1
|
Sun Q, Yu L, Donnelly SC, Fradin C, Thompson RT, Prato FS, Goldhawk DE. Essential magnetosome proteins MamI and MamL from magnetotactic bacteria interact in mammalian cells. Sci Rep 2024; 14:26292. [PMID: 39487238 PMCID: PMC11530650 DOI: 10.1038/s41598-024-77591-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 10/23/2024] [Indexed: 11/04/2024] Open
Abstract
To detect cellular activities deep within the body using magnetic resonance platforms, magnetosomes are the ideal model of genetically-encoded nanoparticles. These membrane-bound iron biominerals produced by magnetotactic bacteria are highly regulated by approximately 30 genes; however, the number of magnetosome genes that are essential and/or constitute the root structure upon which biominerals form is largely undefined. To examine the possibility that key magnetosome genes may interact in a foreign environment, we expressed mamI and mamL as fluorescent fusion proteins in mammalian cells. Localization and potential protein-protein interaction(s) were investigated using confocal microscopy and fluorescence correlation spectroscopy (FCS). Enhanced green fluorescent protein (EGFP)-MamI and the red fluorescent Tomato-MamL displayed distinct intracellular localization, with net-like and punctate fluorescence, respectively. Remarkably, co-expression revealed co-localization of both fluorescent fusion proteins in the same punctate pattern. An interaction between MamI and MamL was confirmed by co-immunoprecipitation. In addition, changes in EGFP-MamI distribution were accompanied by acquisition of intracellular mobility which all Tomato-MamL structures displayed. Analysis of extracts from these cells by FCS was consistent with an interaction between fluorescent fusion proteins, including an increase in particle radius. Co-localization and interaction of MamI and MamL demonstrate that select magnetosome proteins may associate in mammalian cells.
Collapse
Affiliation(s)
- Qin Sun
- Imaging, Lawson Research Institute, London, ON, Canada
- Medical Biophysics, Western University, London, ON, Canada
- Collaborative Graduate Program in Molecular Imaging, Western University, London, ON, Canada
| | - Liu Yu
- Physics & Astronomy, McMaster University, Hamilton, ON, Canada
| | | | - Cécile Fradin
- Physics & Astronomy, McMaster University, Hamilton, ON, Canada
- Biochemistry & Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - R Terry Thompson
- Imaging, Lawson Research Institute, London, ON, Canada
- Medical Biophysics, Western University, London, ON, Canada
- Medical Imaging, Western University, London, ON, Canada
- Physics & Astronomy, Western University, London, ON, Canada
| | - Frank S Prato
- Imaging, Lawson Research Institute, London, ON, Canada
- Medical Biophysics, Western University, London, ON, Canada
- Collaborative Graduate Program in Molecular Imaging, Western University, London, ON, Canada
- Medical Imaging, Western University, London, ON, Canada
| | - Donna E Goldhawk
- Imaging, Lawson Research Institute, London, ON, Canada.
- Medical Biophysics, Western University, London, ON, Canada.
- Collaborative Graduate Program in Molecular Imaging, Western University, London, ON, Canada.
- Imaging Program, Lawson Research Institute, 268 Grosvenor St. , PO Box 5777 Station B, London, ON, N6A 4V2, Canada.
| |
Collapse
|
2
|
Litberg TJ, Horowitz S. Roles of Nucleic Acids in Protein Folding, Aggregation, and Disease. ACS Chem Biol 2024; 19:809-823. [PMID: 38477936 PMCID: PMC11149768 DOI: 10.1021/acschembio.3c00695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2024]
Abstract
The role of nucleic acids in protein folding and aggregation is an area of continued research, with relevance to understanding both basic biological processes and disease. In this review, we provide an overview of the trajectory of research on both nucleic acids as chaperones and their roles in several protein misfolding diseases. We highlight key questions that remain on the biophysical and biochemical specifics of how nucleic acids have large effects on multiple proteins' folding and aggregation behavior and how this pertains to multiple protein misfolding diseases.
Collapse
Affiliation(s)
- Theodore J. Litberg
- Department of Chemistry & Biochemistry and The Knoebel Institute for Healthy Aging, University of Denver, Denver, CO, 80208, USA
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
- Center for Synthetic Biology, Northwestern University, Evanston, IL, 60208, USA
| | - Scott Horowitz
- Department of Chemistry & Biochemistry and The Knoebel Institute for Healthy Aging, University of Denver, Denver, CO, 80208, USA
| |
Collapse
|
3
|
Montgomery K, Carroll EC, Thwin AC, Quddus AY, Hodges P, Southworth DR, Gestwicki JE. Chemical Features of Polyanions Modulate Tau Aggregation and Conformational States. J Am Chem Soc 2023; 145:3926-3936. [PMID: 36753572 PMCID: PMC9951223 DOI: 10.1021/jacs.2c08004] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Indexed: 02/10/2023]
Abstract
The aggregation of tau into insoluble fibrils is a defining feature of neurodegenerative tauopathies. However, tau has a positive overall charge and is highly soluble; so, polyanions, such as heparin, are typically required to promote its aggregation in vitro. There are dozens of polyanions in living systems, and it is not clear which ones might promote this process. Here, we systematically measure the ability of 37 diverse, anionic biomolecules to initiate tau aggregation using either wild-type (WT) tau or the disease-associated P301S mutant. We find that polyanions from many different structural classes can promote fibril formation and that P301S tau is sensitive to a greater number of polyanions (28/37) than WT tau (21/37). We also find that some polyanions preferentially reduce the lag time of the aggregation reactions, while others enhance the elongation rate, suggesting that they act on partially distinct steps. From the resulting structure-activity relationships, the valency of the polyanion seems to be an important chemical feature such that anions with low valency tend to be weaker aggregation inducers, even at the same overall charge. Finally, the identity of the polyanion influences fibril morphology based on electron microscopy and limited proteolysis. These results provide insights into the crucial role of polyanion-tau interactions in modulating tau conformational dynamics with implications for understanding the tau aggregation landscape in a complex cellular environment.
Collapse
Affiliation(s)
- Kelly
M. Montgomery
- Department
of Pharmaceutical Chemistry, University
of California San Francisco, San Francisco, California 94158, United States
- The
Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, California 94158, United States
| | - Emma C. Carroll
- The
Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, California 94158, United States
| | - Aye C. Thwin
- The
Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, California 94158, United States
| | - Athena Y. Quddus
- The
Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, California 94158, United States
| | - Paige Hodges
- Department
of Pharmaceutical Chemistry, University
of California San Francisco, San Francisco, California 94158, United States
- The
Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, California 94158, United States
| | - Daniel R. Southworth
- The
Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, California 94158, United States
- Department
of Biochemistry and Biophysics, University
of California San Francisco, San Francisco, California 94158, United States
| | - Jason E. Gestwicki
- Department
of Pharmaceutical Chemistry, University
of California San Francisco, San Francisco, California 94158, United States
- The
Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, California 94158, United States
| |
Collapse
|
4
|
Hill SE, Esquivel AR, Ospina SR, Rahal LM, Dickey CA, Blair LJ. Chaperoning activity of the cyclophilin family prevents tau aggregation. Protein Sci 2022; 31:e4448. [PMID: 36305768 PMCID: PMC9597375 DOI: 10.1002/pro.4448] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/18/2022] [Accepted: 09/11/2022] [Indexed: 11/09/2022]
Abstract
Tauopathies, such as Alzheimer's disease, are characterized by the misfolding and progressive accumulation of the microtubule associated protein tau. Chaperones, tasked with maintaining protein homeostasis, can become imbalanced with age and contribute to the progression of neurodegenerative disease. Cyclophilins are a promising pool of underinvestigated chaperones with peptidyl-prolyl isomerase activity that may play protective roles in regulating tau aggregation. Using a Thioflavin T fluorescence-based assay to monitor in vitro tau aggregation, all eight cyclophilins, which include PPIA to PPIH prevent tau aggregation, with PPIB, PPIC, PPID, and PPIH showing the greatest inhibition. The low thermal stability of PPID and the strong heparin binding of PPIB undermines the simplistic interpretation of reduced tau aggregation. In a cellular model of tau accumulation, all cyclophilins, except PPID and PPIH, reduce insoluble tau. PPIB, PPIC, PPIE, and PPIF also reduce soluble tau levels with PPIC exclusively protecting cells from tau seeding. Overall, this study demonstrates cyclophilins prevent tau fibril formation and many reduce cellular insoluble tau accumulation with PPIC having the greatest potential as a molecular tool to mitigate tau seeding and accumulation.
