LIVIVO - The Search Portal for Life Sciences

zur deutschen Oberfläche wechseln
Advanced search

Search results

Result 1 - 10 of total 31

Search options

  1. Article ; Online: Attenuated diphtheria toxin mediates siRNA delivery.

    Arnold, Amy E / Smith, Laura J / Beilhartz, Greg L / Bahlmann, Laura C / Jameson, Emma / Melnyk, Roman A / Shoichet, Molly S

    Science advances

    2020  Volume 6, Issue 18

    Abstract: 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 ... ...

    Abstract 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 (
    MeSH term(s) Diphtheria Toxin/genetics ; Diphtheria Toxin/metabolism ; Endocytosis ; Endosomes/metabolism ; Humans ; Lysosomes/metabolism ; RNA, Small Interfering/genetics ; RNA, Small Interfering/metabolism
    Chemical Substances Diphtheria Toxin ; RNA, Small Interfering
    Language English
    Publishing date 2020-05-01
    Publishing country United States
    Document type Journal Article ; Research Support, Non-U.S. Gov't
    ZDB-ID 2810933-8
    ISSN 2375-2548 ; 2375-2548
    ISSN (online) 2375-2548
    ISSN 2375-2548
    DOI 10.1126/sciadv.aaz4848
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  2. Article ; Online: Small Molecules Take A Big Step Against Clostridium difficile.

    Beilhartz, Greg L / Tam, John / Melnyk, Roman A

    Trends in microbiology

    2015  Volume 23, Issue 12, Page(s) 746–748

    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 ... ...

    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.
    MeSH term(s) Animals ; Anti-Bacterial Agents/therapeutic use ; Clostridium Infections/drug therapy ; Clostridium difficile/isolation & purification ; Virulence/drug effects
    Chemical Substances Anti-Bacterial Agents
    Language English
    Publishing date 2015-12
    Publishing country England
    Document type Comment ; Journal Article ; Research Support, Non-U.S. Gov't
    ZDB-ID 1158963-2
    ISSN 1878-4380 ; 0966-842X
    ISSN (online) 1878-4380
    ISSN 0966-842X
    DOI 10.1016/j.tim.2015.10.009
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  3. Article ; Online: Repurposing bacterial toxins for intracellular delivery of therapeutic proteins.

    Beilhartz, Greg L / Sugiman-Marangos, Seiji N / Melnyk, Roman A

    Biochemical pharmacology

    2017  Volume 142, Page(s) 13–20

    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 ... ...

    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.
    MeSH term(s) Animals ; Bacterial Toxins/chemistry ; Bacterial Toxins/metabolism ; Cytosol/metabolism ; Drug Carriers/chemistry ; Drug Carriers/metabolism ; Drug Discovery/methods ; Drug Discovery/trends ; Humans ; Pharmaceutical Preparations/administration & dosage ; Pharmaceutical Preparations/metabolism ; Protein Binding ; Receptors, Cell Surface/metabolism ; Recombinant Proteins/administration & dosage ; Recombinant Proteins/metabolism
    Chemical Substances Bacterial Toxins ; Drug Carriers ; Pharmaceutical Preparations ; Receptors, Cell Surface ; Recombinant Proteins
    Language English
    Publishing date 2017-10-15
    Publishing country England
    Document type Journal Article ; Review
    ZDB-ID 208787-x
    ISSN 1873-2968 ; 0006-2952
    ISSN (online) 1873-2968
    ISSN 0006-2952
    DOI 10.1016/j.bcp.2017.04.009
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  4. Article ; Online: Comment on "A small-molecule antivirulence agent for treating Clostridium difficile infection".

    Beilhartz, Greg L / Tam, John / Zhang, Zhifen / Melnyk, Roman A

    Science translational medicine

    2016  Volume 8, Issue 370, Page(s) 370tc2

    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. ...

    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.
    MeSH term(s) Animals ; Anti-Bacterial Agents ; Clostridium Infections ; Clostridium difficile ; Mice
    Chemical Substances Anti-Bacterial Agents
    Language English
    Publishing date 2016-12-18
    Publishing country United States
    Document type Journal Article ; Comment
    ZDB-ID 2518854-9
    ISSN 1946-6242 ; 1946-6234
    ISSN (online) 1946-6242
    ISSN 1946-6234
    DOI 10.1126/scitranslmed.aad8926
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  5. Article: Small Molecules Take A Big Step Against Clostridium difficile

    Beilhartz, Greg L / John Tam / Roman A. Melnyk

    Trends in microbiology. 2015 Dec., v. 23

    2015  

    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 ... ...

