Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
J Virol ; 97(11): e0091023, 2023 Nov 30.
Article in English | MEDLINE | ID: mdl-37921471

ABSTRACT

IMPORTANCE: The main limitation of oncolytic vectors is neutralization by blood components, which prevents intratumoral administration to patients. Enadenotucirev, a chimeric HAdV-11p/HAdV-3 adenovirus identified by bio-selection, is a low seroprevalence vector active against a broad range of human carcinoma cell lines. At this stage, there's still some uncertainty about tropism and primary receptor utilization by HAdV-11. However, this information is very important, as it has a direct influence on the effectiveness of HAdV-11-based vectors. The aim of this work is to determine which of the two receptors, DSG2 and CD46, is involved in the attachment of the virus to the host, and what role they play in the early stages of infection.


Subject(s)
Adenoviruses, Human , Desmoglein 2 , Membrane Cofactor Protein , Receptors, Virus , Humans , Adenoviruses, Human/genetics , Adenoviruses, Human/metabolism , Cell Line , Desmoglein 2/genetics , Desmoglein 2/metabolism , Membrane Cofactor Protein/genetics , Membrane Cofactor Protein/metabolism , Receptors, Virus/genetics , Receptors, Virus/metabolism
2.
Mol Ther ; 30(5): 1913-1925, 2022 05 04.
Article in English | MEDLINE | ID: mdl-35151843

ABSTRACT

Virus-like particles (VLPs) are highly suited platforms for protein-based vaccines. In the present work, we adapted a previously designed non-infectious adenovirus-inspired 60-mer dodecahedric VLP (ADDomer) to display a multimeric array of large antigens through a SpyTag/SpyCatcher system. To validate the platform as a potential COVID-19 vaccine approach, we decorated the newly designed VLP with the glycosylated receptor binding domain (RBD) of SARS-CoV-2. Cryoelectron microscopy structure revealed that up to 60 copies of this antigenic domain could be bound on a single ADDomer particle, with the symmetrical arrangements of a dodecahedron. Mouse immunization with the RBD decorated VLPs already showed a significant specific humoral response following prime vaccination, greatly reinforced by a single boost. Neutralization assays with SARS-CoV-2 spike pseudo-typed virus demonstrated the elicitation of strong neutralization titers, superior to those of COVID-19 convalescent patients. Notably, the presence of pre-existing immunity against the adenoviral-derived particles did not hamper the immune response against the antigen displayed on its surface. This plug and play vaccine platform represents a promising new highly versatile tool to combat emergent pathogens.


Subject(s)
COVID-19 , SARS-CoV-2 , Adenoviridae/genetics , Animals , Antibodies, Neutralizing , Antibodies, Viral , COVID-19/prevention & control , COVID-19 Vaccines , Cryoelectron Microscopy , Humans , Mice , Vaccination
3.
Viruses ; 12(10)2020 09 25.
Article in English | MEDLINE | ID: mdl-32992715

ABSTRACT

The study of viruses causing acute respiratory distress syndromes (ARDS) is more essential than ever at a time when a virus can create a global pandemic in a matter of weeks. Among human adenoviruses, adenovirus of serotype 7 (HAdV7) is one of the most virulent serotypes. This virus regularly re-emerges in Asia and has just been the cause of several deaths in the United States. A critical step of the virus life cycle is the attachment of the knob domain of the fiber (HAd7K) to the cellular receptor desmoglein-2 (DSG2). Complexes between the fiber knob and two extracellular domains of DSG2 have been produced. Their characterization by biochemical and biophysical methods show that these two domains are sufficient for the interaction and that the trimeric HAd7K could accommodate up to three DSG2 receptor molecules. The cryo-electron microscopy (cryo-EM) structure of these complexes at 3.1 Å resolution confirmed the biochemical data, and allowed the identification of the critical amino acid residues for this interaction, which shows similarities with other DSG2 interacting adenoviruses, despite a low homology in the primary sequences.


