31 results on '"Greg L. Beilhartz"'
Search Results
2. An enhanced intracellular delivery platform based on a distant diphtheria toxin homolog that evades pre-existing antitoxin antibodies
- Author
-
Shivneet K Gill, Seiji N Sugiman-Marangos, Greg L Beilhartz, Elizabeth Mei, Mikko Taipale, and Roman A Melnyk
- Subjects
Immunotoxin ,Intracellular Delivery ,Diphtheria Toxin ,Chelona Toxin ,Antidrug Antibodies ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract Targeted intracellular delivery of therapeutic proteins remains a significant unmet challenge in biotechnology. A promising approach is to leverage the intrinsic capabilities of bacterial toxins like diphtheria toxin (DT) to deliver a potent cytotoxic enzyme into cells with an associated membrane translocation moiety. Despite showing promising clinical efficacy, widespread deployment of DT-based therapeutics is complicated by the prevalence of pre-existing antibodies in the general population arising from childhood DT toxoid vaccinations, which impact the exposure, efficacy, and safety of these potent molecules. Here, we describe the discovery and characterization of a distant DT homolog from the ancient reptile pathogen Austwickia chelonae that we have dubbed chelona toxin (ACT). We show that ACT is comparable to DT structure and function in all respects except that it is not recognized by pre-existing anti-DT antibodies circulating in human sera. Furthermore, we demonstrate that ACT delivers heterologous therapeutic cargos into target cells more efficiently than DT. Our findings highlight ACT as a promising new chassis for building next-generation immunotoxins and targeted delivery platforms with improved pharmacokinetic and pharmacodynamic properties.
- Published
- 2024
- Full Text
- View/download PDF
3. HIV-1 Ribonuclease H: Structure, Catalytic Mechanism and Inhibitors
- Author
-
Greg L. Beilhartz and Matthias Götte
- Subjects
HIV ,reverse transcriptase ,RNase H ,inhibitors ,drug resistance ,Microbiology ,QR1-502 - Abstract
Since the human immunodeficiency virus (HIV) was discovered as the etiological agent of acquired immunodeficiency syndrome (AIDS), it has encouraged much research into antiviral compounds. The reverse transcriptase (RT) of HIV has been a main target for antiviral drugs. However, all drugs developed so far inhibit the polymerase function of the enzyme, while none of the approved antiviral agents inhibit specifically the necessary ribonuclease H (RNase H) function of RT. This review provides a background on structure-function relationships of HIV-1 RNase H, as well as an outline of current attempts to develop novel, potent chemotherapeutics against a difficult drug target.
- Published
- 2010
- Full Text
- View/download PDF
4. An engineered chimeric toxin that cleaves activated mutant and wild-type RAS inhibits tumor growth
- Author
-
Marco Biancucci, David Gius, Minyoung Park, Matthew B. Kieffer, Karla J. F. Satchell, Vania Vidimar, Greg L. Beilhartz, and Roman A. Melnyk
- Subjects
Male ,0301 basic medicine ,Mutant ,Mice, Nude ,Antineoplastic Agents ,Protein Sorting Signals ,Chimeric toxin ,Mice ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,Endopeptidases ,Animals ,Humans ,Diphtheria Toxin ,Tumor growth ,Cells, Cultured ,Diphtheria toxin ,Multidisciplinary ,Chemistry ,Wild type ,rap1 GTP-Binding Proteins ,Neoplasms, Experimental ,Biological Sciences ,HCT116 Cells ,Recombinant Proteins ,Endopeptidase ,3. Good health ,Cell biology ,030104 developmental biology ,030220 oncology & carcinogenesis ,Mutation ,Proteolysis ,ras Proteins ,Female ,Intracellular - Abstract
Significance RAS oncoproteins have long been considered among the most elusive drug targets in cancer research. At issue is the lack of accessible drug-binding sites and the extreme affinity for GTP. Covalent inhibitors against the KRAS G12C mutant have shown early clinical promise; however, targeting the other oncogenic RAS mutants across three RAS isoforms has proven challenging. Inhibition of activated wild-type RAS in the absence of canonical RAS mutations is also highly desirable in certain tumors. Here, we demonstrate delivery of an extremely potent pan-RAS and RAP1-cleaving enzyme in therapeutic quantities to specific receptor-bearing cells in vitro and in vivo. We aim to advance this approach to engineer the first targeted pan-RAS inhibitor for cancer therapy.
- Published
- 2020
- Full Text
- View/download PDF
5. Exploiting the diphtheria toxin internalization receptor enhances delivery of proteins to lysosomes for enzyme replacement therapy
- Author
-
Seiji Sugiman-Marangos, Dongxia Zhou, Greg L. Beilhartz, Berge A. Minassian, Rong Hua, Xiaochu Zhao, Peter K. Kim, Roman A. Melnyk, and James M. Rini
- Subjects
media_common.quotation_subject ,Diseases and Disorders ,complex mixtures ,Receptor, IGF Type 2 ,law.invention ,Mice ,03 medical and health sciences ,0302 clinical medicine ,law ,Hydrolase ,Lysosomal storage disease ,medicine ,Animals ,Diphtheria Toxin ,Enzyme Replacement Therapy ,Receptor ,Internalization ,Research Articles ,030304 developmental biology ,media_common ,Diphtheria toxin ,chemistry.chemical_classification ,0303 health sciences ,Multidisciplinary ,Chemistry ,SciAdv r-articles ,Brain ,Cell Biology ,Enzyme replacement therapy ,medicine.disease ,Recombinant Proteins ,Cell biology ,Enzyme ,Recombinant DNA ,Lysosomes ,030217 neurology & neurosurgery ,Research Article - Abstract
Engineering lysosomal enzymes to bind the diphtheria toxin receptor with high affinity increases uptake into cells and tissues., Enzyme replacement therapy, in which a functional copy of an enzyme is injected either systemically or directly into the brain of affected individuals, has proven to be an effective strategy for treating certain lysosomal storage diseases. The inefficient uptake of recombinant enzymes via the mannose-6-phosphate receptor, however, prohibits the broad utility of replacement therapy. Here, to improve the efficiency and efficacy of lysosomal enzyme uptake, we exploited the strategy used by diphtheria toxin to enter into the endolysosomal network of cells by creating a chimera between the receptor-binding fragment of diphtheria toxin and the lysosomal hydrolase TPP1. We show that chimeric TPP1 binds with high affinity to target cells and is efficiently delivered into lysosomes. Further, we show superior uptake of chimeric TPP1 over TPP1 alone in brain tissue following intracerebroventricular injection in mice lacking TPP1, demonstrating the potential of this strategy for enhancing lysosomal storage disease therapy.
