Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters











Publication year range
1.
Commun Biol ; 7(1): 486, 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38649430

ABSTRACT

The ongoing evolution of SARS-CoV-2 to evade vaccines and therapeutics underlines the need for innovative therapies with high genetic barriers to resistance. Therefore, there is pronounced interest in identifying new pharmacological targets in the SARS-CoV-2 viral life cycle. The small molecule PAV-104, identified through a cell-free protein synthesis and assembly screen, was recently shown to target host protein assembly machinery in a manner specific to viral assembly. In this study, we investigate the capacity of PAV-104 to inhibit SARS-CoV-2 replication in human airway epithelial cells (AECs). We show that PAV-104 inhibits >99% of infection with diverse SARS-CoV-2 variants in immortalized AECs, and in primary human AECs cultured at the air-liquid interface (ALI) to represent the lung microenvironment in vivo. Our data demonstrate that PAV-104 inhibits SARS-CoV-2 production without affecting viral entry, mRNA transcription, or protein synthesis. PAV-104 interacts with SARS-CoV-2 nucleocapsid (N) and interferes with its oligomerization, blocking particle assembly. Transcriptomic analysis reveals that PAV-104 reverses SARS-CoV-2 induction of the type-I interferon response and the maturation of nucleoprotein signaling pathway known to support coronavirus replication. Our findings suggest that PAV-104 is a promising therapeutic candidate for COVID-19 with a mechanism of action that is distinct from existing clinical management approaches.


Subject(s)
Antiviral Agents , Epithelial Cells , SARS-CoV-2 , Virus Replication , Humans , SARS-CoV-2/drug effects , SARS-CoV-2/physiology , Virus Replication/drug effects , Epithelial Cells/virology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Antiviral Agents/pharmacology , Virus Assembly/drug effects , COVID-19/virology , COVID-19 Drug Treatment
2.
Res Sq ; 2023 May 17.
Article in English | MEDLINE | ID: mdl-37292622

ABSTRACT

The ongoing evolution of SARS-CoV-2 to evade vaccines and therapeutics underlines the need for novel therapies with high genetic barriers to resistance. The small molecule PAV-104, identified through a cell-free protein synthesis and assembly screen, was recently shown to target host protein assembly machinery in a manner specific to viral assembly. Here, we investigated the capacity of PAV-104 to inhibit SARS-CoV-2 replication in human airway epithelial cells (AECs). Our data demonstrate that PAV-104 inhibited > 99% of infection with diverse SARS-CoV-2 variants in primary and immortalized human AECs. PAV-104 suppressed SARS-CoV-2 production without affecting viral entry or protein synthesis. PAV-104 interacted with SARS-CoV-2 nucleocapsid (N) and interfered with its oligomerization, blocking particle assembly. Transcriptomic analysis revealed that PAV-104 reversed SARS-CoV-2 induction of the Type-I interferon response and the 'maturation of nucleoprotein' signaling pathway known to support coronavirus replication. Our findings suggest that PAV-104 is a promising therapeutic candidate for COVID-19.

3.
Biosci Rep ; 41(10)2021 10 29.
Article in English | MEDLINE | ID: mdl-34677582

ABSTRACT

The role of human prostatic acid phosphatase (PAcP, P15309|PPAP_HUMAN) in prostate cancer was investigated using a new proteomics tool termed signal sequence swapping (replacement of domains from the native cleaved amino terminal signal sequence of secretory/membrane proteins with corresponding regions of functionally distinct signal sequence subtypes). This manipulation preferentially redirects proteins to different pathways of biogenesis at the endoplasmic reticulum (ER), magnifying normally difficult to detect subsets of the protein of interest. For PAcP, this technique reveals three forms identical in amino acid sequence but profoundly different in physiological functions, subcellular location, and biochemical properties. These three forms of PAcP can also occur with the wildtype PAcP signal sequence. Clinical specimens from patients with prostate cancer demonstrate that one form, termed PLPAcP, correlates with early prostate cancer. These findings confirm the analytical power of this method, implicate PLPAcP in prostate cancer pathogenesis, and suggest novel anticancer therapeutic strategies.


