that pUL54 and ppUL44 can be transported to the nucleus as a complex. Our work thus identified distinct sites within the HCMV DNA polymerase, which represent potential therapeutic targets and establishes the molecular basis of UL54 nuclear import.


Nuclear Import of the Respiratory Syncytial Virus Matrix Protein Is Mediated By Importin β1 Independent of Importin α †

October 2005

·

56 Reads

·

103 Citations

Biochemistry

The matrix (M) protein of respiratory syncytial virus (RSV) plays an important role in virus assembly through specific interactions with RSV nucleocapsids and envelope glycoproteins in the cytoplasm as well as with the host cell membrane. We have previously shown that M localizes to the nucleus of infected cells at an early stage in the RSV infection cycle, where it may be instrumental in inhibiting host cell processes. The present study uses transient expression of M as well as a truncated green fluorescent protein (GFP) fusion derivative to show for the first time that M is able to localize in the nucleus in the absence of other RSV gene products, through the action of amino acids 110-183, encompassing the nucleic acid binding regions of the protein, that are sufficient to target GFP to the nucleus. Using native PAGE, ELISA-based binding assays, a novel Alphascreen assay, and an in vitro nuclear transport assay, we show that M is recognized directly by the importin beta1 nuclear import receptor, which mediates its nuclear import in concert with the guanine nucleotide-binding protein Ran. Retention of M in the nucleus through binding to nuclear components, probably mediated by the putative zinc finger domain of M, also contributes to M nuclear accumulation. This is the first report of the importin binding and nuclear import properties of a gene product from a negative sense RNA virus, with implications for the function of RSV M and possibly other viral M proteins in the nucleus of infected cells.


Apoptin Nuclear Accumulation Is Modulated by a CRM1-Recognized Nuclear Export Signal that Is Active in Normal but not in Tumor Cells

September 2005

·

26 Reads

·

122 Citations

Cancer Research

Tumor cell-specific activity of chicken anemia virus viral protein 3 (VP3 or apoptin) is believed to be dependent on its ability to localize in the nucleus of transformed but not of primary or nontransformed cells. The present study characterizes the signals responsible for the novel nucleocytoplasmic trafficking properties of VP3 using two isogenic tumor/nontumor cell pairs. In addition to the tumor cell-specific nuclear targeting signal, comprising two stretches of basic amino acids in the VP3 COOH terminus which are highly efficient in tumor but not in normal cells, we define the CRM1-recognized nuclear export sequence (NES) within the VP3 tumor cell-specific nuclear targeting signal for the first time. Intriguingly, the NES (amino acids 97-105) is functional in normal but not in tumor cells through the action of the threonine 108 phosphorylation site adjacent to the NES which inhibits its action. In addition, we characterize a leucine-rich sequence (amino acids 33-46) that assists VP3 nuclear accumulation by functioning as a nuclear retention sequence, conferring association with promyelocytic leukemia nuclear bodies. This unique combination of signals is the basis of the tumor cell-specific nuclear targeting abilities of VP3.


Fig. 1. Continued. 
Fig. 1. NLS dependence of Imp binding to the T-ag NLS as shown by native PAGE/fluorimaging; evidence for autoinhibition of Imp α (A) 2 μ M GFP T-ag (102–135) or GFP itself, were incubated either alone or with 10 μ M Tr- α , Imp β or Imp α/β as indicated. (B) A 2 μ M GFP T-ag (102–135) ( upper panel ) or GFP T-ag (102–135/Thr 128 ) ( lower panel ), were incubated with increasing concentrations of Imp α/β as indicated. The 
Quantitative Analysis of Protein–Protein Interactions by Native Page/Fluorimaging

August 2005

·

254 Reads

·

25 Citations

Journal of Fluorescence

We have developed a new quantitative native PAGE mobility shift assay, which allows for the measurement of binding affinities for interacting protein pairs, one of which is fluorescently labelled. We have used it to examine recognition of the Simian virus 40 (SV40) large tumour T-antigen (T-ag) nuclear localisation sequence (NLS) by members of the importin (Imp) superfamily of nuclear transport proteins. We demonstrate that the T-ag NLS binds to the Imp alpha/beta heterodimer in NLS-dependent manner, determining that it binds with eight-fold higher affinity (340 nM), when compared to Imp alpha alone, consistent with autoinhibition of Imp alpha when not complexed with Imp beta. The mobility shift assay is able to detect nM binding affinities, making it a sensitive and useful tool to analyse protein-protein interactions in solution.