Subject(s)
Adenoviruses, Human/metabolism , Capsid Proteins/metabolism , Desmoglein 2/metabolism , Respiratory Distress Syndrome/virology , Adenoviridae Infections/virology , Adenoviruses, Human/pathogenicity , Capsid Proteins/chemistry , Capsid Proteins/genetics , Cryoelectron Microscopy , Desmoglein 2/chemistry , HEK293 Cells , Host-Pathogen Interactions , Humans , Models, Molecular , Mutation , Protein Binding , Protein Conformation , Receptors, Virus/chemistry , Receptors, Virus/metabolism , Serogroup
4.
Viruses ; 12(8)2020 07 30.
Article in English | MEDLINE | ID: mdl-32751671

ABSTRACT

Influenza viruses are negative single-stranded RNA viruses with nuclear transcription and replication. They enter the nucleus by using the cellular importin-α/-ß nuclear import machinery. Influenza nucleoproteins from influenza A, B, C and D viruses possess a nuclear localization signal (NLS) localized on an intrinsically disordered extremity (NPTAIL). In this paper, using size exclusion chromatography (SEC), SEC-multi-angle laser light scattering (SEC-MALLS) analysis, surface plasmon resonance (SPR) and fluorescence anisotropy, we provide the first comparative study designed to dissect the interaction between the four NPTAILs and four importins-α identified as partners. All interactions between NPTAILs and importins-α have high association and dissociation rates and present a distinct and specific behaviour. D/NPTAIL interacts strongly with all importins-α while B/NPTAIL shows weak affinity for importins-α. A/NPTAIL and C/NPTAIL present preferential importin-α partners. Mutations in B/NPTAIL and D/NPTAIL show a loss of importin-α binding, confirming key NLS residues. Taken together, our results provide essential highlights of this complex translocation mechanism.


Subject(s)
Host Microbial Interactions , Nucleocapsid Proteins/metabolism , Orthomyxoviridae/metabolism , alpha Karyopherins/metabolism , Active Transport, Cell Nucleus , Cell Nucleus/metabolism , Fluorescence Polarization , Humans , Mutation , Nuclear Localization Signals , Nucleocapsid Proteins/genetics , Orthomyxoviridae/genetics , Protein Binding , Surface Plasmon Resonance
5.
Viruses ; 12(7)2020 07 02.
Article in English | MEDLINE | ID: mdl-32630840

ABSTRACT

Many geometric forms are found in nature, some of them adhering to mathematical laws or amazing aesthetic rules. One of the best-known examples in microbiology is the icosahedral shape of certain viruses with 20 triangular facets and 12 edges. What is less known, however, is that a complementary object displaying 12 faces and 20 edges called a 'dodecahedron' can be produced in huge amounts during certain adenovirus replication cycles. The decahedron was first described more than 50 years ago in the human adenovirus (HAdV3) viral cycle. Later on, the expression of this recombinant scaffold, combined with improvements in cryo-electron microscopy, made it possible to decipher the structural determinants underlying their architecture. Recently, this particle, which mimics viral entry, was used to fish the long elusive adenovirus receptor, desmoglein-2, which serves as a cellular docking for some adenovirus serotypes. This breakthrough enabled the understanding of the physiological role played by the dodecahedral particles, showing that icosahedral and dodecahedral particles live more than a simple platonic story. All these points are developed in this review, and the potential use of the dodecahedron in therapeutic development is discussed.


Subject(s)
Adenoviridae/physiology , Capsid/physiology , Adenoviridae Infections/pathology , Animals , Capsid Proteins/physiology , Cryoelectron Microscopy , Humans , Virus Replication/physiology
6.
Acta Crystallogr F Struct Biol Commun ; 75(Pt 12): 750-757, 2019 Dec 01.
Article in English | MEDLINE | ID: mdl-31797817

ABSTRACT

The cryo-electron microscopy (cryo-EM) structure of the complex between the trimeric human adenovirus B serotype 3 fibre knob and human desmoglein 2 fragments containing cadherin domains EC2 and EC3 has been published, showing 3:1 and 3:2 complexes. Here, the crystal structure determined at 4.5 Šresolution is presented with one EC2-EC3 desmoglein fragment bound per fibre knob monomer in the asymmetric unit, leading to an apparent 3:3 stoichiometry. However, in concentrated solution the 3:2 complex is predominant, as shown by small-angle X-ray scattering (SAXS), while cryo-EM at lower concentrations showed a majority of the 3:1 complex. Substitution of the calcium ions bound to the desmoglein domains by terbium ions allowed confirmation of the X-ray model using their anomalous scattering and shows that at least one binding site per cluster of calcium ions is intact and exchangeable and, combined with SAXS data, that the cadherin domains are folded even in the distal part that is invisible in the cryo-EM reconstruction.