Subject(s)
Acid Phosphatase/metabolism , Biomarkers, Tumor/metabolism , Cell Proliferation , Endoplasmic Reticulum/enzymology , Prostatic Neoplasms/enzymology , Acid Phosphatase/genetics , Androgens/pharmacology , Antineoplastic Agents, Hormonal/pharmacology , Biomarkers, Tumor/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm , Early Detection of Cancer , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/pathology , Humans , Isoenzymes , Male , Predictive Value of Tests , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Protein Conformation , Structure-Activity Relationship
4.
Biosci Rep ; 2021 Oct 04.
Article in English | MEDLINE | ID: mdl-34605872

ABSTRACT

The role of human prostatic acid phosphatase (PAcP, P15309|PPAP_HUMAN) in prostate cancer was investigated using a new proteomic tool termed signal sequence swapping (replacement of domains from the native cleaved amino terminal signal sequence of secretory/membrane proteins with corresponding regions of functionally distinct signal sequence subtypes). This manipulation preferentially redirects proteins to different pathways of biogenesis at the endoplasmic reticulum, magnifying normally difficult to detect subsets of the protein of interest. For PAcP this technique reveals three forms identical in amino acid sequence but profoundly different in physiological functions, subcellular location, and biochemical properties. These three forms of PAcP can also occur with the wild-type PAcP signal sequence. Clinical specimens from patients with prostate cancer demonstrate that one form, termed PLPAcP, correlates with early prostate cancer. These findings confirm the analytical power of this method, implicate PLPAcP in prostate cancer pathogenesis, and suggest novel anticancer therapeutic strategies.

5.
Open Biol ; 11(6): 200400, 2021 06.
Article in English | MEDLINE | ID: mdl-34186010

ABSTRACT

Wnt gradients elicit distinct cellular responses, such as proliferation, specification, differentiation and survival in a dose-dependent manner. Porcupine (PORCN), a membrane-bound O-acyl transferase (MBOAT) that resides in the endoplasmic reticulum, catalyses the addition of monounsaturated palmitate to Wnt proteins and is required for Wnt gradient formation and signalling. In humans, PORCN mutations are causal for focal dermal hypoplasia (FDH), an X-linked dominant syndrome characterized by defects in mesodermal and endodermal tissues. PORCN is also an emerging target for cancer therapeutics. Despite the importance of this enzyme, its structure remains poorly understood. Recently, the crystal structure of DltB, an MBOAT family member from bacteria, was solved. In this report, we use experimental data along with homology modelling to DltB to determine the membrane topology of PORCN. Our studies reveal that PORCN has 11 membrane domains, comprising nine transmembrane spanning domains and two reentrant domains. The N-terminus is oriented towards the lumen while the C-terminus is oriented towards the cytosol. Like DltB, PORCN has a funnel-like structure that is encapsulated by multiple membrane-spanning helices. This new model for PORCN topology allows us to map residues that are important for biological activity (and implicated in FDH) onto its three-dimensional structure.


Subject(s)
Acyltransferases/metabolism , Cell Membrane/chemistry , Cell Membrane/metabolism , Membrane Proteins/metabolism , Wnt Signaling Pathway , Acyltransferases/chemistry , Algorithms , Animals , Cell Line , Computational Biology/methods , Consensus Sequence , Fluorescent Antibody Technique , Glycosylation , Humans , Membrane Proteins/chemistry , Models, Molecular , Molecular Dynamics Simulation , Protein Binding , Protein Conformation , Structure-Activity Relationship
6.
Antiviral Res ; 191: 105086, 2021 07.
Article in English | MEDLINE | ID: mdl-33992710

ABSTRACT

Decades after the eradication of smallpox and the discontinuation of routine smallpox vaccination, over half of the world's population is immunologically naïve to variola virus and other orthopoxviruses (OPXVs). Even in those previously vaccinated against smallpox, protective immunity wanes over time. As such, there is a concomitant increase in the incidence of human OPXV infections worldwide. To identify novel antiviral compounds with potent anti-OPXV potential, we characterized the inhibitory activity of PAV-866 and other methylene blue derivatives against the prototypic poxvirus, vaccinia virus (VACV). These compounds inactivated virions prior to infection and consequently inhibited viral binding, fusion and entry. The compounds exhibited strong virucidal activity at non-cytotoxic concentrations, and inhibited VACV infection when added before, during or after viral adsorption. The compounds were effective against other OPXVs including monkeypox virus, cowpox virus and the newly identified Akhmeta virus. Altogether, these findings reveal a novel mode of inhibition that has not previously been demonstrated for small molecule compounds against VACV. Additional studies are in progress to determine the in vivo efficacy of these compounds against OPXVs and further characterize the anti-viral effects of these derivatives.


Subject(s)
Antiviral Agents/pharmacology , Methylene Blue/chemistry , Methylene Blue/pharmacology , Orthopoxvirus/drug effects , Antiviral Agents/chemistry , Cell Line , Cowpox virus/drug effects , HeLa Cells , Humans , Monkeypox virus/drug effects , Orthopoxvirus/classification , Vaccinia virus/drug effects , Virus Attachment/drug effects
7.
Viruses ; 13(3)2021 03 10.
Article in English | MEDLINE | ID: mdl-33802145