Collapse
Affiliation(s)
- Shannon E. Hill
- USF Health Byrd Alzheimer's InstituteUniversity of South FloridaTampaFloridaUSA
- Department of Molecular MedicineUniversity of South FloridaTampaFloridaUSA
| | - Abigail R. Esquivel
- USF Health Byrd Alzheimer's InstituteUniversity of South FloridaTampaFloridaUSA
- Department of Molecular MedicineUniversity of South FloridaTampaFloridaUSA
| | - Santiago Rodriguez Ospina
- USF Health Byrd Alzheimer's InstituteUniversity of South FloridaTampaFloridaUSA
- Department of Molecular MedicineUniversity of South FloridaTampaFloridaUSA
| | - Lauren M. Rahal
- USF Health Byrd Alzheimer's InstituteUniversity of South FloridaTampaFloridaUSA
- Department of Molecular MedicineUniversity of South FloridaTampaFloridaUSA
| | - Chad A. Dickey
- USF Health Byrd Alzheimer's InstituteUniversity of South FloridaTampaFloridaUSA
- Department of Molecular MedicineUniversity of South FloridaTampaFloridaUSA
| | - Laura J. Blair
- USF Health Byrd Alzheimer's InstituteUniversity of South FloridaTampaFloridaUSA
- Department of Molecular MedicineUniversity of South FloridaTampaFloridaUSA
- Research ServiceJames A. Haley Veterans HospitalTampaFloridaUSA
| |
Collapse
|
5
|
Furuya G, Katoh H, Atsumi S, Hashimoto I, Komura D, Hatanaka R, Senga S, Hayashi S, Akita S, Matsumura H, Miura A, Mita H, Nakakido M, Nagatoishi S, Sugiyama A, Suzuki R, Konishi H, Yamamoto A, Abe H, Hiraoka N, Aoki K, Kato Y, Seto Y, Yoshimura C, Miyadera K, Tsumoto K, Ushiku T, Ishikawa S. Nucleic acid-triggered tumoral immunity propagates pH-selective therapeutic antibodies through tumor-driven epitope spreading. Cancer Sci 2022; 114:321-338. [PMID: 36136061 PMCID: PMC9807517 DOI: 10.1111/cas.15596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/09/2022] [Accepted: 09/12/2022] [Indexed: 01/07/2023] Open
Abstract
Important roles of humoral tumor immunity are often pointed out; however, precise profiles of dominant antigens and developmental mechanisms remain elusive. We systematically investigated the humoral antigens of dominant intratumor immunoglobulin clones found in human cancers. We found that approximately half of the corresponding antigens were restricted to strongly and densely negatively charged polymers, resulting in simultaneous reactivities of the antibodies to both densely sulfated glycosaminoglycans (dsGAGs) and nucleic acids (NAs). These anti-dsGAG/NA antibodies matured and expanded via intratumoral immunological driving force of innate immunity via NAs. These human cancer-derived antibodies exhibited acidic pH-selective affinity across both antigens and showed specific reactivity to diverse spectrums of human tumor cells. The antibody-drug conjugate exerted therapeutic effects against multiple cancers in vivo by targeting cell surface dsGAG antigens. This study reveals that intratumoral immunological reactions propagate tumor-oriented immunoglobulin clones and demonstrates a new therapeutic modality for the universal treatment of human malignancies.
Collapse
Affiliation(s)
- Genta Furuya
- Department of Preventive medicine, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Hiroto Katoh
- Department of Preventive medicine, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Shinichiro Atsumi
- Department of Preventive medicine, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Itaru Hashimoto
- Department of Preventive medicine, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Daisuke Komura
- Department of Preventive medicine, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Ryo Hatanaka
- Discovery and Preclinical Research DivisionTaiho Pharmaceutical Co., Ltd.IbarakiJapan
| | - Shogo Senga
- Discovery and Preclinical Research DivisionTaiho Pharmaceutical Co., Ltd.IbarakiJapan
| | - Shuto Hayashi
- Department of Preventive medicine, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Shoji Akita
- Discovery and Preclinical Research DivisionTaiho Pharmaceutical Co., Ltd.IbarakiJapan
| | - Hirofumi Matsumura
- Discovery and Preclinical Research DivisionTaiho Pharmaceutical Co., Ltd.IbarakiJapan
| | - Akihiro Miura
- Discovery and Preclinical Research DivisionTaiho Pharmaceutical Co., Ltd.IbarakiJapan
| | - Hideaki Mita
- Department of Preventive medicine, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Makoto Nakakido
- Laboratory of Medical Proteomics, Institute of Medical ScienceThe University of TokyoTokyoJapan
| | - Satoru Nagatoishi
- Laboratory of Medical Proteomics, Institute of Medical ScienceThe University of TokyoTokyoJapan
| | - Akira Sugiyama
- Laboratory of Systems Biology and MedicineResearch Center for Advanced Science and Technology, The University of TokyoTokyoJapan
| | - Ryohei Suzuki
- Department of Preventive medicine, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Hiroki Konishi
- Department of Preventive medicine, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Asami Yamamoto
- Department of Preventive medicine, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Hiroyuki Abe
- Department of Pathology, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Nobuyoshi Hiraoka
- Department of Analytical PathologyNational Cancer Center Research InstituteTokyoJapan
| | - Kazunori Aoki
- Division of Molecular and Cellular MedicineNational Cancer Center Research InstituteTokyoJapan
| | - Yasumasa Kato
- Department of Oral Function and Molecular BiologyOhu University School of DentistryFukushimaJapan
| | - Yasuyuki Seto
- Department of Gastrointestinal SurgeryGraduate School of Medicine, The University of TokyoTokyoJapan
| | - Chihoko Yoshimura
- Discovery and Preclinical Research DivisionTaiho Pharmaceutical Co., Ltd.IbarakiJapan
| | - Kazutaka Miyadera
- Discovery and Preclinical Research DivisionTaiho Pharmaceutical Co., Ltd.IbarakiJapan
| | - Kouhei Tsumoto
- Laboratory of Medical Proteomics, Institute of Medical ScienceThe University of TokyoTokyoJapan
| | - Tetsuo Ushiku
- Department of Pathology, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Shumpei Ishikawa
- Department of Preventive medicine, Graduate School of MedicineThe University of TokyoTokyoJapan
| |
Collapse
|
6
|
Bhaumik SK, Banerjee S. Highly sensitive and ratiometric luminescence sensing of heparin through templated cyanostilbene assemblies. Analyst 2021; 146:2194-2202. [PMID: 33587729 DOI: 10.1039/d0an01808b] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The assembly of organic dyes on bio-molecular templates is an attractive strategy for the creation of bio-materials with intriguing optical properties. This principle is exploited here for the detection of polyanion heparin, a known anticoagulant, by employing di-cationic cyanostilbene derivatives with inherent aggregation induced emission (AIE) features. The cyanostilbene derivatives exhibited weak cyan-blue monomeric emissions in solutions but upon electrostatic co-assembly with heparin, formed highly luminescent clusters on the polyanion surface. The cyanostilbene chromophores in the clusters exhibited greenish-yellow excimer emissions with remarkably longer life-times (up to 70-fold) and higher quantum yields (up to 85-fold) compared to their aqueous solutions. This led to heparin detection in aqueous buffer in low nanomolar concentrations. Additionally, and more importantly, a ratiometric detection of heparin was achieved in highly competitive media such as 50% human serum and 60% human plasma in medically relevant concentrations.