Subject(s)
Adenoviruses, Human/metabolism , Cadherins/metabolism , Capsid Proteins/chemistry , Capsid Proteins/metabolism , Desmoglein 2/chemistry , Desmoglein 2/metabolism , Adenoviruses, Human/classification , Amino Acid Sequence , Cadherins/chemistry , Crystallization , Crystallography, X-Ray , Humans , Models, Molecular , Protein Binding , Protein Conformation , Protein Domains , Serogroup
7.
Sci Rep ; 9(1): 6169, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30992466

ABSTRACT

Disorganized intercellular junctions are critical for maintaining the integrity of solid epithelial tumors and prevent the infiltration of oncological therapies into the bulk of the malignancy. We have developed small, recombinant proteins which bind a critical junction protein, desmoglein 2, triggering the transient and specific opening of tumor tight junctions allowing for infiltration of the tumor with immune cells, oncolytic viruses, drugs, and other therapeutics. Our new molecule, JOC-x, is a promising candidate for a new class of tumor-targeting agents that accumulate both around and within tumors and remodel the tumor microenvironment. Native cysteines were removed from the parental protein, JO-4, followed by addition of a single cysteine to allow for convenient attachment of various payloads that can be targeted directly to the tumor. Our tumor-targeting protein exhibits high avidity, minimal aggregation, and is easily purified at good yields from E. coli. For proof of concept, we demonstrate effective conjugation to biotin as a model for flexible co-targeting, addition of metal ion chelators as models for imaging and radiotherapy, and linkage of the TLR3 agonist poly(I:C) as a model immune-oncologic agent. This second-generation cancer co-therapeutic protein is optimized for activity and primed for cGMP manufacture in preparation for upcoming clinical studies.


Subject(s)
Adenoviridae/metabolism , Capsid Proteins/metabolism , Desmoglein 2/metabolism , Neoplasms/therapy , Tight Junctions/metabolism , Adenoviridae/chemistry , Amino Acid Sequence , Capsid Proteins/chemistry , Drug Carriers/chemistry , Drug Carriers/metabolism , Drug Delivery Systems , HeLa Cells , Humans , Models, Molecular , Neoplasms/metabolism , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
8.
Nat Commun ; 10(1): 1181, 2019 03 12.
Article in English | MEDLINE | ID: mdl-30862836

ABSTRACT

Attachment of human adenovirus (HAd) to the host cell is a critical step of infection. Initial attachment occurs via the adenoviral fibre knob protein and a cellular receptor. Here we report the cryo-electron microscopy (cryo-EM) structure of a <100 kDa non-symmetrical complex comprising the trimeric HAd type 3 fibre knob (HAd3K) and human desmoglein 2 (DSG2). The structure reveals a unique stoichiometry of 1:1 and 2:1 (DSG2: knob trimer) not previously observed for other HAd-receptor complexes. We demonstrate that mutating Asp261 in the fibre knob is sufficient to totally abolish receptor binding. These data shed new light on adenovirus infection strategies and provide insights for adenoviral vector development and structure-based design.


Subject(s)
Adenoviruses, Human/metabolism , Capsid Proteins/metabolism , Desmoglein 2/metabolism , Receptors, Virus/metabolism , Virus Attachment , Adenoviridae Infections/pathology , Adenoviridae Infections/virology , Adenoviruses, Human/pathogenicity , Asparagine/genetics , Capsid Proteins/ultrastructure , Cryoelectron Microscopy , Desmoglein 2/ultrastructure , HEK293 Cells , Humans , Models, Molecular , Protein Domains , Receptors, Virus/ultrastructure , Recombinant Proteins/metabolism , Recombinant Proteins/ultrastructure
9.
Sci Rep ; 8(1): 8381, 2018 05 30.
Article in English | MEDLINE | ID: mdl-29849084

ABSTRACT

High-affinity binding of the trimeric fibre protein to a cell surface primary receptor is a common feature shared by all adenovirus serotypes. Recently, a long elusive species B adenovirus receptor has been identified. Desmoglein 2 (DSG2) a component of desmosomal junction, has been reported to interact at high affinity with Human adenoviruses HAd3, HAd7, HAd11 and HAd14. Little is known with respect to the molecular interactions of adenovirus fibre with the DSG2 ectodomain. By using different DSG2 ectodomain constructs and biochemical and biophysical experiments, we report that the third extracellular cadherin domain (EC3) of DSG2 is critical for HAd3 fibre binding. Unexpectedly, stoichiometry studies using multi-angle laser light scattering (MALLS) and analytical ultra-centrifugation (AUC) revealed a non-classical 1:1 interaction (one DSG2 per trimeric fibre), thus differentiating 'DSG2-interacting' adenoviruses from other protein receptor interacting adenoviruses in their infection strategy.