ABSTRACT

The concerning increase in HIV-1 resistance argues for prioritizing the development of host-targeting antiviral drugs because such drugs can offer high genetic barriers to the selection of drug-resistant viral variants. Targeting host proteins could also yield drugs that act on viral life cycle events that have proven elusive to inhibition, such as intracellular events of HIV-1 immature capsid assembly. Here, we review small molecule inhibitors identified primarily through HIV-1 self-assembly screens and describe how all act either narrowly post-entry or broadly on early and late events of the HIV-1 life cycle. We propose that a different screening approach could identify compounds that specifically inhibit HIV-1 Gag assembly, as was observed when a potent rabies virus inhibitor was identified using a host-catalyzed rabies assembly screen. As an example of this possibility, we discuss an antiretroviral small molecule recently identified using a screen that recapitulates the host-catalyzed HIV-1 capsid assembly pathway. This chemotype potently blocks HIV-1 replication in T cells by specifically inhibiting immature HIV-1 capsid assembly but fails to select for resistant viral variants over 37 passages, suggesting a host protein target. Development of such small molecules could yield novel host-targeting antiretroviral drugs and provide insight into chronic diseases resulting from dysregulation of host machinery targeted by these drugs.


Subject(s)
Anti-Retroviral Agents/pharmacology , Drug Resistance, Viral , HIV-1/drug effects , Host Microbial Interactions/drug effects , Virus Assembly/drug effects , Anti-Retroviral Agents/isolation & purification , Capsid/metabolism , HIV Seropositivity , HIV-1/physiology , Humans , T-Lymphocytes/drug effects , T-Lymphocytes/virology
8.
Trends Neurosci ; 44(5): 352-365, 2021 05.
Article in English | MEDLINE | ID: mdl-33317827

ABSTRACT

The ability of viruses to evolve several orders of magnitude faster than their host cells has enabled them to exploit host cellular machinery by selectively recruiting multiprotein complexes (MPCs) for their catalyzed assembly and replication. This hijacking may depend on alternative, 'moonlighting' functions of host proteins that deviate from their canonical functions thereby inducing cellular pathology. Here, we posit that if virus-induced cellular pathology is similar to that of other, unknown (non-viral) causes, the identification and molecular characterization of the host proteins involved in virus-mediated cellular pathology can be leveraged to decipher the non-viral disease-relevant mechanisms. We focus on how virus-induced aberrant proteostasis and protein aggregation resemble the cellular pathology of sporadic neurodegenerative diseases (NDs) and how this can be exploited for drug discovery.


Subject(s)
Brain , Viruses , Brain/pathology , Brain/virology , Humans , Multiprotein Complexes , Protein Aggregation, Pathological , Proteostasis
9.
J Virol ; 95(3)2021 01 13.
Article in English | MEDLINE | ID: mdl-33148797

ABSTRACT

Given the projected increase in multidrug-resistant HIV-1, there is an urgent need for development of antiretrovirals that act on virus life cycle stages not targeted by drugs currently in use. Host-targeting compounds are of particular interest because they can offer a high barrier to resistance. Here, we report identification of two related small molecules that inhibit HIV-1 late events, a part of the HIV-1 life cycle for which potent and specific inhibitors are lacking. This chemotype was discovered using cell-free protein synthesis and assembly systems that recapitulate intracellular host-catalyzed viral capsid assembly pathways. These compounds inhibit replication of HIV-1 in human T cell lines and peripheral blood mononuclear cells, and are effective against a primary isolate. They reduce virus production, likely by inhibiting a posttranslational step in HIV-1 Gag assembly. Notably, the compound colocalizes with HIV-1 Gag in situ; however, unexpectedly, selection experiments failed to identify compound-specific resistance mutations in gag or pol, even though known resistance mutations developed upon parallel nelfinavir selection. Thus, we hypothesized that instead of binding to Gag directly, these compounds localize to assembly intermediates, the intracellular multiprotein complexes containing Gag and host factors that form during immature HIV-1 capsid assembly. Indeed, imaging of infected cells shows compound colocalized with two host enzymes found in assembly intermediates, ABCE1 and DDX6, but not two host proteins found in other complexes. While the exact target and mechanism of action of this chemotype remain to be determined, our findings suggest that these compounds represent first-in-class, host-targeting inhibitors of intracellular events in HIV-1 assembly.IMPORTANCE The success of antiretroviral treatment for HIV-1 is at risk of being undermined by the growing problem of drug resistance. Thus, there is a need to identify antiretrovirals that act on viral life cycle stages not targeted by drugs in use, such as the events of HIV-1 Gag assembly. To address this gap, we developed a compound screen that recapitulates the intracellular events of HIV-1 assembly, including virus-host interactions that promote assembly. This effort led to the identification of a new chemotype that inhibits HIV-1 replication at nanomolar concentrations, likely by acting on assembly. This compound colocalized with Gag and two host enzymes that facilitate capsid assembly. However, resistance selection did not result in compound-specific mutations in gag, suggesting that the chemotype does not directly target Gag. We hypothesize that this chemotype represents a first-in-class inhibitor of virus production that acts by targeting a virus-host complex important for HIV-1 Gag assembly.