Collapse
Affiliation(s)
- Shubhra Kanti Bhaumik
- The Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur 741246, Nadia, India.
| | | |
Collapse
|
7
|
Aarum J, Cabrera CP, Jones TA, Rajendran S, Adiutori R, Giovannoni G, Barnes MR, Malaspina A, Sheer D. Enzymatic degradation of RNA causes widespread protein aggregation in cell and tissue lysates. EMBO Rep 2020; 21:e49585. [PMID: 32945072 PMCID: PMC7534620 DOI: 10.15252/embr.201949585] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 06/28/2020] [Accepted: 07/30/2020] [Indexed: 12/12/2022] Open
Abstract
Most proteins in cell and tissue lysates are soluble. We show here that in lysate from human neurons, more than 1,300 proteins are maintained in a soluble and functional state by association with endogenous RNA, as degradation of RNA invariably leads to protein aggregation. The majority of these proteins lack conventional RNA‐binding domains. Using synthetic oligonucleotides, we identify the importance of nucleic acid structure, with single‐stranded pyrimidine‐rich bulges or loops surrounded by double‐stranded regions being particularly efficient in the maintenance of protein solubility. These experiments also identify an apparent one‐to‐one protein‐nucleic acid stoichiometry. Furthermore, we show that protein aggregates isolated from brain tissue from Amyotrophic Lateral Sclerosis patients can be rendered soluble after refolding by both RNA and synthetic oligonucleotides. Together, these findings open new avenues for understanding the mechanism behind protein aggregation and shed light on how certain proteins remain soluble.
Collapse
Affiliation(s)
- Johan Aarum
- Barts and The London School of Medicine and Dentistry, Blizard Institute, Queen Mary University of London, London, UK
| | - Claudia P Cabrera
- Barts and The London NIHR Cardiovascular Biomedical Research Centre, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Tania A Jones
- Barts and The London School of Medicine and Dentistry, Blizard Institute, Queen Mary University of London, London, UK
| | - Shiron Rajendran
- Barts and The London School of Medicine and Dentistry, Blizard Institute, Queen Mary University of London, London, UK
| | - Rocco Adiutori
- Barts and The London School of Medicine and Dentistry, Blizard Institute, Queen Mary University of London, London, UK
| | - Gavin Giovannoni
- Barts and The London School of Medicine and Dentistry, Blizard Institute, Queen Mary University of London, London, UK
| | - Michael R Barnes
- Barts and The London NIHR Cardiovascular Biomedical Research Centre, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Andrea Malaspina
- Barts and The London School of Medicine and Dentistry, Blizard Institute, Queen Mary University of London, London, UK
| | - Denise Sheer
- Barts and The London School of Medicine and Dentistry, Blizard Institute, Queen Mary University of London, London, UK
| |
Collapse
|
8
|
Marson D, Laurini E, Aulic S, Fermeglia M, Pricl S. Perceptions and Misconceptions in Molecular Recognition: Key Factors in Self-Assembling Multivalent (SAMul) Ligands/Polyanions Selectivity. Molecules 2020; 25:molecules25041003. [PMID: 32102359 PMCID: PMC7070608 DOI: 10.3390/molecules25041003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 02/21/2020] [Accepted: 02/21/2020] [Indexed: 01/02/2023] Open
Abstract
Biology is dominated by polyanions (cell membranes, nucleic acids, and polysaccharides just to name a few), and achieving selective recognition between biological polyanions and synthetic systems currently constitutes a major challenge in many biomedical applications, nanovectors-assisted gene delivery being a prime example. This review work summarizes some of our recent efforts in this field; in particular, by using a combined experimental/computation approach, we investigated in detail some critical aspects in self-assembled nanomicelles and two major polyanions—DNA and heparin.
Collapse
Affiliation(s)
- Domenico Marson
- Molecular Biology and Nanotechnology Laboratory (MolBNL@UniTS), Department of Engineering and Architecture, University of Trieste, 34127 Trieste, Italy; (D.M.); (S.A.); (M.F.); (S.P.)
| | - Erik Laurini
- Molecular Biology and Nanotechnology Laboratory (MolBNL@UniTS), Department of Engineering and Architecture, University of Trieste, 34127 Trieste, Italy; (D.M.); (S.A.); (M.F.); (S.P.)
- Correspondence: ; Tel.: +39-040-558-3432
| | - Suzana Aulic
- Molecular Biology and Nanotechnology Laboratory (MolBNL@UniTS), Department of Engineering and Architecture, University of Trieste, 34127 Trieste, Italy; (D.M.); (S.A.); (M.F.); (S.P.)
| | - Maurizio Fermeglia
- Molecular Biology and Nanotechnology Laboratory (MolBNL@UniTS), Department of Engineering and Architecture, University of Trieste, 34127 Trieste, Italy; (D.M.); (S.A.); (M.F.); (S.P.)
| | - Sabrina Pricl
- Molecular Biology and Nanotechnology Laboratory (MolBNL@UniTS), Department of Engineering and Architecture, University of Trieste, 34127 Trieste, Italy; (D.M.); (S.A.); (M.F.); (S.P.)
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland
| |
Collapse
|
9
|
Choi SI. A Simple Principle for Understanding the Combined Cellular Protein Folding and Aggregation. Curr Protein Pept Sci 2020; 21:3-21. [DOI: 10.2174/1389203720666190725114550] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 07/09/2019] [Accepted: 07/10/2019] [Indexed: 12/27/2022]
Abstract
Proteins can undergo kinetic/thermodynamic partitioning between folding and aggregation. Proper protein folding and thermodynamic stability are crucial for aggregation inhibition. Thus, proteinfolding principles have been widely believed to consistently underlie aggregation as a consequence of conformational change. However, this prevailing view appears to be challenged by the ubiquitous phenomena that the intrinsic and extrinsic factors including cellular macromolecules can prevent aggregation, independently of (even with sacrificing) protein folding rate and stability. This conundrum can be definitely resolved by ‘a simple principle’ based on a rigorous distinction between protein folding and aggregation: aggregation can be controlled by affecting the intermolecular interactions for aggregation, independently of the intramolecular interactions for protein folding. Aggregation is beyond protein folding. A unifying model that can conceptually reconcile and underlie the seemingly contradictory observations is described here. This simple principle highlights, in particular, the importance of intermolecular repulsive forces against aggregation, the magnitude of which can be correlated with the size and surface properties of molecules. The intermolecular repulsive forces generated by the common intrinsic properties of cellular macromolecules including chaperones, such as their large excluded volume and surface charges, can play a key role in preventing the aggregation of their physically connected polypeptides, thus underlying the generic intrinsic chaperone activity of soluble cellular macromolecules. Such intermolecular repulsive forces of bulky cellular macromolecules, distinct from protein conformational change and attractive interactions, could be the puzzle pieces for properly understanding the combined cellular protein folding and aggregation including how proteins can overcome their metastability to amyloid fibrils in vivo.