- Published
- 2020
- Full Text
- View/download PDF
6. Recognition of Semaphorin Proteins by P. sordellii Lethal Toxin Reveals Principles of Receptor Specificity in Clostridial Toxins
- Author
-
Daniel Schramek, Hunsang Lee, Greg L. Beilhartz, Mikko Taipale, Roman A. Melnyk, Hong Cui, Swetha Raman, Jean-Philippe Julien, John L. Rubinstein, Iga Kucharska, Huazhu Liang, and Mandy H. Y. Lam
- Subjects
bacterial exotoxins ,Frizzled ,Bacterial Toxins ,Exotoxins ,Semaphorins ,Biology ,Clostridial toxins ,semaphorin ,General Biochemistry, Genetics and Molecular Biology ,Article ,CRISPR/Cas9 screening ,law.invention ,Microbiology ,03 medical and health sciences ,0302 clinical medicine ,SEMA6B ,Semaphorin ,SEMA6A ,law ,medicine ,P. sordellii ,CRISPR ,Secretion ,Receptor ,Binding selectivity ,030304 developmental biology ,0303 health sciences ,Toxic shock syndrome ,TcsL ,medicine.disease ,Recombinant DNA ,cryo-EM ,030217 neurology & neurosurgery - Abstract
Summary Pathogenic clostridial species secrete potent toxins that induce severe host tissue damage. Paeniclostridium sordellii lethal toxin (TcsL) causes an almost invariably lethal toxic shock syndrome associated with gynecological infections. TcsL is 87% similar to C. difficile TcdB, which enters host cells via Frizzled receptors in colon epithelium. However, P. sordellii infections target vascular endothelium, suggesting that TcsL exploits another receptor. Here, using CRISPR/Cas9 screening, we establish semaphorins SEMA6A and SEMA6B as TcsL receptors. We demonstrate that recombinant SEMA6A can protect mice from TcsL-induced edema. A 3.3 Å cryo-EM structure shows that TcsL binds SEMA6A with the same region that in TcdB binds structurally unrelated Frizzled. Remarkably, 15 mutations in this evolutionarily divergent surface are sufficient to switch binding specificity of TcsL to that of TcdB. Our findings establish semaphorins as physiologically relevant receptors for TcsL and reveal the molecular basis for the difference in tissue targeting and disease pathogenesis between highly related toxins., Graphical Abstract, Highlights • CRISPR screen identifies SEMA6A and SEMA6B as receptors for P. sordellii lethal toxin TcsL • Soluble SEMA6A ectodomain protects mouse lungs from TcsL-induced edema • 3.3 Å cryo-EM structure of TcsL bound to SEMA6A reveals atomic details of the interaction • 15 mutations in the TcsL receptor-binding interface rewire receptor specificity, A lethal bacterial toxin that plays a major role in toxic shock syndrome uses the host semaphorin proteins as receptors, and structural analysis shows a small binding interface that can be mutated to switch specificity between very different host receptors.
- Published
- 2020
7. bioPROTACs as versatile modulators of intracellular therapeutic targets including proliferating cell nuclear antigen (PCNA)
- Author
-
Jinkai Teo, Greg L. Beilhartz, Khong Ming Peh, Simon Ng, Charles W. Johannes, Regina Khoo, Shih Chieh Chang, Brian Henry, Roman A. Melnyk, Anthony W. Partridge, Shuhui Lim, David P. Lane, and Christopher J. Brown
- Subjects
0301 basic medicine ,Proteolysis ,Ubiquitin-Protein Ligases ,Cell cycle progression ,Protein Engineering ,01 natural sciences ,03 medical and health sciences ,Proliferating Cell Nuclear Antigen ,medicine ,Humans ,Molecular Targeted Therapy ,Gene ,Multidisciplinary ,Binding Sites ,medicine.diagnostic_test ,biology ,DNA synthesis ,010405 organic chemistry ,Chemistry ,Intracellular protein ,Biological Sciences ,Recombinant Proteins ,0104 chemical sciences ,Ubiquitin ligase ,Proliferating cell nuclear antigen ,Cell biology ,030104 developmental biology ,HEK293 Cells ,biology.protein ,Intracellular ,Protein Binding - Abstract
Targeted degradation approaches such as proteolysis targeting chimeras (PROTACs) offer new ways to address disease through tackling challenging targets and with greater potency, efficacy, and specificity over traditional approaches. However, identification of high-affinity ligands to serve as PROTAC starting points remains challenging. As a complementary approach, we describe a class of molecules termed biological PROTACs (bioPROTACs)-engineered intracellular proteins consisting of a target-binding domain directly fused to an E3 ubiquitin ligase. Using GFP-tagged proteins as model substrates, we show that there is considerable flexibility in both the choice of substrate binders (binding positions, scaffold-class) and the E3 ligases. We then identified a highly effective bioPROTAC against an oncology target, proliferating cell nuclear antigen (PCNA) to elicit rapid and robust PCNA degradation and associated effects on DNA synthesis and cell cycle progression. Overall, bioPROTACs are powerful tools for interrogating degradation approaches, target biology, and potentially for making therapeutic impacts.
- Published
- 2020
8. Repurposing bacterial toxins for intracellular delivery of therapeutic proteins
- Author
-
Greg L. Beilhartz, Roman A. Melnyk, and Seiji Sugiman-Marangos
- Subjects
0301 basic medicine ,Endosome ,Bacterial Toxins ,Receptors, Cell Surface ,Computational biology ,Pharmacology ,Biology ,Biochemistry ,03 medical and health sciences ,Cytosol ,0302 clinical medicine ,In vivo ,Drug Discovery ,Animals ,Humans ,Deimmunization ,Repurposing ,Drug Carriers ,Microbial toxins ,Drug discovery ,Recombinant Proteins ,030104 developmental biology ,Pharmaceutical Preparations ,030220 oncology & carcinogenesis ,Intracellular ,Protein Binding - Abstract
Despite enormous efforts, achieving efficacious levels of proteins inside mammalian cells remains one of the greatest challenges in biologics-based drug discovery and development. The inability of proteins to readily cross biological membranes precludes access to the wealth of intracellular targets and applications that lie within mammalian cells. Existing methods of delivery commonly suffer from an inability to target specific cells and tissues, poor endosomal escape, and limited in vivo efficacy. The aim of the present commentary is to highlight the potential of certain classes of bacterial toxins, which naturally deliver a large protein into the cytosolic compartment of target cells after binding a host cell-surface receptor with high affinity, as robust protein delivery platforms. We review the progress made in recent years toward demonstrating the utility of these systems at delivering a wide variety of protein cargo, with special attention paid to three distinct toxin-based platforms. We contend that with recent advances in protein deimmunization strategies, bacterial toxins are poised to introduce biologics into the inner sanctum of cells and treat a wealth of heretofore untreatable diseases with a new generation of therapeutics.
- Published
- 2017
- Full Text
- View/download PDF
9. Exploiting toxin internalization receptors to enhance delivery of proteins to lysosomes for enzyme replacement therapy
- Author
-
Roman A. Melnyk, Seiji Sugiman-Marangos, Xiaochu Zhao, Greg L. Beilhartz, Rong Hua, Berge A. Minassian, James M. Rini, Dongxia Zhou, and Peter K. Kim
- Subjects
Diphtheria toxin ,Chemistry ,media_common.quotation_subject ,Enzyme replacement therapy ,law.invention ,Cell biology ,Chimera (genetics) ,law ,In vivo ,Hydrolase ,Recombinant DNA ,Receptor ,Internalization ,media_common - Abstract
Lysosomal storage diseases are a group of over 70 inherited genetic diseases caused by a defect or deficiency in a lysosomal protein. Enzyme replacement therapy, in which a functional copy of the defective enzyme is injected either systemically or directly into the brain of affected individuals, has proven to be an effective strategy for treating certain lysosomal storage diseases; however, the inefficient uptake of recombinant enzymes into cells and tissuesviathe low-affinity mannose-6-phosphate receptor prohibits broader utility of replacement therapy. Here, to improve the efficiency and efficacy of lysosomal enzyme uptake, we exploited the strategy used by diphtheria toxin to enter into the endo-lysosomal network of cells by creating a chimera between the receptor-binding fragment of diphtheria toxin and the lysosomal hydrolase TPP1. We show that the targeted TPP1 chimera binds with high affinity to target cells and is delivered into lysosomes with much greater efficiency than TPP1 alone. Further, we demonstrate efficient and durable uptake of the chimerain vivofollowing intracerebroventricular injection in mice lacking TPP1. Targeting the highly efficient diphtheria toxin internalization pathway represents a novel approach for improving the efficacy and utility of enzyme replacement therapy for treating lysosomal storage diseases.