Subject(s)
Anti-Retroviral Agents/pharmacology , HIV Infections/drug therapy , HIV-1/drug effects , Leukocytes, Mononuclear/drug effects , Small Molecule Libraries/pharmacology , Virus Assembly/drug effects , ATP-Binding Cassette Transporters/metabolism , DEAD-box RNA Helicases/metabolism , HIV Infections/pathology , HIV Infections/virology , Humans , Leukocytes, Mononuclear/virology , Proto-Oncogene Proteins/metabolism , gag Gene Products, Human Immunodeficiency Virus/metabolism
10.
Proc Natl Acad Sci U S A ; 117(12): 6741-6751, 2020 03 24.
Article in English | MEDLINE | ID: mdl-32152117

ABSTRACT

Neurodegenerative diseases feature specific misfolded or misassembled proteins associated with neurotoxicity. The precise mechanisms by which protein aggregates first arise in the majority of sporadic cases have remained unclear. Likely, a first critical mass of misfolded proteins starts a vicious cycle of a prion-like expansion. We hypothesize that viruses, having evolved to hijack the host cellular machinery for catalyzing their replication, lead to profound disturbances of cellular proteostasis, resulting in such a critical mass of protein aggregates. Here, we investigated the effect of influenza virus (H1N1) strains on proteostasis of proteins associated with neurodegenerative diseases in Lund human mesencephalic dopaminergic cells in vitro and infection of Rag knockout mice in vivo. We demonstrate that acute H1N1 infection leads to the formation of α-synuclein and Disrupted-in-Schizophrenia 1 (DISC1) aggregates, but not of tau or TDP-43 aggregates, indicating a selective effect on proteostasis. Oseltamivir phosphate, an antiinfluenza drug, prevented H1N1-induced α-synuclein aggregation. As a cell pathobiological mechanism, we identified H1N1-induced blocking of autophagosome formation and inhibition of autophagic flux. In addition, α-synuclein aggregates appeared in infected cell populations connected to the olfactory bulbs following intranasal instillation of H1N1 in Rag knockout mice. We propose that H1N1 virus replication in neuronal cells can induce seeds of aggregated α-synuclein or DISC1 that may be able to initiate further detrimental downstream events and should thus be considered a risk factor in the pathogenesis of synucleinopathies or a subset of mental disorders. More generally, aberrant proteostasis induced by viruses may be an underappreciated factor in initiating protein misfolding.


Subject(s)
Homeodomain Proteins/physiology , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza, Human/complications , Orthomyxoviridae Infections/complications , Proteostasis , Synucleinopathies/etiology , alpha-Synuclein/chemistry , Animals , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Female , Humans , Influenza, Human/virology , Mice , Mice, Knockout , Nerve Tissue Proteins/metabolism , Orthomyxoviridae Infections/virology , Protein Multimerization , Synucleinopathies/metabolism , Synucleinopathies/pathology , alpha-Synuclein/metabolism
11.
J Virol ; 92(9)2018 05 01.
Article in English | MEDLINE | ID: mdl-29467316

ABSTRACT

During immature capsid assembly in cells, human immunodeficiency virus type 1 (HIV-1) Gag co-opts a host RNA granule, forming a pathway of intracellular assembly intermediates containing host components, including two cellular facilitators of assembly, ABCE1 and DDX6. A similar assembly pathway has been observed for other primate lentiviruses. Here we asked whether feline immunodeficiency virus (FIV), a nonprimate lentivirus, also forms RNA granule-derived capsid assembly intermediates. First, we showed that the released FIV immature capsid and a large FIV Gag-containing intracellular complex are unstable during analysis, unlike for HIV-1. We identified harvest conditions, including in situ cross-linking, that overcame this problem, revealing a series of FIV Gag-containing complexes corresponding in size to HIV-1 assembly intermediates. Previously, we showed that assembly-defective HIV-1 Gag mutants are arrested at specific assembly intermediates; here we identified four assembly-defective FIV Gag mutants, including three not previously studied, and demonstrated that they appear to be arrested at the same intermediate as the cognate HIV-1 mutants. Further evidence that these FIV Gag-containing complexes correspond to assembly intermediates came from coimmunoprecipitations demonstrating that endogenous ABCE1 and the RNA granule protein DDX6 are associated with FIV Gag, as shown previously for HIV-1 Gag, but are not associated with a ribosomal protein, at steady state. Additionally, we showed that FIV Gag associates with another RNA granule protein, DCP2. Finally, we validated the FIV Gag-ABCE1 and FIV Gag-DCP2 interactions with proximity ligation assays demonstrating colocalization in situ Together, these data support a model in which primate and nonprimate lentiviruses form intracellular capsid assembly intermediates derived from nontranslating host RNA granules.IMPORTANCE Like HIV-1 Gag, FIV Gag assembles into immature capsids; however, it is not known whether FIV Gag progresses through a pathway of immature capsid assembly intermediates derived from host RNA granules, as shown for HIV-1 Gag. Here we showed that FIV Gag forms complexes that resemble HIV-1 capsid assembly intermediates in size and in their association with ABCE1 and DDX6, two host facilitators of HIV-1 immature capsid assembly that are found in HIV-1 assembly intermediates. Our studies also showed that known and novel assembly-defective FIV Gag mutants fail to progress past putative intermediates in a pattern resembling that observed for HIV-1 Gag mutants. Finally, we used imaging to demonstrate colocalization of FIV Gag with ABCE1 and with the RNA granule protein DCP2. Thus, we conclude that formation of assembly intermediates derived from host RNA granules is likely conserved between primate and nonprimate lentiviruses and could provide targets for future antiviral strategies.