Collapse
Affiliation(s)
- Seong Il Choi
- Department of Biochemistry and Biophysics, Stockholm University, SE-106 91 Stockholm, Sweden
| |
Collapse
|
10
|
Mizrak A, Morgan DO. Polyanions provide selective control of APC/C interactions with the activator subunit. Nat Commun 2019; 10:5807. [PMID: 31862931 PMCID: PMC6925294 DOI: 10.1038/s41467-019-13864-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 12/04/2019] [Indexed: 01/18/2023] Open
Abstract
Transient interactions between the anaphase-promoting complex/cyclosome (APC/C) and its activator subunit Cdc20 or Cdh1 generate oscillations in ubiquitylation activity necessary to maintain the order of cell cycle events. Activator binds the APC/C with high affinity and exhibits negligible dissociation kinetics in vitro, and it is not clear how the rapid turnover of APC/C-activator complexes is achieved in vivo. Here, we describe a mechanism that controls APC/C-activator interactions based on the availability of substrates. We find that APC/C-activator dissociation is stimulated by abundant cellular polyanions such as nucleic acids and polyphosphate. Polyanions also interfere with substrate ubiquitylation. However, engagement with high-affinity substrate blocks the inhibitory effects of polyanions on activator binding and APC/C activity. We propose that this mechanism amplifies the effects of substrate affinity on APC/C function, stimulating processive ubiquitylation of high-affinity substrates and suppressing ubiquitylation of low-affinity substrates.
Collapse
Affiliation(s)
- Arda Mizrak
- Department of Physiology, University of California, San Francisco, CA, 94143, USA
| | - David O Morgan
- Department of Physiology, University of California, San Francisco, CA, 94143, USA.
| |
Collapse
|
11
|
Ghiselli G. Heparin Binding Proteins as Therapeutic Target: An Historical Account and Current Trends. MEDICINES (BASEL, SWITZERLAND) 2019; 6:E80. [PMID: 31362364 PMCID: PMC6789896 DOI: 10.3390/medicines6030080] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 07/16/2019] [Accepted: 07/18/2019] [Indexed: 12/16/2022]
Abstract
The polyanionic nature and the ability to interact with proteins with different affinities are properties of sulfated glycosaminoglycans (GAGs) that determine their biological function. In designing drugs affecting the interaction of proteins with GAGs the challenge has been to generate agents with high binding specificity. The example to emulated has been a heparin-derived pentasaccharide that binds to antithrombin-III with high affinity. However, the portability of this model to other biological situations is questioned on several accounts. Because of their structural flexibility, oligosaccharides with different sulfation and uronic acid conformation can display the same binding proficiency to different proteins and produce comparable biological effects. This circumstance represents a formidable obstacle to the design of drugs based on the heparin scaffold. The conceptual framework discussed in this article is that through a direct intervention on the heparin-binding functionality of proteins is possible to achieve a high degree of action specificity. This objective is currently pursued through two strategies. The first makes use of small molecules for which in the text we provide examples from past and present literature concerning angiogenic factors and enzymes. The second approach entails the mutagenesis of the GAG-binding site of proteins as a means to generate a new class of biologics of therapeutic interest.
Collapse
Affiliation(s)
- Giancarlo Ghiselli
- Independent Researcher, 1326 Spruce Street Suite 706, Philadephia, PA 19107, USA.
| |
Collapse
|
12
|
Nunes QM, Su D, Brownridge PJ, Simpson DM, Sun C, Li Y, Bui TP, Zhang X, Huang W, Rigden DJ, Beynon RJ, Sutton R, Fernig DG. The heparin-binding proteome in normal pancreas and murine experimental acute pancreatitis. PLoS One 2019; 14:e0217633. [PMID: 31211768 PMCID: PMC6581253 DOI: 10.1371/journal.pone.0217633] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 05/15/2019] [Indexed: 02/07/2023] Open
Abstract
Acute pancreatitis (AP) is acute inflammation of the pancreas, mainly caused by gallstones and alcohol, driven by changes in communication between cells. Heparin-binding proteins (HBPs) play a central role in health and diseases. Therefore, we used heparin affinity proteomics to identify extracellular HBPs in pancreas and plasma of normal mice and in a caerulein mouse model of AP. Many new extracellular HBPs (360) were discovered in the pancreas, taking the total number of HBPs known to 786. Extracellular pancreas HBPs form highly interconnected protein-protein interaction networks in both normal pancreas (NP) and AP. Thus, HBPs represent an important set of extracellular proteins with significant regulatory potential in the pancreas. HBPs in NP are associated with biological functions such as molecular transport and cellular movement that underlie pancreatic homeostasis. However, in AP HBPs are associated with additional inflammatory processes such as acute phase response signalling, complement activation and mitochondrial dysfunction, which has a central role in the development of AP. Plasma HBPs in AP included known AP biomarkers such as serum amyloid A, as well as emerging targets such as histone H2A. Other HBPs such as alpha 2-HS glycoprotein (AHSG) and histidine-rich glycoprotein (HRG) need further investigation for potential applications in the management of AP. Pancreas HBPs are extracellular and so easily accessible and are potential drug targets in AP, whereas plasma HBPs represent potential biomarkers for AP. Thus, their identification paves the way to determine which HBPs may have potential applications in the management of AP.
Collapse
Affiliation(s)
- Quentin M. Nunes
- Liverpool Pancreatitis Research Group, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
- * E-mail:
| | - Dunhao Su
- Liverpool Pancreatitis Research Group, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
- Department of Biochemistry, Institute of Integrative Biology, Biosciences Building, University of Liverpool, Liverpool, United Kingdom
| | - Philip J. Brownridge
- Department of Biochemistry, Institute of Integrative Biology, Biosciences Building, University of Liverpool, Liverpool, United Kingdom
- Centre for Proteome Research, Institute of Integrative Biology, Biosciences Building, University of Liverpool, Liverpool, United Kingdom
| | - Deborah M. Simpson
- Department of Biochemistry, Institute of Integrative Biology, Biosciences Building, University of Liverpool, Liverpool, United Kingdom
- Centre for Proteome Research, Institute of Integrative Biology, Biosciences Building, University of Liverpool, Liverpool, United Kingdom
| | - Changye Sun
- Department of Biochemistry, Institute of Integrative Biology, Biosciences Building, University of Liverpool, Liverpool, United Kingdom
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, Henan, China
| | - Yong Li
- Department of Biochemistry, Institute of Integrative Biology, Biosciences Building, University of Liverpool, Liverpool, United Kingdom
- College of Life and Environmental Science, Wen Zhou University, Wenzhou, China
| | - Thao P. Bui
- Department of Biochemistry, Institute of Integrative Biology, Biosciences Building, University of Liverpool, Liverpool, United Kingdom
| | - Xiaoying Zhang
- Liverpool Pancreatitis Research Group, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Wei Huang
- Liverpool Pancreatitis Research Group, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
- Department of Integrated Traditional Chinese and Western Medicine, Sichuan Provincial Pancreatitis Centre and West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, China
| | - Daniel J. Rigden
- Department of Biochemistry, Institute of Integrative Biology, Biosciences Building, University of Liverpool, Liverpool, United Kingdom
| | - Robert J. Beynon
- Department of Biochemistry, Institute of Integrative Biology, Biosciences Building, University of Liverpool, Liverpool, United Kingdom
- Centre for Proteome Research, Institute of Integrative Biology, Biosciences Building, University of Liverpool, Liverpool, United Kingdom
| | - Robert Sutton
- Liverpool Pancreatitis Research Group, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
| | - David G. Fernig
- Liverpool Pancreatitis Research Group, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
- Department of Biochemistry, Institute of Integrative Biology, Biosciences Building, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
13
|
Semenyuk P, Muronetz V. Protein Interaction with Charged Macromolecules: From Model Polymers to Unfolded Proteins and Post-Translational Modifications. Int J Mol Sci 2019; 20:E1252. [PMID: 30871103 PMCID: PMC6429204 DOI: 10.3390/ijms20051252] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 03/05/2019] [Accepted: 03/07/2019] [Indexed: 12/18/2022] Open
Abstract
Interaction of proteins with charged macromolecules is involved in many processes in cells. Firstly, there are many naturally occurred charged polymers such as DNA and RNA, polyphosphates, sulfated glycosaminoglycans, etc., as well as pronouncedly charged proteins such as histones or actin. Electrostatic interactions are also important for "generic" proteins, which are not generally considered as polyanions or polycations. Finally, protein behavior can be altered due to post-translational modifications such as phosphorylation, sulfation, and glycation, which change a local charge of the protein region. Herein we review molecular modeling for the investigation of such interactions, from model polyanions and polycations to unfolded proteins. We will show that electrostatic interactions are ubiquitous, and molecular dynamics simulations provide an outstanding opportunity to look inside binding and reveal the contribution of electrostatic interactions. Since a molecular dynamics simulation is only a model, we will comprehensively consider its relationship with the experimental data.