- Published
- 2020
- Full Text
- View/download PDF
10. Inhibition of tumor growth by a novel engineered chimeric toxin that cleaves activated mutant and wild-type RAS
- Author
-
David Gius, Vania Vidimar, Greg L. Beilhartz, Karla J. F. Satchell, Marco Biancucci, Minyoung Park, Matthew B. Kieffer, and Roman A. Melnyk
- Subjects
Diphtheria toxin ,In vivo ,Chemistry ,Mutant ,Wild type ,medicine ,Rap1 ,KRAS ,Binding site ,medicine.disease_cause ,In vitro ,Cell biology - Abstract
SummaryDespite nearly four decades of effort, broad inhibition of oncogenic RAS using small molecule approaches has proven to be a major challenge. Here we describe the development of a novel pan-RAS biologic inhibitor comprised of the RAS-RAP1-specific endopeptidase fused to the protein delivery machinery of diphtheria toxin. We show that this engineered chimeric toxin irreversibly cleaves and inactivates intracellular RAS at low picomolar concentrations terminating downstream signaling in receptor-bearing cells. Further, we demonstrate in vivo target engagement and reduction of tumor burden in three mouse xenograft models driven by either wild-type or mutant RAS. Intracellular delivery of a potent anti-RAS biologic through a receptor-mediated mechanism represents a promising new approach to developing RAS therapeutics against a broad array of cancers.SignificanceRAS oncoproteins have long been considered among the most elusive drug targets in cancer research. At issue is the lack of accessible drug binding sites and the extreme affinity for its GTP substrate. Covalent inhibitors against the KRAS G12C mutant have shown early clinical promise, however, targeting the other oncogenic RAS mutants across three RAS isoforms has proven challenging. Inhibition of activated wild-type RAS in the absence of canonical RAS mutations is also highly desirable in certain tumors. Here, we demonstrate delivery of an extremely potent pan-RAS and RAP1 cleaving enzyme in therapeutic quantities to specific receptor-bearing cells in vitro and in vivo. We aim to advance this approach to engineer the first targeted pan-RAS inhibitor for cancer therapy.One Sentence SummaryEngineered chimeric toxin halts tumor growth in vivo via RAS cleavage
- Published
- 2019
- Full Text
- View/download PDF
11. Identification of the C. sordellii lethal toxin receptor elucidates principles of receptor specificity in clostridial toxins
- Author
-
Greg L. Beilhartz, Mikko Taipale, Hunsang Lee, Jean-Philippe Julien, Daniel Schramek, Swetha Raman, Iga Kucharska, Roman A. Melnyk, John L. Rubinstein, Hong Cui, and Mandy H. Y. Lam
- Subjects
0303 health sciences ,Frizzled ,Toxin ,Cas9 ,Toxic shock syndrome ,Biology ,medicine.disease ,medicine.disease_cause ,3. Good health ,Microbiology ,03 medical and health sciences ,0302 clinical medicine ,Semaphorin ,medicine ,Extracellular ,CRISPR ,Receptor ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
Clostridium sordellii lethal toxin (TcsL) is responsible for an almost invariably lethal toxic shock syndrome associated with gynecological C. sordellii infections. Here, using CRISPR/Cas9 screening, we identify semaphorins SEMA6A and SEMA6B as the cellular receptors for TcsL and demonstrate that soluble extracellular SEMA6A can protect mice from TcsL-induced edema. A 3.3 Å cryo-EM structure shows that TcsL binds SEMA6A with the same region that the highly related C. difficile TcdB toxin uses to bind structurally unrelated Frizzled receptors. Remarkably, reciprocal mutations in this evolutionarily divergent surface are sufficient to switch receptor specificity between the toxins. Our findings establish semaphorins as physiologically relevant receptors for TcsL, and reveal the molecular basis for the difference in tissue targeting and disease pathogenesis between highly related toxins.
- Published
- 2019
- Full Text
- View/download PDF
12. Attenuated diphtheria toxin mediates siRNA delivery
- Author
-
Amy E. Arnold, Laura C. Bahlmann, Emma Jameson, Greg L. Beilhartz, Molly S. Shoichet, Roman A. Melnyk, and Laura Smith
- Subjects
Small interfering RNA ,Cell ,Endosomes ,Endocytosis ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,medicine ,Initiation factor ,Humans ,Diphtheria Toxin ,RNA, Small Interfering ,Research Articles ,030304 developmental biology ,Cancer ,Diphtheria toxin ,0303 health sciences ,Gene knockdown ,Multidisciplinary ,Chemistry ,SciAdv r-articles ,3. Good health ,Cell biology ,medicine.anatomical_structure ,Applied Sciences and Engineering ,030220 oncology & carcinogenesis ,Cancer cell ,Lysosomes ,human activities ,Research Article - Abstract
Hitching a ride out of the endosome results in gene knockdown and functional response after siRNA delivery to brain cancer cells., Toxins efficiently deliver cargo to cells by binding to cell surface ligands, initiating endocytosis, and escaping the endolysosomal pathway into the cytoplasm. We took advantage of this delivery pathway by conjugating an attenuated diphtheria toxin to siRNA, thereby achieving gene downregulation in patient-derived glioblastoma cells. We delivered siRNA against integrin-β1 (ITGB1)—a gene that promotes invasion and metastasis—and siRNA against eukaryotic translation initiation factor 3 subunit b (eIF-3b)—a survival gene. We demonstrated mRNA downregulation of both genes and the corresponding functional outcomes: knockdown of ITGB1 led to a significant inhibition of invasion, shown with an innovative 3D hydrogel model; and knockdown of eIF-3b resulted in significant cell death. This is the first example of diphtheria toxin being used to deliver siRNAs, and the first time a toxin-based siRNA delivery strategy has been shown to induce relevant genotypic and phenotypic effects in cancer cells.
- Published
- 2019
13. bioPROTACs as versatile modulators of intracellular therapeutic targets: Application to proliferating cell nuclear antigen (PCNA)
- Author
-
Shuhui Lim, Regina Khoo, Khong Ming Peh, Jinkai Teo, Shih Chieh Chang, Simon Ng, Greg L. Beilhartz, Roman A. Melnyk, Charles W. Johannes, Christopher J. Brown, David P. Lane, Brian Henry, and Anthony W. Partridge
- Subjects
DNA clamp ,biology ,RNA interference ,Chemistry ,biology.protein ,DNA replication ,Target protein ,Fusion protein ,Small molecule ,Ubiquitin ligase ,Proliferating cell nuclear antigen ,Cell biology - Abstract
Targeted degradation approaches have recently generated much excitement as a paradigm shift to address human disease in unprecedented ways. Amongst these, small molecule based approaches such as Proteolysis targeting chimeras (PROTACs) have attracted the lion’s share of attention due to their potential to tackle historically intractable targets and achieve greater potency, efficacy, and specificity over traditional small molecule inhibitors. Despite their promise, the identification of high-affinity ligands that can serve as starting points for PROTAC strategies remains challenging. As a complementary approach, we describe herein a class of intracellular biologics termed bioPROTACs. The substrate binding component of these fusion proteins consists of a peptide or an antibody-mimetic which allows for an unprecedented diversity of protein targets that can be addressed. The high-affinity binder is linked directly to an E3 ubiquitin ligase to harness the power of targeted degradation. Using GFP-tagged proteins as model substrates, we show that there is considerable flexibility in both the choice of substrate binders (binding positions, scaffold-class) and the E3 ligases. Indeed, 9 out of 16 binder-E3 combinations tested resulted in greater than 70% target clearance. Through a systematic approach, we then identified a highly effective bioPROTAC against an oncology target, proliferating cell nuclear antigen (PCNA), a sliding DNA clamp with critical roles in DNA replication and repair. The bioPROTAC, termed Con1-SPOP, elicited rapid and robust PCNA degradation and associated effects on DNA synthesis and cell cycle progression. Compared to RNAi-based approaches which typically take days to manifest, PCNA knockdown using Con1-SPOP was evident within 4 h. The advantage of degradation versus stoichiometric inhibition was also clearly demonstrated with bioPROTAC strategies. Combining superior pharmacological inhibition and relative ease of development, bioPROTACs are powerful tools for interrogating the degradability of a substrate, for guiding the identification of the fittest E3 ligase, for studying the functional consequences associated with target protein down-regulation, and potentially for making therapeutic impacts.