Subject(s)
Capsid Proteins/metabolism , Capsid/metabolism , Gene Products, gag/genetics , HIV-1/metabolism , Immunodeficiency Virus, Feline/metabolism , Virus Assembly/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , COS Cells , Capsid Proteins/genetics , Cats , Cell Line , Chlorocebus aethiops , DEAD-box RNA Helicases/metabolism , Endoribonucleases/metabolism , HIV-1/genetics , Immunodeficiency Virus, Feline/genetics , RNA-Binding Proteins/biosynthesis
12.
Exp Cell Res ; 347(1): 171-183, 2016 09 10.
Article in English | MEDLINE | ID: mdl-27492485

ABSTRACT

Loss-of-function studies have identified Porcupine (PORCN) and Wntless (WLS) as essential mediators of Wnt secretion and signaling. Whereas PORCN is thought to palmitoylate Wnt proteins, WLS is believed to transport palmitoylated Wnt proteins to the cell surface. However, little is known about how these two proteins cooperate to regulate Wnt palmitoylation, trafficking, secretion, and signaling. We first investigated possible interactions between PORCN, WLS, and WNT1, by carrying out co-immunoprecipitation studies. These studies demonstrate the existence of a complex containing PORCN and WLS. They further show that PORCN and WLS compete for binding to WNT1. Then, we used gain-of-function studies to investigate the cooperation between PORCN and WLS as well as possible biochemical interactions between PORCN, WLS, and WNT1. Consistent with the proposed roles for PORCN and WLS, we show that overexpression of PORCN promotes palmitoylation of WNT1 while overexpression of WLS does not. Overexpression of PORCN enhances the ability of WLS to promote WNT1 trafficking to the cell surface as well as secretion, but decreases the ability of WLS to activate WNT1 signaling in target cell. These observations suggest that the levels of WNT1 on the cell surface and in the media are not the sole determinants of the activation of Wnt signaling in target cells.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Wnt Signaling Pathway , Wnt1 Protein/metabolism , Acyltransferases , Animals , Autocrine Communication/drug effects , COS Cells , Chickens , Chlorocebus aethiops , HEK293 Cells , Humans , Hydrophobic and Hydrophilic Interactions , Immunoprecipitation , Lipoylation/drug effects , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Paracrine Communication/drug effects , Protein Binding/drug effects , Protein Transport/drug effects
13.
Biol Direct ; 11: 25, 2016 05 14.
Article in English | MEDLINE | ID: mdl-27179769

ABSTRACT

BACKGROUND: Viral capsid assembly involves the oligomerization of the capsid nucleoprotein (NP), which is an essential step in viral replication and may represent a potential antiviral target. An in vitro transcription-translation reaction using a wheat germ (WG) extract in combination with a sandwich ELISA assay has recently been used to identify small molecules with antiviral activity against the rabies virus. RESULTS: Here, we examined the application of this system to viruses with capsids with a different structure, such as the Rift Valley fever virus (RVFV), the etiological agent of a severe emerging infectious disease. The biochemical and immunological characterization of the in vitro-generated RVFV NP assembly products enabled the distinction between intermediately and highly ordered capsid structures. This distinction was used to establish a screening method for the identification of potential antiviral drugs for RVFV countermeasures. CONCLUSIONS: These results indicated that this unique analytical system, which combines nucleoprotein oligomerization with the specific immune recognition of a highly ordered capsid structure, can be extended to various viral families and used both to study the early stages of NP assembly and to assist in the identification of potential antiviral drugs in a cost-efficient manner. REVIEWERS: Reviewed by Jeffry Skolnick and Noah Isakov. For the full reviews please go to the Reviewers' comments section.