FIG. 1. The mouse caspase-2 prodomain. A, the N-terminal region is shown in italics, and the putative bipartite NLS is indicated. The locations of the six-helices of the CARD are underlined. The linker region, which lies between the CARD and the caspase domain, is shown in gray. B, schematic representation of caspase-2 GFP fusion proteins used in localization and immunoprecipitation studies. The GFP tag is located at the C terminus of caspase-2. Link, linker region (shown as a black box); p18/p14, the caspase-2 catalytic region. FIG. 2. The linker region of the prodomain mediates nuclear localization of caspase-2. COS cells were transfected with the indicated GFP constructs and 24 h later were fixed and analyzed by confocal microscopy. A, prodomain-GFP; B, CARD-GFP; C, PDMLink-GFP; D, PDMCARD-GFP; E, caspase-3-GFP; and F, caspase-2Link caspase-3-GFP. 
FIG. 3. Identification of a classical NLS at the C terminus of the caspase-2 prodomain. Alignment of amino acid residues (149 –156) of mouse caspase-2 with human and rat caspase-2 and human c-Myc protein. A, conserved residues are indicated in boldface type. COS cells transfected with the indicated wild-type or mutant (K152A) GFP constructs were fixed and analyzed using a confocal microscope. B, caspase2-GFP; C, caspase-2(K152A)-GFP; D, prodomain-GFP; E, prodomain (K152A)-GFP; F, PDMCARD-GFP; G, PDMCARD(K152A)-GFP. 
FIG. 4. Caspase-2 NLS is recognized by importin /. Recognition of caspase 2 by the importin / heterodimer as indicated by native gel electrophoresis and fluorescent gel imaging. The indicated GFP fusion proteins were incubated in the absence or presence of importins for 15 min at room temperature, prior to native electrophoresis on a 12% polyacrylamide gel. After 5 h electrophoresis at 80 V, the gel was imaged using a Fluorimager. The position of the different importin (Imp)-GFP fusion protein complexes are indicated. 
FIG. 5. A short region of CARD is necessary for higher order structures but not for homodimerization of caspase-2. COS cells transfected with the indicated GFP constructs were fixed and analyzed using a confocal microscope. A, WT caspase-2-GFP; B, PDM25-GFP; C, PDM44-GFP. D, HEK293T cells were co-transfected with GFP-tagged and HAtagged caspase-2 expression constructs and lysed 24 h following transfection. Expression of HA-tagged proteins in each lysate was checked by immunoblot analysis using an anti-HA monoclonal antibody (top panel). GFP fusion proteins were immunoprecipitated (IP) from each sample using an anti-GFP monoclonal antibody. Co-precipitated HA-tagged proteins were detected by immunoblot analysis using an anti-HA monoclonal antibody (middle panel). Immunoprecipitation of GFP constructs was checked by immunoblot analysis with an anti-GFP monoclonal antibody (bottom panel). WB, Western blot. E, PDM44 Gal4 DNA binding domain and Gal4 activation domain fusion constructs were co-transfected together, or with the appropriate empty vector, into the S. cerevisiae Y190 strain. Colonies were assayed for-galactosidase activity by colony filter assay using X-gal as a substrate. 
Role of Prodomain in Importin-mediated Nuclear Localization and Activation of Caspase-2