Collapse
Affiliation(s)
- Pavel Semenyuk
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119234 Moscow, Russia.
| | - Vladimir Muronetz
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119234 Moscow, Russia.
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia.
| |
Collapse
|
14
|
The polyanions heparin and suramin impede binding of free adenine to a DNA glycosylase from C. pseudotuberculosis. Int J Biol Macromol 2019; 125:459-468. [DOI: 10.1016/j.ijbiomac.2018.12.067] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 12/05/2018] [Accepted: 12/06/2018] [Indexed: 12/31/2022]
|
15
|
Eberle RJ, Kawai LA, de Moraes FR, Olivier D, do Amaral MS, Tasic L, Arni RK, Coronado MA. Inhibition of thioredoxin A1 from Corynebacterium pseudotuberculosis by polyanions and flavonoids. Int J Biol Macromol 2018; 117:1066-1073. [DOI: 10.1016/j.ijbiomac.2018.06.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 06/04/2018] [Accepted: 06/05/2018] [Indexed: 11/17/2022]
|
16
|
Iliev D, Strandskog G, Nepal A, Aspar A, Olsen R, Jørgensen J, Wolfson D, Ahluwalia BS, Handzhiyski J, Mironova R. Stimulation of exosome release by extracellular DNA is conserved across multiple cell types. FEBS J 2018; 285:3114-3133. [PMID: 29953723 DOI: 10.1111/febs.14601] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 05/18/2018] [Accepted: 06/26/2018] [Indexed: 01/01/2023]
Abstract
Exosomes are distinguished from other types of extracellular vesicles by their small and relatively uniform size (30-100 nm) and their composition which reflects their endo-lysosomal origin. Involvement of these extracellular organelles in intercellular communication and their implication in pathological conditions has fuelled intensive research on mammalian exosomes; however, currently, very little is known about exosomes in lower vertebrates. Here we show that, in primary cultures of head kidney leukocytes from Atlantic salmon (Salmo salar), phosphorothioate CpG oligodeoxynucleotides induce secretion of vesicles with characteristics very similar to these of mammalian exosomes. Further experiments revealed that the oligonucleotide-induced exosome secretion did not depend on the CpG motifs but it relied on the phosphorothioate modification of the internucleotide linkage. Exosome secretion was also induced by genomic bacterial and eukaryotic DNA in toll-like receptor 9-negative piscine and human cell lines demonstrating that this is a phylogenetically conserved phenomenon which does not depend on activation of immune signaling pathways. In addition to exosomes, stimulation with phosphorothioate oligonucleotides and genomic DNA induced secretion of LC3B-II, an autophagosome marker, which was associated with vesicles of diverse size and morphology, possibly derived from autophagosome-related intracellular compartments. Overall, this work reveals a previously unrecognized biological activity of phosphorothioate ODNs and genomic DNA - their capacity to induce secretion of exosomes and other types of extracellular vesicles. This finding might help shed light on the side effects of therapeutic phosphorothioate oligodeoxynucleotides and the biological activity of extracellular genomic DNA which is often upregulated in pathological conditions.
Collapse
Affiliation(s)
- Dimitar Iliev
- Norwegian College of Fishery Science, UiT The Arctic University of Norway, Tromsø, Norway
| | - Guro Strandskog
- Norwegian College of Fishery Science, UiT The Arctic University of Norway, Tromsø, Norway
| | - Arpita Nepal
- Norwegian College of Fishery Science, UiT The Arctic University of Norway, Tromsø, Norway
| | - Augusta Aspar
- Institute of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| | - Randi Olsen
- Institute of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| | - Jorunn Jørgensen
- Norwegian College of Fishery Science, UiT The Arctic University of Norway, Tromsø, Norway
| | - Deanna Wolfson
- Department of Physics and Technology, UiT The Arctic University of Norway, Tromsø, Norway
| | | | - Jordan Handzhiyski
- Department of Gene Regulation, Institute of Molecular Biology 'Roumen Tsanev', Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Roumyana Mironova
- Department of Gene Regulation, Institute of Molecular Biology 'Roumen Tsanev', Bulgarian Academy of Sciences, Sofia, Bulgaria
| |
Collapse
|
17
|
Thornalley KA, Laurini E, Pricl S, Smith DK. Enantiomeric and Diastereomeric Self‐Assembled Multivalent Nanostructures: Understanding the Effects of Chirality on Binding to Polyanionic Heparin and DNA. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201803298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
| | - Erik Laurini
- Simulation Engineering (MOSE) Laboratory Department of Engineering and Architectures (DEA) University of Trieste 34127 Trieste Italy
| | - Sabrina Pricl
- Simulation Engineering (MOSE) Laboratory Department of Engineering and Architectures (DEA) University of Trieste 34127 Trieste Italy
| | - David K. Smith
- Department of Chemistry University of York Heslington York YO10 5DD UK
| |
Collapse
|
18
|
Thornalley KA, Laurini E, Pricl S, Smith DK. Enantiomeric and Diastereomeric Self‐Assembled Multivalent Nanostructures: Understanding the Effects of Chirality on Binding to Polyanionic Heparin and DNA. Angew Chem Int Ed Engl 2018; 57:8530-8534. [DOI: 10.1002/anie.201803298] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 05/11/2018] [Indexed: 01/09/2023]
Affiliation(s)
| | - Erik Laurini
- Simulation Engineering (MOSE) Laboratory Department of Engineering and Architectures (DEA) University of Trieste 34127 Trieste Italy
| | - Sabrina Pricl
- Simulation Engineering (MOSE) Laboratory Department of Engineering and Architectures (DEA) University of Trieste 34127 Trieste Italy
| | - David K. Smith
- Department of Chemistry University of York Heslington York YO10 5DD UK
| |
Collapse
|
19
|
Smith DK. From fundamental supramolecular chemistry to self-assembled nanomaterials and medicines and back again – how Sam inspired SAMul. Chem Commun (Camb) 2018; 54:4743-4760. [DOI: 10.1039/c8cc01753k] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Personal inspiration led to the development of a programme of research targeting the use of self-assembled systems in nanomedicine, which in the process of approaching a range of applications has uncovered new fundamental concepts in supramolecular science.