Subject(s)
Antiviral Agents/analysis , Capsid/physiology , Drug Discovery/methods , Drug Evaluation, Preclinical , Rift Valley fever virus/physiology , Cell-Free System , Nucleoproteins/chemistry
14.
Virus Res ; 207: 155-64, 2015 Sep 02.
Article in English | MEDLINE | ID: mdl-25451064

ABSTRACT

Viruses can be conceptualized as self-replicating multiprotein assemblies, containing coding nucleic acids. Viruses have evolved to exploit host cellular components including enzymes to ensure their replicative life cycle. New findings indicate that also viral capsid proteins recruit host factors to accelerate their assembly. These assembly machines are RNA-containing multiprotein complexes whose composition is governed by allosteric sites. In the event of viral infection, the assembly machines are recruited to support the virus over the host and are modified to achieve that goal. Stress granules and processing bodies may represent collections of such assembly machines, readily visible by microscopy but biochemically labile and difficult to isolate by fractionation. We hypothesize that the assembly of protein multimers such as encountered in neurodegenerative or other protein conformational diseases, is also catalyzed by assembly machines. In the case of viral infection, the assembly machines have been modified by the virus to meet the virus' need for rapid capsid assembly rather than host homeostasis. In the case of the neurodegenerative diseases, it is the monomers and/or low n oligomers of the so-called aggregated proteins that are substrates of assembly machines. Examples for substrates are amyloid ß peptide (Aß) and tau in Alzheimer's disease, α-synuclein in Parkinson's disease, prions in the prion diseases, Disrupted-in-schizophrenia 1 (DISC1) in subsets of chronic mental illnesses, and others. A likely continuum between virus capsid assembly and cell-to-cell transmissibility of aggregated proteins is remarkable. Protein aggregation diseases may represent dysfunction and dysregulation of these assembly machines analogous to the aberrations induced by viral infection in which cellular homeostasis is pathologically reprogrammed. In this view, as for viral infection, reset of assembly machines to normal homeostasis should be the goal of protein aggregation therapeutics. A key basis for the commonality between viral and neurodegenerative disease aggregation is a broader definition of assembly as more than just simple aggregation, particularly suited for the crowded cytoplasm. The assembly machines are collections of proteins that catalytically accelerate an assembly reaction that would occur spontaneously but too slowly to be relevant in vivo. Being an enzyme complex with a functional allosteric site, appropriated for a non-physiological purpose (e.g. viral infection or conformational disease), these assembly machines present a superior pharmacological target because inhibition of their active site will amplify an effect on their substrate reaction. Here, we present this hypothesis based on recent proof-of-principle studies against Aß assembly relevant in Alzheimer's disease.


Subject(s)
Capsid Proteins/metabolism , Protein Aggregation, Pathological/metabolism , Virus Diseases/metabolism , Viruses/metabolism , Animals , Capsid/metabolism , Capsid Proteins/genetics , Humans , Models, Biological , Protein Aggregation, Pathological/drug therapy , Virus Assembly , Virus Diseases/drug therapy , Virus Diseases/virology , Viruses/genetics
15.
J Virol Methods ; 193(1): 226-31, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23764417

ABSTRACT

Venezuelan (VEEV), eastern, and western equine encephalitis viruses, members of the genus Alphavirus, are causative agents of debilitative and sometimes fatal encephalitis. Although human cases are rare, these viruses pose a threat to military personnel, and to public health, due to their potential use as bioweapons. Currently, there are no licensed therapeutics for treating alphavirus infections. To address this need, small-molecules with potential anti-alphavirus activity, provided by collaborators, are tested routinely in live alphavirus assays utilizing time-consuming virus yield-reduction assays. To expedite the screening/hit-confirmation process, a cell-based enzyme-linked immunosorbent assay (ELISA) was developed and validated for the measurement of VEEV infection. A signal-to-background ratio of >900, and a z-factor of >0.8 indicated the robustness of this assay. For validation, the cell-based ELISA was compared directly to results from virus yield reduction assays in a single dose screen of 21 compounds. Using stringent criteria for anti-VEEV activity there was 90% agreement between the two assays (compounds displaying either antiviral activity, or no effect, in both assays). A concurrent compound-induced cell toxicity assay effectively filtered out false-positive hits. The cell-based ELISA also reproduced successfully compound dose-response virus inhibition data observed using the virus yield reduction assay. With available antibodies, this assay can be adapted readily to other viruses of interest to the biodefense community. Additionally, it is cost-effective, rapid, and amenable to automation and scale-up. Therefore, this assay could expedite greatly screening efforts and the identification of effective anti-alphavirus inhibitors.