March 2003

·

39 Reads

·

107 Citations

Journal of Biological Chemistry

Caspase-2 is unique among mammalian caspases because it localizes to the nucleus in a prodomain-dependent manner. The caspase-2 prodomain also regulates caspase-2 activity via a caspase recruitment domain that mediates oligomerization of procaspase-2 molecules and their subsequent autoactivation. In this study we sought to map specific functional regions in the caspase-2 prodomain that regulate its nuclear transport and also its activation. Our data indicate that caspase-2 contains a classical nuclear localization signal (NLS) at the C terminus of the prodomain which is recognized by the importin alpha/beta heterodimer. The mutation of a conserved Lys residue in the NLS abolishes nuclear localization of caspase-2 and binding to the importin alpha/beta heterodimer. Although caspase-2 is imported into the nucleus, mutants lacking the NLS were still capable of inducing apoptosis upon overexpression in transfected cells. We define a region in the prodomain that regulates the ability of caspase-2 to form dot- and filament-like structures when ectopically expressed, which in turn promotes cell killing. Our data provides a mechanism for caspase-2 nuclear import and demonstrate that association of procaspase-2 into higher order structures, rather than its nuclear localization, is required for caspase-2 activation and its ability to induce apoptosis.

Citations (10)


... Dysregulation of nucleocytoplasmic transport can severely impact cell function [1,[6][7][8], with the inhibition of transport through the use of small molecules a promising therapeutic strategy for cancer and infectious diseases [1,4,9]. Various inhibitors of IMPα such as the macrocyclic lactone ivermectin [10] have been a focus of interest as antiviral agents [1,4,9,[11][12][13][14][15][16], and more recently as agents to combat the apicomplexan Plasmodium species and Toxoplasma gondii [17,18], the causative agents of malaria and toxoplasmosis, respectively [19][20][21][22][23][24]. ...

Reference:

The Ivermectin Related Compound Moxidectin Can Target Apicomplexan Importin α and Limit Growth of Malarial Parasites
High-Throughput Screening to Identify Inhibitors of Plasmodium falciparum Importin α

Cells

... In this assay, SUFU protein bound quite tightly to GLI1, with a Kd of~4 nM (Fig 2). These results are consistent with other published measurements of the binding affinity between GLI1 and SUFU (Cherry et al, 2013;Szczepny et al, 2014). ...

Overlapping binding sites for importin β1 and Suppressor of Fused on Glioma-associated oncogene homolog 1 (Gli1) regulate its nuclear localisation.
  • Citing Article
  • May 2014

Biochemical Journal

... Indeed, cargo phosphorylation can either promote [10][11][12][13][14][15] or impair the process [11,[16][17][18][19][20]. Secondly, although phosphorylation-dependent regulation of nuclear import has been proposed to rely on the modulation of cargo-IMP interactions, it remains uncertain whether the latter depends on a change in affinity for IMPs [13,17] or for other factors that compete with IMPs [21][22][23]. Additionally, phosphorylation of certain cargoes has been proposed to result in conformational changes regulating NLS exposure, and therefore IMP binding and subsequent nuclear import, as seen in hepatitis B core protein where phosphorylation likely acts as an important modulator of the viral life cycle [24]. ...

Binding of p110 Retinoblastoma Protein Inhibits Nuclear Import of Simian Virus SV40 Large Tumor Antigen

Journal of Biological Chemistry

... Molecular dynamic simulations indicated that the structure of TcNTPDase1 undergoes a certain degree of conformational changes, and it is predicted that this enzyme may assume a more open conformation (Fig. 8). We know that structural studies of the crystals of NTPDases from Rattus norvegicus [71] and Legionella pneumophila [94] revealed the presence of two structural domains (α and β) in addition to identifying the catalytic site of the enzyme and proposing a mechanism of action. The crystal of LpNTPDase1 was obtained both in the presence and absence of a ligand and showed a more open conformation without the ligand [94]. ...

Crystal Structure of a Legionella pneumophila Ecto -Triphosphate Diphosphohydrolase, A Structural and Functional Homolog of the Eukaryotic NTPDases
  • Citing Article
  • February 2010

Structure

... The mammalian expression construct encoding full-length wild-type GFP-M has been described previously [22]. A mutant construct was also generated, where the Valine at 153 was mutated to Alanine (Mmut, M-V153A) using site-directed mutagenesis, followed by recombination into the pEpiDESTC vector [22]. ...