- Published
- 2019
- Full Text
- View/download PDF
14. Intracellular Delivery of Human Purine Nucleoside Phosphorylase by Engineered Diphtheria Toxin Rescues Function in Target Cells
- Author
-
Weixian Min, Xiaobai Xu, Roman A. Melnyk, Minyoung Park, Eyal Grunebaum, Seiji Sugiman-Marangos, Sachdev S. Sidhu, Greg L. Beilhartz, and Jarret J. Adams
- Subjects
0301 basic medicine ,Purine-Pyrimidine Metabolism, Inborn Errors ,Primary Immunodeficiency Diseases ,Recombinant Fusion Proteins ,T-Lymphocytes ,Induced Pluripotent Stem Cells ,Pharmaceutical Science ,Purine nucleoside phosphorylase ,Protein Engineering ,03 medical and health sciences ,0302 clinical medicine ,Cytosol ,Drug Delivery Systems ,Drug Discovery ,Extracellular ,Cytotoxic T cell ,Humans ,Diphtheria Toxin ,Receptor ,chemistry.chemical_classification ,Diphtheria toxin ,B-Lymphocytes ,Cell biology ,030104 developmental biology ,Enzyme ,chemistry ,Purine-Nucleoside Phosphorylase ,Molecular Medicine ,030217 neurology & neurosurgery ,Intracellular - Abstract
Despite a wealth of potential applications inside target cells, protein-based therapeutics are largely limited to extracellular targets due to the inability of proteins to readily cross biological membranes and enter the cytosol. Bacterial toxins, which deliver a cytotoxic enzyme into cells as part of their intoxication mechanism, hold great potential as platforms for delivering therapeutic protein cargo into cells. Diphtheria toxin (DT) has been shown to be capable of delivering an array of model proteins of varying sizes, structures, and stabilities into mammalian cells as amino-terminal fusions. Here, seeking to expand the utility of DT as a delivery vector, we asked whether an active human enzyme, purine nucleoside phosphorylase (PNP), could be delivered by DT into cells to rescue PNP deficiency. Using a series of biochemical and cellular readouts, we demonstrate that PNP is efficiently delivered into target cells in a receptor- and translocation-dependent manner. In patient-derived PNP-deficient lymphocytes and pluripotent stem cell-differentiated neurons, we show that human PNP is efficiently translocated into target cells by DT, where it is able to restore intracellular hypoxanthine levels. Further, through replacement of the native receptor-binding moiety of DT with single-chain variable fragments that were selected to bind mouse HBEGF, we show that PNP can be retargeted into mouse splenocytes from PNP-deficient mice, resulting in restoration of the proliferative capacity of T-cells. These findings highlight the versatility of the DT delivery platform and provide an attractive approach for the delivery of PNP as well as other cytosolic enzymes implicated in disease.
- Published
- 2018
15. Derivatives of Mesoxalic Acid Block Translocation of HIV-1 Reverse Transcriptase
- Author
-
Lianhai Li, Albert M. Berghuis, Daria J. Hazuda, Greg L. Beilhartz, Jay A. Grobler, Rico Lavoie, Roman A. Melnyk, Egor P. Tchesnokov, Sidney M. Hecht, Rakesh Paul, Anick Auger, Marianne Ngure, Michael D. Miller, Matthias Götte, and Jean A. Bernatchez
- Subjects
Models, Molecular ,Stereochemistry ,Mesoxalic acid ,Ribonuclease H ,Drug Evaluation, Preclinical ,Plasma protein binding ,Biochemistry ,Catalysis ,Structure-Activity Relationship ,chemistry.chemical_compound ,Catalytic Domain ,Structure–activity relationship ,Binding site ,RNase H ,Molecular Biology ,Ions ,biology ,Mutagenesis ,Hydrazones ,Active site ,Cell Biology ,HIV Reverse Transcriptase ,Malonates ,Reverse transcriptase ,Anti-Retroviral Agents ,chemistry ,Metals ,Mutation ,Enzymology ,HIV-1 ,biology.protein ,Reverse Transcriptase Inhibitors ,Protein Multimerization ,Protein Binding - Abstract
The pyrophosphate mimic and broad spectrum antiviral phosphonoformic acid (PFA, foscarnet) was shown to freeze the pre-translocational state of the reverse transcriptase (RT) complex of the human immunodeficiency virus type 1 (HIV-1). However, PFA lacks a specificity domain, which is seen as a major reason for toxic side effects associated with the clinical use of this drug. Here, we studied the mechanism of inhibition of HIV-1 RT by the 4-chlorophenylhydrazone of mesoxalic acid (CPHM) and demonstrate that this compound also blocks RT translocation. Hot spots for inhibition with PFA or CPHM occur at template positions with a bias toward pre-translocation. Mutations at active site residue Asp-185 compromise binding of both compounds. Moreover, divalent metal ions are required for the formation of ternary complexes with either of the two compounds. However, CPHM contains both an anchor domain that likely interacts with the catalytic metal ions and a specificity domain. Thus, although the inhibitor binding sites may partly overlap, they are not identical. The K65R mutation in HIV-1 RT, which reduces affinity to PFA, increases affinity to CPHM. Details with respect to the binding sites of the two inhibitors are provided on the basis of mutagenesis studies, structure-activity relationship analyses with newly designed CPHM derivatives, and in silico docking experiments. Together, these findings validate the pre-translocated complex of HIV-1 RT as a specific target for the development of novel classes of RT inhibitors.
- Published
- 2015
- Full Text
- View/download PDF
16. Inhibition of the Ribonuclease H Activity of HIV-1 Reverse Transcriptase by GSK5750 Correlates with Slow Enzyme-Inhibitor Dissociation
- Author
-
Greg L. Beilhartz, Peter Gerondelis, Felix Deanda, Brian A. Johns, Marianne Ngure, and Matthias Götte
- Subjects
Pyridines ,RNase P ,Ribonuclease H ,Pyrimidinones ,Biology ,Biochemistry ,Ribonuclease ,RNase H ,Molecular Biology ,chemistry.chemical_classification ,Base Sequence ,virus diseases ,Active site ,Cell Biology ,Molecular biology ,HIV Reverse Transcriptase ,Reverse transcriptase ,Dissociation constant ,Kinetics ,Enzyme ,Oligodeoxyribonucleotides ,chemistry ,Enzyme inhibitor ,Enzymology ,biology.protein ,Reverse Transcriptase Inhibitors - Abstract
Compounds that efficiently inhibit the ribonuclease (RNase) H activity of the human immunodeficiency virus type 1 (HIV-1) reverse transcriptase (RT) have yet to be developed. Here, we demonstrate that GSK5750, a 1-hydroxy-pyridopyrimidinone analog, binds to the enzyme with an equilibrium dissociation constant (K(d)) of ~400 nM. Inhibition of HIV-1 RNase H is specific, as DNA synthesis is not affected. Moreover, GSK5750 does not inhibit the activity of Escherichia coli RNase H. Order-of-addition experiments show that GSK5750 binds to the free enzyme in an Mg(2+)-dependent fashion. However, as reported for other active site inhibitors, binding of GSK5750 to a preformed enzyme-substrate complex is severely compromised. The bound nucleic acid prevents access to the RNase H active site, which represents a possible biochemical hurdle in the development of potent RNase H inhibitors. Previous studies suggested that formation of a complex with the prototypic RNase H inhibitor β-thujaplicinol is slow, and, once formed, it dissociates rapidly. This unfavorable kinetic behavior can limit the potency of RNase H active site inhibitors. Although the association kinetics of GSK5750 remains slow, our data show that this compound forms a long lasting complex with HIV-1 RT. We conclude that slow dissociation of the inhibitor and HIV-1 RT improves RNase H active site inhibitors and may circumvent the obstacle posed by the inability of these compounds to bind to a preformed enzyme-substrate complex.