Subject(s)
Alphavirus/drug effects , Antiviral Agents/isolation & purification , Drug Evaluation, Preclinical/methods , Animals , Antiviral Agents/pharmacology , Chlorocebus aethiops , Enzyme-Linked Immunosorbent Assay/methods , Vero Cells
16.
Proc Natl Acad Sci U S A ; 110(10): E861-8, 2013 Mar 05.
Article in English | MEDLINE | ID: mdl-23404707

ABSTRACT

We present an unconventional approach to antiviral drug discovery, which is used to identify potent small molecules against rabies virus. First, we conceptualized viral capsid assembly as occurring via a host-catalyzed biochemical pathway, in contrast to the classical view of capsid formation by self-assembly. This suggested opportunities for antiviral intervention by targeting previously unappreciated catalytic host proteins, which were pursued. Second, we hypothesized these host proteins to be components of heterogeneous, labile, and dynamic multi-subunit assembly machines, not easily isolated by specific target protein-focused methods. This suggested the need to identify active compounds before knowing the precise protein target. A cell-free translation-based small molecule screen was established to recreate the hypothesized interactions involving newly synthesized capsid proteins as host assembly machine substrates. Hits from the screen were validated by efficacy against infectious rabies virus in mammalian cell culture. Used as affinity ligands, advanced analogs were shown to bind a set of proteins that effectively reconstituted drug sensitivity in the cell-free screen and included a small but discrete subfraction of cellular ATP-binding cassette family E1 (ABCE1), a host protein previously found essential for HIV capsid formation. Taken together, these studies advance an alternate view of capsid formation (as a host-catalyzed biochemical pathway), a different paradigm for drug discovery (whole pathway screening without knowledge of the target), and suggest the existence of labile assembly machines that can be rendered accessible as next-generation drug targets by the means described.


Subject(s)
Antiviral Agents/pharmacology , Host-Pathogen Interactions/drug effects , Rabies virus/drug effects , Rabies virus/physiology , Viral Proteins/physiology , Amino Acid Sequence , Animals , Cell-Free System , Chlorocebus aethiops , Drug Discovery , Host-Pathogen Interactions/physiology , Humans , Microbial Sensitivity Tests , Molecular Sequence Data , Nucleocapsid Proteins/chemistry , Nucleocapsid Proteins/genetics , Nucleocapsid Proteins/physiology , Protein Interaction Domains and Motifs , Rabies virus/genetics , Vero Cells , Viral Proteins/chemistry , Viral Proteins/genetics , Virus Assembly/drug effects
17.
Curr Pharm Biotechnol ; 14(5): 513-23, 2013.
Article in English | MEDLINE | ID: mdl-22429134

ABSTRACT

In general, drug discovery in the therapeutic field of infectious disease has a stellar track record. And yet, subsets of pathogens, for example many classes of viruses other than HIV, HSV, influenza, and HCV, have been poorly addressed. In addition, the development of resistance remains a specter of great concern for almost all current chemotherapy directed against infectious diseases, including viruses. Within the viral lifecycle, capsid assembly stands out as a step occurring in all viruses, which has not been the subject of extensive drug discovery programs. Until recently, the common view of assembly was that all the necessary information for assembly was contained in the sequence of the viral protein, in other words, the capsid self-assembles. In the last few years, a body of data has opened new opportunities for antiviral pharmaceutical research. Evidence that host proteins may play catalytic or essential structural roles in viral capsid assembly suggests that these host proteins and their functions are novel targets for small molecule therapeutics. Here we review the current understanding of the capsid assembly process with emphasis on recent data that demonstrate the essential role of host proteins in capsid assembly. Furthermore, this dependency of assembly on host factors appears quite sensitive to small molecule intervention. Implications of this alternate mechanism of capsid assembly are also considered. For example, the dependency on host factors could impose a potent barrier to development of viral resistance to a host-targeted anti-capsid chemotherapeutic. Finally, we give specific examples of the current state of drug discovery programs that have focused on therapeutic inhibition of host-assisted viral capsid assembly.


Subject(s)
Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Capsid/metabolism , Virus Assembly/drug effects , Animals , Capsid Proteins/metabolism , Drug Discovery/methods , Humans
18.
Nephron Exp Nephrol ; 120(4): e134-40, 2012.
Article in English | MEDLINE | ID: mdl-23037926