    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.
    Keywords antibiotics ; Clostridium difficile ; drugs
    Language English
    Dates of publication 2015-12
    Size p. 746-748.
    Publishing place Elsevier Ltd
    Document type Article
    ZDB-ID 1158963-2
    ISSN 1878-4380 ; 0966-842X
    ISSN (online) 1878-4380
    ISSN 0966-842X
    DOI 10.1016/j.tim.2015.10.009
    Database NAL-Catalogue (AGRICOLA)

    More links

    Kategorien

  6. Article ; Online: An engineered chimeric toxin that cleaves activated mutant and wild-type RAS inhibits tumor growth.

    Vidimar, Vania / Beilhartz, Greg L / Park, Minyoung / Biancucci, Marco / Kieffer, Matthew B / Gius, David R / Melnyk, Roman A / Satchell, Karla J F

    Proceedings of the National Academy of Sciences of the United States of America

    2020  Volume 117, Issue 29, Page(s) 16938–16948

    Abstract: Despite 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 pan-RAS biologic inhibitor composed of the RAS-RAP1-specific endopeptidase ... ...

    Abstract Despite 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 pan-RAS biologic inhibitor composed 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. Furthermore, we demonstrate in vivo target engagement and reduction of tumor burden in three mouse xenograft models driven by either wild-type or mutant
    MeSH term(s) Animals ; Antineoplastic Agents/therapeutic use ; Cells, Cultured ; Diphtheria Toxin/chemistry ; Diphtheria Toxin/genetics ; Diphtheria Toxin/metabolism ; Endopeptidases/chemistry ; Endopeptidases/genetics ; Endopeptidases/metabolism ; Female ; HCT116 Cells ; Humans ; Male ; Mice ; Mice, Nude ; Mutation ; Neoplasms, Experimental/drug therapy ; Protein Sorting Signals ; Proteolysis ; Recombinant Proteins/therapeutic use ; rap1 GTP-Binding Proteins/metabolism ; ras Proteins/genetics ; ras Proteins/metabolism
    Chemical Substances Antineoplastic Agents ; Diphtheria Toxin ; Protein Sorting Signals ; Recombinant Proteins ; Endopeptidases (EC 3.4.-) ; Rap1 protein, mouse (EC 3.6.5.2) ; rap1 GTP-Binding Proteins (EC 3.6.5.2) ; ras Proteins (EC 3.6.5.2)
    Language English
    Publishing date 2020-07-02
    Publishing country United States
    Document type Journal Article ; Research Support, N.I.H., Extramural ; Research Support, Non-U.S. Gov't
    ZDB-ID 209104-5
    ISSN 1091-6490 ; 0027-8424
    ISSN (online) 1091-6490
    ISSN 0027-8424
    DOI 10.1073/pnas.2000312117
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  7. Article ; Online: A designer peptide against the EAG2-Kvβ2 potassium channel targets the interaction of cancer cells and neurons to treat glioblastoma.

    Dong, Weifan / Fekete, Adam / Chen, Xiaodi / Liu, Hongwei / Beilhartz, Greg L / Chen, Xin / Bahrampour, Shahrzad / Xiong, Yi / Yang, Qi / Zhao, Hongyu / Kong, Tian / Morioka, Malia S / Jung, Geena / Kim, Ji-Eun / Schramek, Daniel / Dirks, Peter B / Song, Yuanquan / Kim, Tae-Hee / He, Ye /
    Wanggou, Siyi / Li, Xuejun / Melnyk, Roman A / Wang, Lu-Yang / Huang, Xi

    Nature cancer

    2023  Volume 4, Issue 10, Page(s) 1418–1436

    Abstract: Glioblastoma (GBM) is an incurable brain cancer that lacks effective therapies. Here we show that EAG2 and Kvβ2, which are predominantly expressed by GBM cells at the tumor-brain interface, physically interact to form a potassium channel complex due to a ...

    Abstract Glioblastoma (GBM) is an incurable brain cancer that lacks effective therapies. Here we show that EAG2 and Kvβ2, which are predominantly expressed by GBM cells at the tumor-brain interface, physically interact to form a potassium channel complex due to a GBM-enriched Kvβ2 isoform. In GBM cells, EAG2 localizes at neuron-contacting regions in a Kvβ2-dependent manner. Genetic knockdown of the EAG2-Kvβ2 complex decreases calcium transients of GBM cells, suppresses tumor growth and invasion and extends the survival of tumor-bearing mice. We engineered a designer peptide to disrupt EAG2-Kvβ2 interaction, thereby mitigating tumor growth in patient-derived xenograft and syngeneic mouse models across GBM subtypes without overt toxicity. Neurons upregulate chemoresistant genes in GBM cells in an EAG2-Kvβ2-dependent manner. The designer peptide targets neuron-associated GBM cells and possesses robust efficacy in treating temozolomide-resistant GBM. Our findings may lead to the next-generation therapeutic agent to benefit patients with GBM.
    MeSH term(s) Humans ; Mice ; Animals ; Glioblastoma/drug therapy ; Temozolomide/pharmacology ; Temozolomide/therapeutic use ; Ether-A-Go-Go Potassium Channels/therapeutic use ; Disease Models, Animal ; Peptides/therapeutic use ; Neurons/pathology
    Chemical Substances Temozolomide (YF1K15M17Y) ; Ether-A-Go-Go Potassium Channels ; Peptides
    Language English
    Publishing date 2023-09-11
    Publishing country England
    Document type Journal Article
    ISSN 2662-1347
    ISSN (online) 2662-1347
    DOI 10.1038/s43018-023-00626-8
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  8. Article ; Online: HIV-1 Ribonuclease H: Structure, Catalytic Mechanism and Inhibitors.