- Published
- 2014
- Full Text
- View/download PDF
17. Abstract C031: Ras processing by RRSP protease as a strategy to inhibit Ras-driven tumors
- Author
-
Greg L. Beilhartz, K.J.F. Satchell, Roman A. Melnyk, and Vania Vidimar
- Subjects
Cancer Research ,Chemistry ,Colorectal cancer ,Growth factor ,medicine.medical_treatment ,medicine.disease_cause ,medicine.disease ,Oncology ,Downregulation and upregulation ,Cell culture ,Cancer cell ,medicine ,Cancer research ,KRAS ,Viability assay ,Receptor - Abstract
RAS genes are the most frequently mutated oncogenes in human cancers encoding four highly-related proteins that are critical molecular hubs for cancer cell proliferation and survival. Although promising results have been recently reported in early trials for KRas G12C inhibitors, there are no FDA-approved drugs that are effective against Ras-driven cancers, and Ras is still considered one of the most elusive targets in cancer research. We discovered the Ras-Rap1-Specific Protease (RRSP) from bacterium Vibrio vulnificus. RRSP is a naturally occurring endopeptidase that site-specifically cleaves the major Ras isoforms (H, N and K) and the most common mutated Ras oncoproteins (G12V, G13D and Q61R) within the Switch I region. In order to bring RRSP into cancer cells, RRSP was fused to diphtheria toxin binding subunit B (DTB). DTB delivers RRSP into the cytosol of targeted cells via binding to its receptor hHB-EGF (human heparin-binding epidermal growth factor-like growth factor). KRAS mutations occur in up to 50% of colorectal cancer (CRC) and in approximately 2% of triple-negative breast cancer (TNBC). However, 50% of TNBC show Ras upregulation via mutation-independent activation mechanisms. We investigated the anticancer potential of RRSP in CRC HCT-116 KRAS G13D as well as TNBC MDA-MB-231 KRAS G13D and MDA-MB-436 KRAS WT cell lines. RRSP was successfully delivered via DTB into CRC and TNBC cancer cell lines as indicated by Ras cleavage and pERK1/2 dephosphorylation at concentrations as low as 10 pM. As a consequence of Ras processing, a strong decrease in cell viability and proliferation was observed in all cell lines tested as per crystal violet staining and CellTiter Glo assay. A catalytically-dead mutant of RRSP carrying an amino acid substitution in its active site (RRSP*-DTB) did not show Ras processing nor reduced cell viability. Interestingly, our in vivo xenograft data demonstrated that, unlike RRSP*-DTB, RRSP-DTB effectively inhibited TNBC MDA-MB-436 and MDA-MB-231 tumor growth with no obvious toxicity. Proof of RRSP-target engagement was confirmed by loss of pERK1/2 phosphorylation via immunohistochemical analysis of tumor sections. We also found that RRSP-DTB strongly reduced the size and viability of 3-dimensional CRC HCT-116 spheroids in a time and dose-dependent manner. Moreover, an independent experiment at Charles River showed significant inhibition of tumor growth in a CRC HCT-116 xenograft model. Altogether our results revealed that, by physically processing Ras, RRSP strongly inhibits tumor growth in subcutaneously implanted human CRC and TNBC Ras-dependent tumors bearing either mutant or wild-type KRAS. This work supports further development of RRSP as an anticancer therapeutic for a broad spectrum of Ras-driven tumors and has the potential to take anti-Ras therapy to the next level. Citation Format: Vania Vidimar, Greg L Beilhartz, Roman A Melnyk, Karla JF Satchell. Ras processing by RRSP protease as a strategy to inhibit Ras-driven tumors [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference on Molecular Targets and Cancer Therapeutics; 2019 Oct 26-30; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2019;18(12 Suppl):Abstract nr C031. doi:10.1158/1535-7163.TARG-19-C031
- Published
- 2019
- Full Text
- View/download PDF
18. Small Molecules Take A Big Step Against Clostridium difficile
- Author
-
Greg L. Beilhartz, John Kit Chung Tam, and Roman A. Melnyk
- Subjects
Microbiology (medical) ,medicine.drug_class ,Antibiotics ,Clostridium Infections ,Biology ,Clostridium difficile ,Microbiology ,Small molecule ,Virology ,Clostridium difficile infections ,Infectious Diseases ,medicine ,Effective treatment - Abstract
Effective treatment of Clostridium difficile infections demands a shift away from antibiotics towards toxin-neutralizing agents. Work by Bender et al., using a drug that attenuates toxin action in vivo without affecting bacterial survival, demonstrates the exciting potential of small molecules as a new modality in the fight against C. difficile.
- Published
- 2015
- Full Text
- View/download PDF
19. The anticancer activity of the combination therapy of Gemcitabine and Doxorubicin encapsulated in a nanoemulsion
- Author
-
Anick Auger, Greg L. Beilhartz, Roman A. Melnyk, and Minyoung Park
- Subjects
Microbial toxins ,Toxin ,Chemistry ,medicine ,medicine.disease_cause ,Microbiology - Published
- 2017
- Full Text
- View/download PDF
20. Comment on 'A small-molecule antivirulence agent for treating Clostridium difficile infection'
- Author
-
Greg L. Beilhartz, Zhifen Zhang, Roman A. Melnyk, and John Kit Chung Tam
- Subjects
0301 basic medicine ,Antivirulence ,Ebselen ,Clostridioides difficile ,030106 microbiology ,Clostridium Infections ,General Medicine ,Biology ,Clostridium difficile ,Small molecule ,Virology ,3. Good health ,Microbiology ,Anti-Bacterial Agents ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,030104 developmental biology ,Mechanism of action ,chemistry ,medicine ,Molecular targets ,Animals ,medicine.symptom - Abstract
New insights into the mechanism of action of ebselen, a small-molecule antivirulence agent that reduces disease pathology in a mouse model of Clostridium difficile infection, suggest a different molecular target may be responsible for its efficacy.
- Published
- 2016
21. Telbivudine Exerts no Antiviral Activity against HIV-1 In Vitro and in Humans
- Author
-
Andy I. M. Hoepelman, Dorien de Jong, Noortje M van Maarseveen, Greg L. Beilhartz, Raymond F. Schinazi, Annemarie M. J. Wensing, Joop E. Arends, Aleksandr Obikhod, Matthias Götte, Monique Nijhuis, Sijia Tao, and Marieke Pingen
- Subjects
Adult ,Male ,Drug ,Hepatitis B virus ,Genotype ,media_common.quotation_subject ,Human immunodeficiency virus (HIV) ,HIV Infections ,Pyrimidinones ,Pharmacology ,medicine.disease_cause ,Antiviral Agents ,Article ,Cell Line ,Hepatitis B, Chronic ,Telbivudine ,Drug Resistance, Viral ,Humans ,Medicine ,Pharmacology (medical) ,media_common ,Coinfection ,business.industry ,virus diseases ,Nucleosides ,Middle Aged ,Viral Load ,In vitro ,CD4 Lymphocyte Count ,HEK293 Cells ,Phenotype ,Infectious Diseases ,DNA, Viral ,HIV-1 ,RNA, Viral ,business ,Thymidine ,medicine.drug - Abstract
Background HIV–HBV-coinfected individuals who need to be treated only for their HBV infection have limited therapeutic options, since most approved anti-HBV agents have a risk of selecting for drug-resistant HIV mutants. In vivo data are inconclusive as to whether telbivudine (LdT) may exert antiviral effects against HIV. Thus, we investigated in further detail the antiviral activity and the biochemical properties of LdT against HIV-1. Methods To investigate the activity of LdT against HIV-1 in humans we analysed viral dynamics and genotypic and phenotypic resistance development in two HIV–HBV-coinfected individuals with no prior antiviral exposure. To investigate the activity of LdT against HIV-1 in vitro, LdT susceptibility for HIV-1 wild-type strains as well as drug-resistant strains was determined. Furthermore, we studied whether the 5′-triphosphate form of LdT (LdT-TP) can act as a substrate for wild-type HIV-1 RT. Results In the two patients studied, LdT treatment did not result in a significant decline of HIV-1 RNA load nor in selection of genotypic or phenotypic resistance in HIV-1 RT. In vitro virological analyses demonstrated that LdT had no activity (50% effective concentration >100 μM) against wild type HIV and drug-resistant variants. Biochemical analyses demonstrated that LdT-TP is not incorporated by wild-type HIV-1 RT. Conclusions Based on the in vivo and in vitro evidence obtained in this study, we conclude that LdT has no anti-HIV-1 activity and is currently the only selective anti-HBV agent among the five FDA-approved nucleoside/nucleotide analogues for treatment of HBV infections in HIV-infected individuals.