ABSTRACT

BACKGROUND: Protein translation and translocation at the rough endoplasmic reticulum (RER) are the first steps in the secretory pathway. The translocon through which newly made proteins are translocated into or across the RER membrane consists of three main subunits: Sec61α, -ß, and -γ. Sec61ß facilitates translocation, and we and others have shown that the highly conserved eight-protein exocyst complex interacts with Sec61ß. We have also shown that the exocyst is involved in basolateral, not apical, protein synthesis and delivery. Recently, however, exocyst involvement in apical protein delivery has been reported. Furthermore, we have shown that the exocyst is necessary for formation of primary cilia, organelles found on the apical surface. METHODS: GST pulldown was performed on lysate of renal tubule cells to investigate biochemical interactions. Cell-free assays consisting of cell-free extracts from rabbit reticulocytes, pancreatic endoplasmic reticulum (ER) microsomal membranes, transcripts of cDNA from apical and basolateral proteins, ATP/GTP, amino acids, and (35)S-methionine for protein detection were used to investigate the role of the exocyst in synthesis of polarized proteins. P(32)-orthophosphate and immunoprecipitation with antibody against Sec61ß was used to investigate Sec61ß phosphorylation in exocyst Sec10-overexpressing cells. RESULTS: Sec10 biochemically interacts with Sec61ß using GST pulldown. Using cell-free assays, there is enhanced exocyst recruitment to endoplasmic reticulum membranes following exocyst depletion and basolateral G protein of vesicular stomatitis virus protein translation, compared to apical hemagglutinin of influenza virus protein translation. Finally, Sec10 overexpression increases Sec61ß phosphorylation. CONCLUSION: These data confirm that the exocyst is preferentially involved in basolateral protein translation and translocation, and may well act through the phosphorylation of Sec61ß.


Subject(s)
Endoplasmic Reticulum/metabolism , Membrane Proteins/metabolism , Protein Biosynthesis/physiology , Protein Transport/physiology , Vesicular Transport Proteins/metabolism , Animals , Humans , Rabbits , SEC Translocation Channels
19.
Int J Mol Sci ; 11(10): 4051-62, 2010 Oct 20.
Article in English | MEDLINE | ID: mdl-21152319

ABSTRACT

We recently reported the presence of a novel 32 kDa protein immunoreactive to a copper, zinc superoxide dismutase (SOD1) antibody within the spinal cord of patients with amyotrophic lateral sclerosis (ALS). This unique protein species was generated by biotinylation of spinal cord tissue extracts to detect conformational changes of SOD1 specific to ALS patients. To further characterize this protein, we enriched the protein by column chromatography and determined its protein identity by mass spectrometry. The protein that gave rise to the 32 kDa species upon biotinylation was identified as carbonic anhydrase I (CA I). Biotinylation of CA I from ALS spinal cord resulted in the generation of a novel epitope recognized by the SOD1 antibody. This epitope could also be generated by biotinylation of extracts from cultured cells expressing human CA I. Peptide competition assays identified the amino acid sequence in carbonic anhydrase I responsible for binding the SOD1 antibody. We conclude that chemical modifications used to identify pathogenic protein conformations can lead to the identification of unanticipated proteins that may participate in disease pathogenesis.


Subject(s)
Carbonic Anhydrase I/immunology , Spinal Cord/enzymology , Superoxide Dismutase/immunology , Amyotrophic Lateral Sclerosis/enzymology , Amyotrophic Lateral Sclerosis/immunology , Biotinylation , Humans , Immunoassay , Proteomics , Spinal Cord/immunology , Superoxide Dismutase-1
20.
Biochem Biophys Res Commun ; 393(3): 439-44, 2010 Mar 12.
Article in English | MEDLINE | ID: mdl-20138835

ABSTRACT

The cellular prion protein (PrP(C)) is a GPI-anchored cell-surface protein. A small subset of PrP(C) molecules, however, can be integrated into the ER-membrane via a transmembrane domain (TM), which also harbors the most highly conserved regions of PrP(C), termed the hydrophobic core (HC). A mutation in HC is associated with prion disease resulting in an enhanced formation of a transmembrane form ((Ctm)PrP), which has thus been postulated to be a neurotoxic molecule besides PrP(Sc). To elucidate a possible physiological function of the transmembrane domain, we created a set of mutants carrying microdeletions of 2-8 aminoacids within HC between position 114 and 121. Here, we show that these mutations display reduced propensity for transmembrane topology. In addition, the mutants exhibited alterations in the formation of the C1 proteolytic fragment, which is generated by alpha-cleavage during normal PrP(C) metabolism, indicating that HC might function as recognition site for the protease(s) responsible for PrP(C) alpha-cleavage. Interestingly, the mutant G113V, corresponding to a hereditary form of prion disease in humans, displayed increased (Ctm)PrP topology and decreased alpha-cleavage in our in vitro assay. In conclusion, HC represents an essential determinant for transmembrane PrP topology, whereas the high evolutionary conservation of this region is rather based upon preservation of PrP(C) alpha-cleavage, thus highlighting the biological importance of this cleavage.


Subject(s)
Cell Membrane/metabolism , PrPC Proteins/metabolism , Amino Acid Sequence , Animals , Hydrophobic and Hydrophilic Interactions , Mice , Mice, Transgenic , Molecular Sequence Data , PrPC Proteins/genetics , Protein Structure, Tertiary/genetics , Sequence Deletion
SELECTION OF CITATIONS
SEARCH DETAIL