Collapse
|
20
|
Chongsiriwatana NP, Lin JS, Kapoor R, Wetzler M, Rea JAC, Didwania MK, Contag CH, Barron AE. Intracellular biomass flocculation as a key mechanism of rapid bacterial killing by cationic, amphipathic antimicrobial peptides and peptoids. Sci Rep 2017; 7:16718. [PMID: 29196622 PMCID: PMC5711933 DOI: 10.1038/s41598-017-16180-0] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 11/08/2017] [Indexed: 12/19/2022] Open
Abstract
Many organisms rely on antimicrobial peptides (AMPs) as a first line of defense against pathogens. In general, most AMPs are thought to kill bacteria by binding to and disrupting cell membranes. However, certain AMPs instead appear to inhibit biomacromolecule synthesis, while causing less membrane damage. Despite an unclear understanding of mechanism(s), there is considerable interest in mimicking AMPs with stable, synthetic molecules. Antimicrobial N-substituted glycine (peptoid) oligomers ("ampetoids") are structural, functional and mechanistic analogs of helical, cationic AMPs, which offer broad-spectrum antibacterial activity and better therapeutic potential than peptides. Here, we show through quantitative studies of membrane permeabilization, electron microscopy, and soft X-ray tomography that both AMPs and ampetoids trigger extensive and rapid non-specific aggregation of intracellular biomacromolecules that correlates with microbial death. We present data demonstrating that ampetoids are "fast killers", which rapidly aggregate bacterial ribosomes in vitro and in vivo. We suggest intracellular biomass flocculation is a key mechanism of killing for cationic, amphipathic AMPs, which may explain why most AMPs require micromolar concentrations for activity, show significant selectivity for killing bacteria over mammalian cells, and finally, why development of resistance to AMPs is less prevalent than developed resistance to conventional antibiotics.
Collapse
Affiliation(s)
- Nathaniel P Chongsiriwatana
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, Illinois, United States
| | - Jennifer S Lin
- Department of Bioengineering, Stanford University, Stanford, California, United States
| | - Rinki Kapoor
- Biophysics Program, Stanford University, Stanford, California, United States
| | - Modi Wetzler
- Department of Bioengineering, Stanford University, Stanford, California, United States
| | - Jennifer A C Rea
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, Illinois, United States
| | - Maruti K Didwania
- Department of Bioengineering, Stanford University, Stanford, California, United States
| | - Christopher H Contag
- Departments of Microbiology and Immunology, Pediatrics, and Radiology, Stanford University, Stanford, California, United States
| | - Annelise E Barron
- Department of Bioengineering, Stanford University, Stanford, California, United States.
| |
Collapse
|
21
|
Minsky BB, Dubin PL, Kaltashov IA. Electrostatic Forces as Dominant Interactions Between Proteins and Polyanions: an ESI MS Study of Fibroblast Growth Factor Binding to Heparin Oligomers. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2017; 28:758-767. [PMID: 28211013 PMCID: PMC5808462 DOI: 10.1007/s13361-017-1596-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Revised: 12/26/2016] [Accepted: 01/04/2017] [Indexed: 05/24/2023]
Abstract
The interactions between fibroblast growth factors (FGFs) and their receptors (FGFRs) are facilitated by heparan sulfate (HS) and heparin (Hp), highly sulfated biological polyelectrolytes. The molecular basis of FGF interactions with these polyelectrolytes is highly complex due to the structural heterogeneity of HS/Hp, and many details still remain elusive, especially the significance of charge density and minimal chain length of HS/Hp in growth factor recognition and multimerization. In this work, we use electrospray ionization mass spectrometry (ESI MS) to investigate the association of relatively homogeneous oligoheparins (octamer, dp8, and decamer, dp10) with acidic fibroblast growth factor (FGF-1). This growth factor forms 1:1, 2:1, and 3:1 protein/heparinoid complexes with both dp8 and dp10, and the fraction of bound protein is highly dependent on protein/heparinoid molar ratio. Multimeric complexes are preferentially formed on the highly sulfated Hp oligomers. Although a variety of oligomers appear to be binding-competent, there is a strong correlation between the affinity and the overall level of sulfation (the highest charge density polyanions binding FGF most strongly via multivalent interactions). These results show that the interactions between FGF-1 and Hp oligomers are primarily directed by electrostatics, and also demonstrate the power of ESI MS as a tool to study multiple binding equilibria between proteins and structurally heterogeneous polyanions. Graphical Abstract ᅟ.
Collapse
Affiliation(s)
- Burcu Baykal Minsky
- Department of Chemistry, University of Massachusetts-Amherst, Amherst, MA, 01003, USA
| | - Paul L Dubin
- Department of Chemistry, University of Massachusetts-Amherst, Amherst, MA, 01003, USA
| | - Igor A Kaltashov
- Department of Chemistry, University of Massachusetts-Amherst, Amherst, MA, 01003, USA.
| |
Collapse
|
22
|
FDA-Approved Oligonucleotide Therapies in 2017. Mol Ther 2017; 25:1069-1075. [PMID: 28366767 DOI: 10.1016/j.ymthe.2017.03.023] [Citation(s) in RCA: 467] [Impact Index Per Article: 58.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 03/08/2017] [Accepted: 03/15/2017] [Indexed: 01/16/2023] Open
Abstract
Oligonucleotides (oligos) have been under clinical development for approximately the past 30 years, beginning with antisense oligonucleotides (ASOs) and apatmers and followed about 15 years ago by siRNAs. During that lengthy period of time, numerous clinical trials have been performed and thousands of trial participants accrued onto studies. Of all the molecules evaluated as of January 2017, the regulatory authorities assessed that six provided clear clinical benefit in rigorously controlled trials. The story of these six is given in this review.
Collapse
|
23
|
Vieira VMP, Liljeström V, Posocco P, Laurini E, Pricl S, Kostiainen MA, Smith DK. Emergence of highly-ordered hierarchical nanoscale aggregates on electrostatic binding of self-assembled multivalent (SAMul) cationic micelles with polyanionic heparin. J Mater Chem B 2017; 5:341-347. [DOI: 10.1039/c6tb02512a] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Self assembled cationic micelles form well-defined structurally ordered hierarchical nanoscale aggregates on interaction with polyanionic heparin in solution.
Collapse
Affiliation(s)
| | - Ville Liljeström
- Biohybrid Materials
- Department of Biotechnology and Chemical Technology
- Aalto University
- 00076 Aalto
- Finland
| | - Paola Posocco
- Simulation Engineering (MOSE) Laboratory
- Department of Engineering and Architectures (DEA)
- University of Trieste
- Trieste
- Italy
| | - Erik Laurini
- Simulation Engineering (MOSE) Laboratory
- Department of Engineering and Architectures (DEA)
- University of Trieste
- Trieste
- Italy
| | - Sabrina Pricl
- Simulation Engineering (MOSE) Laboratory
- Department of Engineering and Architectures (DEA)
- University of Trieste
- Trieste
- Italy
| | - Mauri A. Kostiainen
- Biohybrid Materials
- Department of Biotechnology and Chemical Technology
- Aalto University
- 00076 Aalto
- Finland
| | | |
Collapse
|
24
|
Xu Y, Liu M, Faisal M, Si Y, Guo Y. Selective protein complexation and coacervation by polyelectrolytes. Adv Colloid Interface Sci 2017; 239:158-167. [PMID: 27378068 DOI: 10.1016/j.cis.2016.06.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 06/03/2016] [Indexed: 12/17/2022]
Abstract
This review discusses the possible relationship between protein charge anisotropy, protein binding affinity, polymer structure, and selective phase separation. We hope that a fundamental understanding of primarily electrostatically driven protein-polyelectrolyte (PE) interactions can enable the prediction of selective protein binding, and hence selective coacervation through non-specific electrostatics. Such research will partially challenge the assumption that specific binding has to be realized through specific binding sites with a variety of short-range interactions and some geometric match. More specifically, the recent studies on selective binding of proteins by polyelectrolytes were examined from different assemblies in addition to the electrostatic features of proteins and PEs. At the end, the optimization of phase separation based on binding affinity for selective coacervation and some considerations relevant to using PEs for protein purification were also overviewed.