The Respiratory Syncytial Virus Matrix Protein Possesses a Crm1-Mediated Nuclear Export Mechanism

... Similar studies have observed phosphorylation mediated nuclear export as tumour specific regulator for the viral protein apoptin [66]. Where apoptin contains a CRM1 recognised nuclear export signal that is turned off in transformed human osteosarcoma cells following phosphorylation [67]. Our study was unable to identify a functioning nuclear export signal, even after investigating four possible candidates (unpublished data), but the point remains that ENO1-S419 phosphorylation appears to play a role in the recognition or regulation of CRM1/ENO1 interaction. ...

Apoptin Nuclear Accumulation Is Modulated by a CRM1-Recognized Nuclear Export Signal that Is Active in Normal but not in Tumor Cells
  • Citing Article
  • September 2005

Cancer Research

... For analysis of binding in a native gel, EMSAs were performed using IMPαΔIBB proteins ± FITC-cNLS peptides, as described previously (Wagstaff et al., 2005). Briefly, a 1.5% agarose gel in TB buffer (0.45 mM tris, 0.45 mM boric acid, pH 8.5) was loaded with recombinant IMPαΔIBBs ± FITC-cNLS peptides and run at 70 V for 2 h. ...

Quantitative Analysis of Protein–Protein Interactions by Native Page/Fluorimaging

Journal of Fluorescence

... In the process of RSV genome replication and mRNA transcription and translation, the negative strand of the genome is wrapped in the nucleoprotein (N) to form the nucleocapsid. The RNAdependent RNA polymerase complex (RdRp) is composed of the large polymerase subunit (L), the phosphoprotein cofactor (P) of the polymerase ribonucleoprotein, and the M2-1 cofactor is formed [14,15]. The last residue of the P protein (Phe241) is essential for its binding to the nucleoprotein [16,17], and structural details on the interactions between the N-terminal domain of the nucleoprotein and the C-terminal dipeptide of the P protein (Asp240-Phe241) reported by Ouizougun-Oubari et al. (2015) showed the possibility that this interaction of this hydrophobic pocket (N/P) might be druggable and that the aromatic chemicals (1-benzyl-1Hpyrazole-3,5-dicarboxylate) might bind to it and prevent hRSV replication [18]. ...

Nuclear Import of the Respiratory Syncytial Virus Matrix Protein Is Mediated By Importin β1 Independent of Importin α †
  • Citing Article
  • October 2005

Biochemistry

... In addition to the HSV-1 DNA polymerase complex (UL30/ UL42), the DNA polymerase catalytic and accessory subunits of a number of other herpesviruses may also replicate the AAV genome. These include UL54 and UL44 of Human cytomegalovirus (HCMV) (Alvisi et al. 2006), BALF5 and BMRF1 of Epstein-Barr virus (EBV) (Kawashima et al. 2013) and Pol-8 and PF-8 of Kaposi's sarcoma-associated herpesvirus (KSHV) (Chen, Ciustea, and Ricciardi 2005). Their potential for use in our rAAV producer cells is yet to be assessed. ...

Human Cytomegalovirus DNA Polymerase Catalytic Subunit pUL54 Possesses Independently Acting Nuclear Localization and ppUL44 Binding Motifs
  • Citing Article
  • November 2006

Traffic

... CASP2 contains a conservative caspase activation and recruitment domain (CARD), a large p19 subunit and a small p12 subunit 46 . Previous findings indicate that the CARD domain is required for nuclear distribution 47 . In our assays the CARD domain of CASP2 (CASP2-CARD) was sufficient to form a punctum, whereas deletion of CARD domain (CASP2-ΔCARD) failed (Fig. 6a). ...

Role of Prodomain in Importin-mediated Nuclear Localization and Activation of Caspase-2

Journal of Biological Chemistry