Subject(s)
Adenoviridae/metabolism , Desmoglein 2/metabolism , Serogroup , Adenoviridae/genetics , Desmoglein 2/chemistry , Glycosylation , Humans , Protein Binding , Protein Domains
10.
Curr Cancer Drug Targets ; 15(1): 2-13, 2015.
Article in English | MEDLINE | ID: mdl-25543663

ABSTRACT

Microtubule drugs have been widely used in cancer chemotherapies. Although microtubules are subject to regulation by signal transduction mechanisms, their pharmacological modulation has so far relied on compounds that bind to the tubulin subunit. Using a cell-based assay designed to probe the microtubule polymerization status, we identified two pharmacophores, CM09 and CM10, as cell-permeable microtubule stabilizing agents. These synthetic compounds do not affect the assembly state of purified microtubules in vitro but they profoundly suppress microtubule dynamics in vivo. Moreover, they exert cytotoxic effects on several cancer cell lines including multidrug resistant cell lines. Therefore, these classes of compounds represent novel attractive leads for cancer chemotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Cycle/drug effects , Drug Design , HeLa Cells/drug effects , Microtubules/drug effects , Tubulin Modulators/pharmacology , Tubulin/drug effects , Cell Culture Techniques , Cell Survival , Fluorescent Antibody Technique , Humans , Microtubules/physiology
11.
FEBS Lett ; 588(24): 4694-700, 2014 Dec 20.
Article in English | MEDLINE | ID: mdl-25447524

ABSTRACT

Ficolin-2 has been reported to bind to DNA and heparin, but the mechanism involved has not been thoroughly investigated. X-ray studies of the ficolin-2 fibrinogen-like domain in complex with several new ligands now show that sulfate and phosphate groups are prone to bind to the S3 binding site of the protein. Composed of Arg132, Asp133, Thr136 and Lys221, the S3 site was previously shown to mainly bind N-acetyl groups. Furthermore, DNA and heparin compete for binding to ficolin-2. Mutagenesis studies reveal that Arg132, and to a lesser extent Asp133, are important for this binding property. The versatility of the S3 site in binding N-acetyl, sulfate and phosphate groups is discussed through comparisons with homologous fibrinogen-like recognition proteins.


Subject(s)
Carbohydrate Metabolism , Lectins/metabolism , Phosphates/metabolism , Sulfates/metabolism , Amino Acid Sequence , Animals , Binding Sites , Humans , Lectins/chemistry , Lectins/genetics , Mice , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Binding , Protein Structure, Tertiary , Ficolins
12.
J Immunol ; 193(11): 5699-708, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25344472

ABSTRACT

Human L-ficolin is a soluble protein of the innate immune system able to sense pathogens through its fibrinogen (FBG) recognition domains and to trigger activation of the lectin complement pathway through associated serine proteases. L-Ficolin has been previously shown to recognize pneumococcal clinical isolates, but its ligands and especially its molecular specificity remain to be identified. Using solid-phase binding assays, serum and recombinant L-ficolins were shown to interact with serotype 2 pneumococcal strain D39 and its unencapsulated R6 derivative. Incubation of both strains with serum triggered complement activation, as measured by C4b and C3b deposition, which was decreased by using ficolin-depleted serum. Recombinant L-ficolin and its FBG-like recognition domain bound to isolated pneumococcal cell wall extracts, whereas binding to cell walls depleted of teichoic acid (TA) was decreased. Both proteins were also shown to interact with two synthetic TA compounds, each comprising part structures of the complete lipoteichoic acid molecule with two PCho residues. Competition studies and direct interaction measurements by surface plasmon resonance identified PCho as a novel L-ficolin ligand. Structural analysis of complexes of the FBG domain of L-ficolin and PCho revealed that the phosphate moiety interacts with amino acids previously shown to define an acetyl binding site. Consequently, binding of L-ficolin to immobilized acetylated BSA was inhibited by PCho and synthetic TA. Binding of serum L-ficolin to immobilized synthetic TA and PCho-conjugated BSA triggered activation of the lectin complement pathway, thus further supporting the hypothesis of L-ficolin involvement in host antipneumococcal defense.