Collapse
Affiliation(s)
- Yisheng Xu
- State Key Laboratory of Chemical Engineering, East China University of Science and Technology, Shanghai 200237, China; Engineering Research Center of Materials Chemical Engineering of Xinjiang Bintuan, Shihezi University, Xinjiang 832000, China.
| | - Miaomiao Liu
- State Key Laboratory of Chemical Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Mostufa Faisal
- State Key Laboratory of Chemical Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Yi Si
- Department of Cardiovascular Surgery, Xinhua Hospital Affiliated of Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Yanchuan Guo
- Technical Institute of Physics and Chemistry, Chinese Academy of Science, Beijing 100190,China.
| |
Collapse
|
25
|
Rodrigo AC, Laurini E, Vieira VMP, Pricl S, Smith DK. Effect of buffer at nanoscale molecular recognition interfaces – electrostatic binding of biological polyanions. Chem Commun (Camb) 2017; 53:11580-11583. [DOI: 10.1039/c7cc07413a] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The electrostatic binding of polyanionic heparin by cationic receptors is highly dependent on the buffer in which the binding assay is carried out.
Collapse
Affiliation(s)
| | - Erik Laurini
- Simulation Engineering (MOSE) Laboratory
- Department of Engineering and Architectures (DEA)
- University of Trieste
- Trieste
- Italy
| | | | - Sabrina Pricl
- Simulation Engineering (MOSE) Laboratory
- Department of Engineering and Architectures (DEA)
- University of Trieste
- Trieste
- Italy
| | | |
Collapse
|
26
|
Ngo T, Miao X, Robinson DN, Zhou QQ. An RNA-binding protein, RNP-1, protects microtubules from nocodazole and localizes to the leading edge during cytokinesis and cell migration in Dictyostelium cells. Acta Pharmacol Sin 2016; 37:1449-1457. [PMID: 27569394 DOI: 10.1038/aps.2016.57] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 03/21/2016] [Indexed: 01/02/2023] Open
Abstract
AIM RNA-binding proteins are a large group of regulators (800-1000 in humans), some of which play significant roles in mRNA local translation. In this study, we analyzed the functions of the protein RNP-1, which was previously discovered in a genetic selection screen for nocodazole suppression. METHODS The growth rates and the microtubule networks of Dictyostelium cells were assessed with or without nocodazole (10 μmol/L) in suspension culture. Fluorescent images of RNP-1-GFP and RFP-tubulin were captured when cells were undergoing cytokinesis, then the GFP signal intensity and distance to the nearest centrosome were analyzed by using a computer program written in Matlab®. The RNP-1-GFP-expresseding cells were polarized, and the time-lapse images of cells were captured when cells were chemotaxing to a cAMP source. RESULTS Over-expression of RNP-1 rescued the growth defects caused by the microtubule-destabilizing agent nocodazole. Over-expression of RNP-1 protected microtubules from nocodazole treatment. In cells undergoing cytokinesis, the RNP-1 protein was localized to the polar regions of the cell cortex, and protein levels decreased proportionally as the power of the distance from the cell cortex to the nearest centrosome. In chemotactic cells, the RNP-1 protein localized to the leading edge of moving cells. Sequence analysis revealed that RNP-1 has two RNA-binding domains and is related to cytosolic poly(A)-binding proteins (PABPCs) in humans. CONCLUSION RNP-1 has roles in protecting microtubules and in directing cortical movement during cytokinesis and cell migration in Dictyostelium cells. The sequence similarity of RNP-1 to human PABPCs suggests that PABPCs may have similar functions in mammalian cells, perhaps in regulating microtubule dynamics and functions during cortical movement in cytokinesis and cell migration.
Collapse
|
27
|
Affiliation(s)
- Ching W. Chan
- Department of Chemistry, University of York, York, UK
| | | |
Collapse
|
28
|
Fechner LE, Albanyan B, Vieira VMP, Laurini E, Posocco P, Pricl S, Smith DK. Electrostatic binding of polyanions using self-assembled multivalent (SAMul) ligand displays - structure-activity effects on DNA/heparin binding. Chem Sci 2016; 7:4653-4659. [PMID: 30155113 PMCID: PMC6013769 DOI: 10.1039/c5sc04801j] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 04/07/2016] [Indexed: 01/11/2023] Open
Abstract
This paper reports that modifying the ligands in self-assembled multivalent (SAMul) displays has an impact on apparent binding selectivity towards two nanoscale biological polyanions - heparin and DNA. For the nanostructures assayed here, spermidine ligands are optimal for heparin binding but spermine ligands are preferred for DNA. Probing subtle differences in such nanoscale binding interfaces is a significant challenge, and as such, several experimental binding assays - competition assays and isothermal calorimetry - are employed to confirm differences in affinity and provide thermodynamic insights. Given the dynamic nature and hierarchical binding processes involved in SAMul systems, we employed multiscale modelling to propose reasons for the origins of polyanion selectivity differences. The modelling results, when expressed in thermodynamic terms and compared with the experimental data, suggest that DNA is a shape-persistent polyanion, and selectivity originates only from ligand preferences, whereas heparin is more flexible and adaptive, and as such, actively reinforces ligand preferences. As such, this study suggests that inherent differences between polyanions may underpin subtle binding selectivity differences, and that even simple electrostatic interfaces such as these can have a degree of tunability, which has implications for biological control and regulation on the nanoscale.
Collapse
Affiliation(s)
- Loryn E Fechner
- Department of Chemistry , University of York , Heslington , York , YO10 5DD , UK .
| | - Buthaina Albanyan
- Department of Chemistry , University of York , Heslington , York , YO10 5DD , UK .
| | - Vânia M P Vieira
- Department of Chemistry , University of York , Heslington , York , YO10 5DD , UK .
| | - Erik Laurini
- Simulation Engineering (MOSE) Laboratory , Department of Engineering and Architectures (DEA) , University of Trieste , Trieste , 34127 , Italy .
| | - Paola Posocco
- Simulation Engineering (MOSE) Laboratory , Department of Engineering and Architectures (DEA) , University of Trieste , Trieste , 34127 , Italy .
| | - Sabrina Pricl
- Simulation Engineering (MOSE) Laboratory , Department of Engineering and Architectures (DEA) , University of Trieste , Trieste , 34127 , Italy .
| | - David K Smith
- Department of Chemistry , University of York , Heslington , York , YO10 5DD , UK .
| |
Collapse
|
29
|
Chan CW, Laurini E, Posocco P, Pricl S, Smith DK. Chiral recognition at self-assembled multivalent (SAMul) nanoscale interfaces – enantioselectivity in polyanion binding. Chem Commun (Camb) 2016; 52:10540-3. [DOI: 10.1039/c6cc04470k] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
We investigate structure–activity effect relationships at the nanoscale chiral molecular recognition interface between enantiomeric self-assembled multivalent (SAMul) systems and biological polyanions, heparin and DNA.
Collapse
Affiliation(s)
| | - Erik Laurini
- Simulation Engineering (MOSE) Laboratory
- Department of Engineering and Architectures (DEA)
- University of Trieste
- Trieste
- Italy
| | - Paola Posocco
- Simulation Engineering (MOSE) Laboratory
- Department of Engineering and Architectures (DEA)
- University of Trieste
- Trieste
- Italy
| | - Sabrina Pricl
- Simulation Engineering (MOSE) Laboratory
- Department of Engineering and Architectures (DEA)
- University of Trieste
- Trieste
- Italy
| | | |
Collapse
|
30
|
Stein C, Castanotto D, Krishnan A, Nikolaenko L. Defibrotide (Defitelio): A New Addition to the Stockpile of Food and Drug Administration-approved Oligonucleotide Drugs. MOLECULAR THERAPY. NUCLEIC ACIDS 2016; 5:e346. [PMID: 28131276 PMCID: PMC5023395 DOI: 10.1038/mtna.2016.42] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Accepted: 05/16/2016] [Indexed: 02/08/2023]
Affiliation(s)
- Cy Stein
- Department of Medical Oncology and Experimental Therapeutics, City of Hope, Duarte, California, USA; Department of Molecular and Cellular Biology, City of Hope, Duarte, California, USA.