- Published
- 2011
- Full Text
- View/download PDF
22. HIV-1 reverse transcriptase inhibitors: beyond classic nucleosides and non-nucleosides
- Author
-
Greg L. Beilhartz, Maryam Ehteshami, Brian J. Scarth, and Matthias Götte
- Subjects
chemistry.chemical_classification ,Drug discovery ,Allosteric regulation ,Human immunodeficiency virus (HIV) ,Biology ,medicine.disease_cause ,Pyrophosphate ,Reverse transcriptase ,Discovery and development of non-nucleoside reverse-transcriptase inhibitors ,chemistry.chemical_compound ,Enzyme ,Biochemistry ,chemistry ,Virology ,medicine ,Nucleoside - Abstract
Reverse transcriptase (RT) of HIV-1 remains an important target in current treatments of HIV-1 infection. Clinically available inhibitors of HIV-1 RT include nucleoside analog RT inhibitors and non-nucleoside RT inhibitors. Nucleoside analog RT inhibitors compete with the natural dNTP substrate and act as chain terminators, while non-nucleoside RT inhibitors bind to an allosteric pocket, inhibiting polymerization noncompetitively. In addition to these two classes of approved drugs, there are a number of RT inhibitors that target the enzyme in different ways. These include nonobligate chain terminators, nucleotide-competing RT inhibitors, pyrophosphate analogs and compounds that inhibit the RT-associated RNase H activity. Here, we review the mechanisms of action associated with these compounds and discuss opportunities and challenges in drug discovery and development efforts.
- Published
- 2011
- Full Text
- View/download PDF
23. Nuclear translocation of the 1,25D3-MARRS (membrane associated rapid response to steroids) receptor protein and NFκB in differentiating NB4 leukemia cells
- Author
-
Yvette Roy, Danielle Cadieux, Greg L. Beilhartz, Kelly A. Meckling, Wenqing Wu, Cynthia L. Richard, Mary C. Farach-Carson, Marc G. Coppolino, Ilka Nemere, Lauren Brown, and Maureen Curtin
- Subjects
Active Transport, Cell Nucleus ,Protein Disulfide-Isomerases ,Biology ,Cell Fractionation ,Calcitriol receptor ,Gene product ,Calcitriol ,Leukemia, Promyelocytic, Acute ,Cell surface receptor ,Gene expression ,Tumor Cells, Cultured ,medicine ,Humans ,Tissue Distribution ,Receptor ,Transcription factor ,Cell Nucleus ,Microscopy, Confocal ,Gene Expression Regulation, Leukemic ,Monocyte ,NF-kappa B ,Membrane Proteins ,Cell Differentiation ,Cell Biology ,Molecular biology ,Protein Transport ,medicine.anatomical_structure ,Nuclear localization sequence - Abstract
1,25 Dihydroxyvitamin D{sub 3} (1,25D{sub 3}) primes NB4 promyelocytic leukemia cells to differentiate along the monocyte/macrophage lineage through a non-genomic mechanism. Here we show that NB4 cells express high levels of the recently identified membrane receptor for 1,25D{sub 3}, which is a distinct gene product from the classical nuclear vitamin D receptor. This 57 kDa protein, named 1,25D{sub 3}-MARRS (Membrane Activated Rapid Response to Steroids)/ERp57/PIA3 appears to associate in a complex with the transcription factor, nuclear factor kappa B (NF{kappa}B). In unstimulated cells, 1,25D{sub 3}-MARRS can be co-immunoprecipitated with antibodies directed at NF{kappa}B, and NF{kappa}B is co-precipitated when antibodies against 1,25D{sub 3}-MARRS or ERp57 are used. Confocal microscopy and subcellular fractionation studies demonstrate that both 1,25D{sub 3}-MARRS and NF{kappa}B begin translocating to the nucleus within minutes of co-stimulation with 1,25D{sub 3} and phorbol ester. The predominant nuclear localization of both proteins precedes the expression of the monocyte/macrophage phenotype and suggests that this event may be critical to the differentiation pathway. This suggests a role for 1,25D{sub 3}-MARRS in the nucleus as a regulator of gene expression. Here it may also regulate the activity of NF{kappa}B and other factors with which it may be interacting.
- Published
- 2010
- Full Text
- View/download PDF
24. HIV ribonuclease H: continuing the search for small molecule antagonists
- Author
-
John A. Beutler, Michaela Wendeler, Greg L. Beilhartz, Stuart F.J. Le Grice, and Matthias Götte
- Subjects
Pharmacology ,chemistry.chemical_classification ,RNase P ,RNA ,Dermatology ,Biology ,Molecular biology ,Reverse transcriptase ,RNase MRP ,Infectious Diseases ,Enzyme ,Endoribonucleases ,chemistry ,Virology ,Drug Discovery ,Phosphodiester bond ,biology.protein ,Pharmacology (medical) ,RNase H - Abstract
Members of the ribonuclease H (RNase H) family of enzymes (EC 3.1.26.4), which are found in nearly all organisms, are endoribonucleases that specifically hydrolyze the phosphodiester bond of RNA in a RNA–DNA hybrid. In retroviruses such as HIV-1, the RNase H activity is part of reverse transcriptase, the enzyme that converts the viral ssRNA into dsDNA suitable for integration into the host cell genome. In HIV-1, RNase H plays an essential role in various stages of reverse transcription, and it has been known for 20 years that inhibiting RNase H activity renders HIV noninfectious. However, the development of potent and selective antagonists of HIV RNase H has made surprisingly slow progress, and so far no RNase H inhibitor is in clinical trial, rendering this enzyme an important, but as yet underexplored, drug target. The recently described crystal structure of human RNase H in complex with a RNA–DNA hybrid provides new insight into the mechanism of HIV RNase H activity, with the potential to unveil new niches for therapeutic intervention. The current status of RNase H screening efforts is reviewed here.