Subject(s)
Lectins/metabolism , Pneumococcal Infections/immunology , Streptococcus pneumoniae/metabolism , Teichoic Acids/metabolism , Acetylation , Cell Wall/metabolism , Complement Activation , Complement C3b/metabolism , Complement C4b/metabolism , Fibrinogen/genetics , Host-Pathogen Interactions , Humans , Immunity, Innate , Lectins/genetics , Phosphorylcholine/chemistry , Protein Binding , Protein Structure, Tertiary/genetics , Streptococcus pneumoniae/immunology , Surface Plasmon Resonance , Teichoic Acids/chemistry , Ficolins
13.
Br J Pharmacol ; 168(3): 673-85, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23004938

ABSTRACT

BACKGROUND AND PURPOSE: Drugs targeting microtubules are commonly used for cancer treatment. However, the potency of microtubule inhibitors used clinically is limited by the emergence of resistance. We thus designed a strategy to find new cell-permeable microtubule-targeting agents. EXPERIMENTAL APPROACH: Using a cell-based assay designed to probe for microtubule polymerization status, we screened a chemical library and identified two azaindole derivatives, CM01 and CM02, as cell-permeable microtubule-depolymerizing agents. The mechanism of the anti-tumour effects of these two compounds was further investigated both in vivo and in vitro. KEY RESULTS: CM01 and CM02 induced G2/M cell cycle arrest and exerted potent cytostatic effects on several cancer cell lines including multidrug-resistant (MDR) cell lines. In vitro experiments revealed that the azaindole derivatives inhibited tubulin polymerization and competed with colchicines for this effect, strongly indicating that tubulin is the cellular target of these azaindole derivatives. In vivo experiments, using a chicken chorioallantoic xenograft tumour assay, established that these compounds exert a potent anti-tumour effect. Furthermore, an assay probing the growth of vessels out of endothelial cell spheroids showed that CM01 and CM02 exert anti-angiogenic activities. CONCLUSIONS AND IMPLICATIONS: CM01 and CM02 are reversible microtubule-depolymerizing agents that exert potent cytostatic effects on human cancer cells of diverse origins, including MDR cells. They were also shown to inhibit angiogenesis and tumour growth in chorioallantoic breast cancer xenografts. Hence, these azaindole derivatives are attractive candidates for further preclinical investigations.


Subject(s)
Antineoplastic Agents/pharmacology , Indoles/pharmacology , Tubulin Modulators/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cells, Cultured , Chick Embryo , Chorioallantoic Membrane/pathology , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Endothelial Cells/drug effects , Endothelial Cells/physiology , Humans , Indoles/therapeutic use , Neoplasms/drug therapy , Neoplasms/pathology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Tubulin Modulators/therapeutic use , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
14.
PLoS One ; 7(11): e50722, 2012.
Article in English | MEDLINE | ID: mdl-23226363

ABSTRACT

Langerin is a C-type lectin specifically expressed in Langerhans cells. As recently shown for HIV, Langerin is thought to capture pathogens and mediate their internalisation into Birbeck Granules for elimination. However, the precise functions of Langerin remain elusive, mostly because of the lack of information on its binding properties and physiological ligands. Based on recent reports that Langerin binds to sulfated sugars, we conducted here a comparative analysis of Langerin interaction with mannose-rich HIV glycoprotein gp120 and glycosaminoglycan (GAGs), a family of sulfated polysaccharides expressed at the surface of most mammalian cells. Our results first revealed that Langerin bound to these different glycans through very distinct mechanisms and led to the identification of a novel, GAG-specific binding mode within Langerin. In contrast to the canonical lectin domain, this new binding site showed no Ca(2+)-dependency, and could only be detected in entire, trimeric extracellular domains of Langerin. Interestingly binding to GAGs, did not simply rely on a net charge effect, but rather on more discrete saccharide features, such as 6-O-sulfation, or iduronic acid content. Using molecular modelling simulations, we proposed a model of Langerin/heparin complex, which located the GAG binding site at the interface of two of the three Carbohydrate-recognition domains of the protein, at the edge of the a-helix coiled-coil. To our knowledge, the binding properties that we have highlighted here for Langerin, have never been reported for C-type lectins before. These findings provide new insights towards the understanding of Langerin biological functions.