    Beilhartz, Greg L / Götte, Matthias

    Viruses

    2010  Volume 2, Issue 4, Page(s) 900–926

    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 ... ...

    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.
    Language English
    Publishing date 2010-03-30
    Publishing country Switzerland
    Document type Journal Article
    ZDB-ID 2516098-9
    ISSN 1999-4915 ; 1999-4915
    ISSN (online) 1999-4915
    ISSN 1999-4915
    DOI 10.3390/v2040900
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  9. Article ; Online: Recognition of Semaphorin Proteins by P. sordellii Lethal Toxin Reveals Principles of Receptor Specificity in Clostridial Toxins.

    Lee, Hunsang / Beilhartz, Greg L / Kucharska, Iga / Raman, Swetha / Cui, Hong / Lam, Mandy Hiu Yi / Liang, Huazhu / Rubinstein, John L / Schramek, Daniel / Julien, Jean-Philippe / Melnyk, Roman A / Taipale, Mikko

    Cell

    2020  Volume 182, Issue 2, Page(s) 345–356.e16

    Abstract: 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% ... ...

    Abstract 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.
    MeSH term(s) Animals ; Bacterial Toxins/chemistry ; Bacterial Toxins/metabolism ; Bacterial Toxins/toxicity ; Binding Sites ; CRISPR-Cas Systems/genetics ; Cell Line ; Clostridium sordellii/metabolism ; Cryoelectron Microscopy ; Edema/pathology ; Edema/prevention & control ; Female ; Humans ; Lung/drug effects ; Lung/pathology ; Mice ; Mice, Inbred C57BL ; Molecular Dynamics Simulation ; Mutagenesis, Site-Directed ; Protein Binding ; Protein Structure, Tertiary ; Recombinant Proteins/biosynthesis ; Recombinant Proteins/isolation & purification ; Recombinant Proteins/therapeutic use ; Semaphorins/chemistry ; Semaphorins/genetics ; Semaphorins/metabolism
    Chemical Substances Bacterial Toxins ; Recombinant Proteins ; Sema6a protein, mouse ; Sema6b protein, mouse ; Semaphorins
    Keywords covid19
    Language English
    Publishing date 2020-06-25
    Publishing country United States
    Document type Journal Article ; Research Support, Non-U.S. Gov't
    ZDB-ID 187009-9
    ISSN 1097-4172 ; 0092-8674
    ISSN (online) 1097-4172
    ISSN 0092-8674
    DOI 10.1016/j.cell.2020.06.005
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  10. Article ; Online: Exploiting the diphtheria toxin internalization receptor enhances delivery of proteins to lysosomes for enzyme replacement therapy.

    Sugiman-Marangos, Seiji N / Beilhartz, Greg L / Zhao, Xiaochu / Zhou, Dongxia / Hua, Rong / Kim, Peter K / Rini, James M / Minassian, Berge A / Melnyk, Roman A

    Science advances

    2020  Volume 6, Issue 50

    Abstract: 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 ... ...

    Abstract 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.
    MeSH term(s) Animals ; Brain/metabolism ; Diphtheria Toxin/metabolism ; Diphtheria Toxin/pharmacology ; Enzyme Replacement Therapy ; Lysosomes/metabolism ; Mice ; Receptor, IGF Type 2/genetics ; Receptor, IGF Type 2/metabolism ; Recombinant Proteins/metabolism
    Chemical Substances Diphtheria Toxin ; Receptor, IGF Type 2 ; Recombinant Proteins
    Language English
    Publishing date 2020-12-11
    Publishing country United States
    Document type Journal Article ; Research Support, Non-U.S. Gov't
    ZDB-ID 2810933-8
    ISSN 2375-2548 ; 2375-2548
    ISSN (online) 2375-2548
    ISSN 2375-2548
    DOI 10.1126/sciadv.abb0385
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

To top