- Published
- 2009
- Full Text
- View/download PDF
25. Connection Domain Mutations N348I and A360V in HIV-1 Reverse Transcriptase Enhance Resistance to 3′-Azido-3′-deoxythymidine through Both RNase H-dependent and -independent Mechanisms
- Author
-
Brian Wynhoven, Suzanne McCormick, Maryam Ehteshami, Brian J. Scarth, Greg L. Beilhartz, Matthias Götte, P. Richard Harrigan, and Egor P. Tchesnokov
- Subjects
Anti-HIV Agents ,RNase P ,Molecular Sequence Data ,Ribonuclease H ,Mutant ,Biochemistry ,03 medical and health sciences ,chemistry.chemical_compound ,Zidovudine ,Drug Resistance, Viral ,medicine ,RNase H ,Molecular Biology ,030304 developmental biology ,0303 health sciences ,Base Sequence ,Enzyme Catalysis and Regulation ,biology ,030306 microbiology ,Cell Biology ,Processivity ,Molecular biology ,HIV Reverse Transcriptase ,Reverse transcriptase ,Protein Structure, Tertiary ,3. Good health ,chemistry ,DNA, Viral ,Mutation ,HIV-1 ,biology.protein ,RNA, Viral ,Thymidine ,DNA ,Protein Binding ,medicine.drug - Abstract
Thymidine analogue-associated mutations (TAMs) in reverse transcriptase (RT) of the human immunodeficiency virus type 1 (HIV-1) cause resistance to 3'-azido-3'-deoxythymidine (AZT) through excision of the incorporated monophosphate. Mutations in the connection domain of HIV-1 RT can augment AZT resistance. It has been suggested that these mutations compromise RNase H cleavage, providing more time for AZT excision to occur. However, the underlying mechanism remains elusive. Here, we focused on connection mutations N348I and A360V that are frequently observed in clinical samples of treatment-experienced patients. We show that both N348I and A360V, in combination with TAMs, decrease the efficiency of RNase H cleavage and increase excision of AZT in the presence of the pyrophosphate donor ATP. The TAMs/N348I/A360V mutant accumulates transiently formed, shorter hybrids that can rebind to RT before the template is irreversibly degraded. These hybrids dissociate selectively from the RNase H-competent complex, whereas binding in the polymerase-competent mode is either not affected with N348I or modestly improved with A360V. Both connection domain mutations can compensate for TAM-mediated deficits in processive DNA synthesis, and experiments with RNase H negative mutant enzymes confirm an RNase H-independent contribution to increased levels of resistance to AZT. Moreover, the combination of diminished RNase H cleavage and increased processivity renders the use of both PP(i) and ATP advantageous, whereas classic TAMs solely enhance the ATP-dependent reaction. Taken together, our findings demonstrate that distinct, complementary mechanisms can contribute to higher levels of excision of AZT, which in turn can amplify resistance to this drug.
- Published
- 2008
- Full Text
- View/download PDF
26. Translocation domain mutations affecting cellular toxicity identify the Clostridium difficile toxin B pore
- Author
-
Greg L. Beilhartz, John Kit Chung Tam, Zhifen Zhang, Anick Auger, Minyoung Park, Roman A. Melnyk, and D. Borden Lacy
- Subjects
Models, Molecular ,Pore Forming Cytotoxic Proteins ,media_common.quotation_subject ,Bacterial Toxins ,Molecular Sequence Data ,Chromosomal translocation ,Clostridium difficile toxin B ,Biology ,Fluorescence ,Amino Acid Sequence ,Internalization ,Peptide sequence ,media_common ,Diphtheria toxin ,Multidisciplinary ,Clostridioides difficile ,Biological membrane ,Biological Sciences ,Transmembrane protein ,Cell biology ,High-Throughput Screening Assays ,Protein Structure, Tertiary ,Cytosol ,Biochemistry ,Mutagenesis ,Mutation ,Rubidium Radioisotopes - Abstract
Disease associated with Clostridium difficile infection is caused by the actions of the homologous toxins TcdA and TcdB on colonic epithelial cells. Binding to target cells triggers toxin internalization into acidified vesicles, whereupon cryptic segments from within the 1,050-aa translocation domain unfurl and insert into the bounding membrane, creating a transmembrane passageway to the cytosol. Our current understanding of the mechanisms underlying pore formation and the subsequent translocation of the upstream cytotoxic domain to the cytosol is limited by the lack of information available regarding the identity and architecture of the transmembrane pore. Here, through systematic perturbation of conserved sites within predicted membrane-insertion elements of the translocation domain, we uncovered highly sensitive residues—clustered between amino acids 1,035 and 1,107—that when individually mutated, reduced cellular toxicity by as much as >1,000-fold. We demonstrate that defective variants are defined by impaired pore formation in planar lipid bilayers and biological membranes, resulting in an inability to intoxicate cells through either apoptotic or necrotic pathways. These findings along with the unexpected similarities uncovered between the pore-forming “hotspots” of TcdB and the well-characterized α-helical diphtheria toxin translocation domain provide insights into the structure and mechanism of formation of the translocation pore for this important class of pathogenic toxins.
- Published
- 2014
27. Impact of primer-induced conformational dynamics of HIV-1 reverse transcriptase on polymerase translocation and inhibition
- Author
-
Jean-Pierre Falgueyret, Greg L. Beilhartz, Maryam Ehteshami, Siqi Zhu, Jay A. Grobler, Anick Auger, Roman A. Melnyk, Elizabeth Cauchon, and Matthias Götte
- Subjects
Indoles ,DNA polymerase ,Pyridones ,Allosteric regulation ,Biochemistry ,Catalysis ,chemistry.chemical_compound ,Nitriles ,Molecular Biology ,Polymerase ,DNA Primers ,chemistry.chemical_classification ,biology ,Cell Biology ,Reverse Transcription ,Molecular biology ,Footprinting ,Reverse transcriptase ,HIV Reverse Transcriptase ,Enzyme ,chemistry ,DNA, Viral ,biology.protein ,HIV-1 ,RNA, Viral ,Primer (molecular biology) ,DNA ,Allosteric Site ,Molecular Biophysics - Abstract
The rapid emergence and the prevalence of resistance mutations in HIV-1 reverse transcriptase (RT) underscore the need to identify RT inhibitors with novel binding modes and mechanisms of inhibition. Recently, two structurally distinct inhibitors, phosphonoformic acid (foscarnet) and INDOPY-1 were shown to disrupt the translocational equilibrium of RT during polymerization through trapping of the enzyme in the pre- and the post-translocation states, respectively. Here, we show that foscarnet and INDOPY-1 additionally display a shared novel inhibitory preference with respect to substrate primer identity. In RT-catalyzed reactions using RNA-primed substrates, translocation inhibitors were markedly less potent at blocking DNA polymerization than in equivalent DNA-primed assays; i.e. the inverse pattern observed with marketed non-nucleoside inhibitors that bind the allosteric pocket of RT. This potency profile was shown to correspond with reduced binding on RNA·DNA primer/template substrates versus DNA·DNA substrates. Furthermore, using site-specific footprinting with chimeric RNA·DNA primers, we demonstrate that the negative impact of the RNA primer on translocation inhibitor potency is overcome after 18 deoxyribonucleotide incorporations, where RT transitions primarily into polymerization-competent binding mode. In addition to providing a simple means to identify similarly acting translocation inhibitors, these findings suggest a broader role for the primer-influenced binding mode on RT translocation equilibrium and inhibitor sensitivity.
- Published
- 2011
28. Structure-activity analysis of vinylogous urea inhibitors of human immunodeficiency virus-encoded ribonuclease H
- Author
-
Greg L. Beilhartz, Stuart F.J. Le Grice, Alun Bermingham, Matthias Götte, Xiaowei Zhuang, Suhman Chung, Barry R. O'Keefe, Michaela Wendeler, Shixin Liu, Jason W. Rausch, and John A. Beutler
- Subjects
Models, Molecular ,Molecular model ,Stereochemistry ,Anti-HIV Agents ,Antiviral Agents ,chemistry.chemical_compound ,Amide ,Catalytic Domain ,Thiophene ,Cyclooctane ,Humans ,Pharmacology (medical) ,Binding site ,Enzyme Inhibitors ,RNase H ,Pharmacology ,biology ,Molecular Structure ,Temperature ,Infectious Diseases ,Ribonuclease H, Human Immunodeficiency Virus ,chemistry ,biology.protein ,Thermodynamics ,Pharmacophore ,Cycloheptane - Abstract
Vinylogous ureas 2-amino-5,6,7,8-tetrahydro-4 H -cyclohepta[ b ]thiophene-3-carboxamide and N -[3-(aminocarbonyl)-4,5-dimethyl-2-thienyl]-2-furancarboxamide (compounds 1 and 2, respectively) were recently identified to be modestly potent inhibitors of the RNase H activity of HIV-1 and HIV-2 reverse transcriptase (RT). Both compounds shared a 3-CONH 2 -substituted thiophene ring but were otherwise structurally unrelated, which prevented a precise definition of the pharmacophore. We have therefore examined a larger series of vinylogous ureas carrying amide, amine, and cycloalkane modifications of the thiophene ring of compound 1. While cycloheptane- and cyclohexane-substituted derivatives retained potency, cyclopentane and cyclooctane substitutions eliminated activity. In the presence of a cycloheptane ring, modifying the 2-NH 2 or 3-CONH 2 functions decreased the potency. With respect to compound 2, vinylogous ureas whose dimethylthiophene ring contained modifications of the 2-NH 2 and 3-CONH 2 functions were investigated. 2-NH 2 -modified analogs displayed potency equivalent to or enhanced over that of compound 2, the most active of which, compound 16, reflected intramolecular cyclization of the 2-NH 2 and 3-CONH 2 groups. Molecular modeling was used to define an inhibitor binding site in the p51 thumb subdomain, suggesting that an interaction with the catalytically conserved His539 of the p66 RNase H domain could underlie inhibition of RNase H activity. Collectively, our data indicate that multiple functional groups of vinylogous ureas contribute to their potencies as RNase H inhibitors. Finally, single-molecule spectroscopy indicates that vinylogous ureas have the property of altering the reverse transcriptase orientation on a model RNA-DNA hybrid mimicking initiation plus-strand DNA synthesis.