Subject(s)
Antigens, CD/chemistry , Antigens, CD/metabolism , Calcium/metabolism , Glycosaminoglycans/metabolism , HIV Envelope Protein gp120/metabolism , Lectins, C-Type/chemistry , Lectins, C-Type/metabolism , Mannose-Binding Lectins/chemistry , Mannose-Binding Lectins/metabolism , Epidermal Cells , Glycosaminoglycans/chemistry , HIV Envelope Protein gp120/chemistry , Heparin/metabolism , Humans , Langerhans Cells/metabolism , Models, Molecular , Molecular Docking Simulation , Mucous Membrane/cytology , Protein Binding , Protein Structure, Tertiary , Software
15.
PLoS One ; 7(10): e47933, 2012.
Article in English | MEDLINE | ID: mdl-23110134

ABSTRACT

Proteoglycans (PGs) are critically involved in major cellular processes. Most PG activities are due to the large interactive properties of their glycosaminoglycan (GAG) polysaccharide chains, whose expression and fine structural features are tightly controlled by a complex and highly regulated biosynthesis machinery. Xylosides are known to bypass PG-associated GAG biosynthesis and prime the assembly of free polysaccharide chains. These are, therefore, attractive molecules to interfere with GAG expression and function. Recently, we have developed a new xyloside derivative, C-Xyloside, that shares classical GAG-inducing xyloside activities while exhibiting improved metabolic stability. We have previously shown that C-Xyloside had beneficial effects on skin homoeostasis/regeneration using a number of models, but its precise effects on GAG expression and fine structure remained to be addressed. In this study, we have therefore investigated this in details, using a reconstructed dermal tissue as model. Our results first confirmed that C-Xyloside strongly enhanced synthesis of GAG chains, but also induced significant changes in their structure. C-Xyloside primed GAGs were exclusively chondroitin/dermatan sulfate (CS/DS) that featured reduced chain size, increased O-sulfation, and changes in iduronate content and distribution. Surprisingly, C-Xyloside also affected PG-borne GAGs, the main difference being observed in CS/DS 4-O/6-O-sulfation ratio. Such changes were found to affect the biological properties of CS/DS, as revealed by the significant reduction in binding to Hepatocyte Growth Factor observed upon C-Xyloside treatment. Overall, this study provides new insights into the effect of C-Xyloside on GAG structure and activities, which opens up perspectives and applications of such compound in skin repair/regeneration. It also provides a new illustration about the use of xylosides as tools for modifying GAG fine structure/function relationships.


Subject(s)
Dermis/metabolism , Gene Expression Regulation/drug effects , Glycosaminoglycans/metabolism , Glycosides/pharmacology , Chondroitin , Chromatography, Gel , Dermatan Sulfate , Glycosaminoglycans/biosynthesis , Glycosaminoglycans/isolation & purification , Glycosides/metabolism , Hepatocyte Growth Factor/metabolism , Humans , Scintillation Counting , Tritium
16.
Cancer Res ; 72(17): 4429-39, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22761334

ABSTRACT

The emergence of tumor resistance to conventional microtubule-targeting drugs restricts their clinical use. Using a cell-based assay that recognizes microtubule polymerization status to screen for chemicals that interact with regulators of microtubule dynamics, we identified Pyr1, a cell permeable inhibitor of LIM kinase, which is the enzyme that phosphorylates and inactivates the actin-depolymerizing factor cofilin. Pyr1 reversibly stabilized microtubules, blocked actin microfilament dynamics, inhibited cell motility in vitro and showed anticancer properties in vivo, in the absence of major side effects. Pyr1 inhibition of LIM kinase caused a microtubule-stabilizing effect, which was independent of any direct effects on the actin cytoskeleton. In addition, Pyr1 retained its activity in multidrug-resistant cancer cells that were resistant to conventional microtubule-targeting agents. Our findings suggest that LIM kinase functions as a signaling node that controls both actin and microtubule dynamics. LIM kinase may therefore represent a targetable enzyme for cancer treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Lim Kinases/antagonists & inhibitors , Microtubules/metabolism , Neoplasms/metabolism , Protein Kinase Inhibitors/pharmacology , Tubulin Modulators/pharmacology , Actins/metabolism , Animals , Antineoplastic Agents/administration & dosage , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Resistance, Neoplasm , Female , HeLa Cells , Humans , Mice , Neoplasms/drug therapy , Neoplasms/mortality , Phenotype , Protein Kinase Inhibitors/administration & dosage , Protein Stability/drug effects , Tubulin/metabolism , Tubulin Modulators/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...