- Published
- 2010
29. N348I in HIV-1 reverse transcriptase can counteract the nevirapine-mediated bias toward RNase H cleavage during plus-strand initiation
- Author
-
Suzanne McCormick, Mia J. Biondi, Greg L. Beilhartz, and Matthias Götte
- Subjects
Cyclopropanes ,Efavirenz ,Nevirapine ,RNase P ,Ribonuclease H ,Mutation, Missense ,Biology ,Biochemistry ,chemistry.chemical_compound ,Drug Resistance, Viral ,medicine ,RNase H ,Molecular Biology ,Polymerase ,DNA synthesis ,virus diseases ,Cell Biology ,Virology ,Molecular biology ,Reverse transcriptase ,HIV Reverse Transcriptase ,Benzoxazines ,chemistry ,Amino Acid Substitution ,Alkynes ,DNA, Viral ,Nucleic acid ,biology.protein ,HIV-1 ,Enzymology ,RNA, Viral ,Reverse Transcriptase Inhibitors ,medicine.drug - Abstract
Drug resistance-associated mutations in HIV-1 reverse transcriptase (RT) can affect the balance between polymerase and ribonuclease H (RNase H) activities of the enzyme. We have recently demonstrated that the N348I mutation in the connection domain causes selective dissociation from RNase H-competent complexes, whereas the functional integrity of the polymerase-competent complex remains largely unaffected. N348I has been associated with resistance to the non-nucleoside RT inhibitor (NNRTI), nevirapine; however, a possible mechanism that links changes in RNase H activity to changes in NNRTI susceptibility remains to be established. To address this problem, we consider recent findings suggesting that NNRTIs may affect the orientation of RT on its nucleic acid substrate and increase RNase H activity. Here we demonstrate that RNase H-mediated primer removal is indeed more efficient in the presence of NNRTIs; however, the N348I mutant enzyme is able to counteract this effect. Efavirenz, a tight binding inhibitor, restricts the influence of the mutation. These findings provide strong evidence to suggest that N348I can thwart the inhibitory effects of nevirapine during initiation of (+)-strand DNA synthesis, which provides a novel mechanism for resistance. The data are in agreement with clinical data, which demonstrate a stronger effect of N348I on susceptibility to nevirapine as compared with efavirenz.
- Published
- 2010
30. HIV-1 Reverse Transcriptase Can Simultaneously Engage its DNA/RNA Substrate at both the DNA Polymerase and RNase H Active Sites: Implications for RNase H Inhibition
- Author
-
Michaela Wendeler, Noel Baichoo, Jason W. Rausch, Greg L. Beilhartz, Stuart F.J. Le Grice, and Matthias Götte
- Subjects
biology ,DNA polymerase ,RNase P ,Active site ,RNase PH ,Molecular biology ,Reverse transcriptase ,Article ,HIV Reverse Transcriptase ,RNase MRP ,Biochemistry ,Structural Biology ,Catalytic Domain ,DNA, Viral ,biology.protein ,HIV-1 ,RNA, Viral ,RNase H ,Molecular Biology ,Polymerase ,Protein Binding - Abstract
Reverse transcriptase of the human immunodeficiency virus possesses DNA polymerase and ribonuclease (RNase) H activities. Although the nucleic acid binding cleft separating these domains can accommodate structurally diverse duplexes, it is currently unknown whether regular DNA/RNA hybrids can simultaneously contact both active sites. In this study, we demonstrate that ligands capable of trapping the 3'-end of the primer at the polymerase active site affect the specificity of RNase H cleavage without altering the efficiency of the reaction. Experiments under single-turnover conditions reveal that complexes with a bound nucleotide substrate show specific RNase H cleavage at template position -18, while complexes with the pyrophosphate analogue foscarnet show a specific cut at position -19. This pattern is indicative of post-translocated and pre-translocated conformations. The data are inconsistent with models postulating that the substrate toggles between both active sites, such that the primer 3'-terminus is disengaged from the polymerase active site when the template is in contact with the RNase H active site. In contrast, our findings provide strong evidence to suggest that the nucleic acid substrate can engage both active sites at the same time. As a consequence, the bound and intact DNA/RNA hybrid can restrict access of RNase H active site inhibitors. We have mapped the binding site of the recently discovered inhibitor beta-thujaplicinol between the RNase H active site and Y501 of the RNase H primer grip, and have shown that the inhibitor is unable to bind to a preformed reverse transcriptase-DNA/RNA complex. In conclusion, the bound nucleic acid substrate and in turn, active DNA synthesis can represent an obstacle to RNase H inhibition with compounds that bind to the RNase H active site.
- Published
- 2009
31. Small Molecule Inhibitors of Clostridium difficile Toxin B-Induced Cellular Damage
- Author
-
John Kit Chung Tam, Anick Auger, Pulkit Gupta, Greg L. Beilhartz, Alex G. Therien, and Roman A. Melnyk
- Subjects
Cell Survival ,Phenotypic screening ,Bacterial Toxins ,Cell ,Clinical Biochemistry ,Clostridium difficile toxin B ,Plasma protein binding ,Biology ,medicine.disease_cause ,Biochemistry ,Catechin ,Cell Line ,Microbiology ,Small Molecule Libraries ,Necrosis ,03 medical and health sciences ,Bacterial Proteins ,Gallic Acid ,Chlorocebus aethiops ,Drug Discovery ,medicine ,Animals ,Biflavonoids ,Humans ,Vero Cells ,Molecular Biology ,030304 developmental biology ,Pharmacology ,0303 health sciences ,Clostridioides difficile ,030306 microbiology ,Toxin ,General Medicine ,Clostridium difficile ,Small molecule ,3. Good health ,Kinetics ,medicine.anatomical_structure ,Phloretin ,Molecular Medicine ,Cholates ,Protein Binding - Abstract
SummaryClostridium difficile causes life-threatening diarrhea through the actions of its homologous toxins TcdA and TcdB on human colonocytes. Therapeutic agents that block toxin-induced damage are urgently needed to prevent the harmful consequences of toxin action that are not addressed with current antibiotic-based treatments. Here, we developed an imaging-based phenotypic screen to identify small molecules that protected human cells from TcdB-induced cell rounding. A series of structurally diverse compounds with antitoxin activity were identified and found to act through one of a small subset of mechanisms, including direct binding and sequestration of TcdB, inhibition of endosomal maturation, and noncompetitive inhibition of the toxin glucosyltransferase activity. Distinct classes of inhibitors were used further to dissect the determinants of the toxin-mediated necrosis phenotype occurring at higher doses of toxin. These findings validate and inform novel targeting strategies for discovering small molecule agents to treat C. difficile infection.
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.