| | - Daniela Castanotto
- Department of Medical Oncology and Experimental Therapeutics, City of Hope, Duarte, California, USA; Department of Molecular and Cellular Biology, City of Hope, Duarte, California, USA
| | - Amrita Krishnan
- Department of Hematologic Oncology, City of Hope, Duarte, California, USA
| | - Liana Nikolaenko
- Department of Hematologic Oncology, City of Hope, Duarte, California, USA
| |
Collapse
|
31
|
Bromfield SM, Smith DK. Heparin versus DNA: Chiral Preferences in Polyanion Binding to Self-Assembled Multivalent (SAMul) Nanostructures. J Am Chem Soc 2015; 137:10056-9. [DOI: 10.1021/jacs.5b04344] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
| | - David K. Smith
- Department
of Chemistry, University of York, Heslington, York YO10 5DD, U.K
| |
Collapse
|
32
|
Abstract
The immediate early gene product Arc (activity-regulated cytoskeleton-associated protein) is posited as a master regulator of long-term synaptic plasticity and memory. However, the physicochemical and structural properties of Arc have not been elucidated. In the present study, we expressed and purified recombinant human Arc (hArc) and performed the first biochemical and biophysical analysis of hArc's structure and stability. Limited proteolysis assays and MS analysis indicate that hArc has two major domains on either side of a central more disordered linker region, consistent with in silico structure predictions. hArc's secondary structure was estimated using CD, and stability was analysed by CD-monitored thermal denaturation and differential scanning fluorimetry (DSF). Oligomerization states under different conditions were studied by dynamic light scattering (DLS) and visualized by AFM and EM. Biophysical analyses show that hArc is a modular protein with defined secondary structure and loose tertiary structure. hArc appears to be pyramid-shaped as a monomer and is capable of reversible self-association, forming large soluble oligomers. The N-terminal domain of hArc is highly basic, which may promote interaction with cytoskeletal structures or other polyanionic surfaces, whereas the C-terminal domain is acidic and stabilized by ionic conditions that promote oligomerization. Upon binding of presenilin-1 (PS1) peptide, hArc undergoes a large structural change. A non-synonymous genetic variant of hArc (V231G) showed properties similar to the wild-type (WT) protein. We conclude that hArc is a flexible multi-domain protein that exists in monomeric and oligomeric forms, compatible with a diverse, hub-like role in plasticity-related processes. A series of complementary biochemical and biophysical analyses revealed that the human activity-regulated cytoskeleton-associated protein (hArc) protein has a modular structure with two domains, is monomeric but also forms oligomers, which posits Arc to be a flexible hub protein.
Collapse
|
33
|
Heinrich UR, Schmidtmann I, Strieth S, Helling K. Cell-specific accumulation patterns of gentamicin in the guinea pig cochlea. Hear Res 2015; 326:40-8. [PMID: 25882166 DOI: 10.1016/j.heares.2015.03.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 03/19/2015] [Accepted: 03/20/2015] [Indexed: 11/24/2022]
Abstract
Intratympanic gentamicin therapy has become a popular treatment modality for Ménière's disease (MD) through controlled elimination of vertigo spells caused by the balance organ. However, the known ototoxic properties of aminoglycosides lead to cochlear damage. In order to gain more information about cellular preferences for aminoglycoside accumulation within the cochlea, gentamicin was immuno histochemically localized by light microscopy in male guinea pigs 1 and 7 days after intratympanic application (n = 8 ears/incubation time). Differences in the gentamicin-specific cellular storage capacities were quantified by determination of the local immuno staining intensities. Gentamicin was detected in every cochlear cell type, but with spatiotemporal variability. One day after application, an intense staining reaction was found in all cell types except the spiral ganglion cells and the stria vascularis. Six days later, gentamicin staining intensities were additionally reduced in the nerve fibers and the spiral ligament. Statistic analysis revealed strong cellular associations in respect to aminoglycoside accumulation. Furthermore, associations with recorded hearing losses were identified comparing the cellular gentamicin content in the organ of Corti, in the stria vascularis, in the spiral ganglion cells and in fibrocytes of the Limbus. In the lateral wall, clear differences in cellular gentamicin accumulation were found between type I fibrocytes of the spiral ligament compared with basal and intermediate cells of the stria vascularis. This finding was unexpected as these three cell types belong to a well-developed gap-junction system which normally enables unhampered cell communication. Cellular differences in local gentamicin storage capacities, transport processes and inherent diffusion barriers are discussed.
Collapse
Affiliation(s)
- Ulf-Rüdiger Heinrich
- Department of Otorhinolaryngology, Head and Neck Surgery, University Medical Center of the Johannes Gutenberg University Mainz, Germany
| | - Irene Schmidtmann
- Institute for Medical Biostatistics, Epidemiology and Informatics, University Medical Center of the Johannes Gutenberg University Mainz, Germany
| | - Sebastian Strieth
- Department of Otorhinolaryngology, Head and Neck Surgery, University Medical Center of the Johannes Gutenberg University Mainz, Germany
| | - Kai Helling
- Department of Otorhinolaryngology, Head and Neck Surgery, University Medical Center of the Johannes Gutenberg University Mainz, Germany.
| |
Collapse
|
34
|
Shah RB, Schwendeman SP. A biomimetic approach to active self-microencapsulation of proteins in PLGA. J Control Release 2014; 196:60-70. [PMID: 25219750 PMCID: PMC4268178 DOI: 10.1016/j.jconrel.2014.08.029] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 08/19/2014] [Accepted: 08/30/2014] [Indexed: 01/10/2023]
Abstract
A biomimetic approach to organic solvent-free microencapsulation of proteins based on the self-healing capacity of poly (DL)-lactic-co-glycolic acid (PLGA) microspheres containing glycosaminoglycan-like biopolymers (BPs), was examined. To screen BPs, aqueous solutions of BP [high molecular weight dextran sulfate (HDS), low molecular weight dextran sulfate (LDS), chondroitin sulfate (CS), heparin (HP), hyaluronic acid (HA), chitosan (CH)] and model protein lysozyme (LYZ) were combined in different molar and mass ratios, at 37 °C and pH7. The BP-PLGA microspheres (20-63 μm) were prepared by a double water-oil-water emulsion method with a range of BP content, and trehalose and MgCO3 to control microclimate pH and to create percolating pores for protein. Biomimetic active self-encapsulation (ASE) of proteins [LYZ, vascular endothelial growth factor165 (VEGF) and fibroblast growth factor (FgF-20)] was accomplished by incubating blank BP-PLGA microspheres in low concentration protein solutions at ~24 °C, for 48 h. Pore closure was induced at 42.5 °C under mild agitation for 42h. Formulation parameters of BP-PLGA microspheres and loading conditions were studied to optimize protein loading and subsequent release. LDS and HP were found to bind >95% LYZ at BP:LYZ>0.125 w/w, whereas HDS and CS bound >80% LYZ at BP:LYZ of 0.25-1 and <0.33, respectively. HA-PLGA microspheres were found to be not ideal for obtaining high protein loading (>2% w/w of LYZ). Sulfated BP-PLGA microspheres were capable of loading LYZ (~2-7% w/w), VEGF (~4% w/w), and FgF-20 (~2% w/w) with high efficiency. Protein loading was found to be dependent on the loading solution concentration, with higher protein loading obtained at higher loading solution concentration within the range investigated. Loading also increased with content of sulfated BP in microspheres. Release kinetics of proteins was evaluated in-vitro with complete release media replacement. Rate and extent of release were found to depend upon volume of release (with non-sink conditions observed <5 ml release volume for ~18 mg loaded BP-PLGA microspheres), ionic strength of release media and loading solution concentration. HDS-PLGA formulations were identified as having ideal loading and release characteristics. These optimal microspheres released ~73-80% of the encapsulated LYZ over 60 days, with >90% of protein being enzymatically active. Nearly 72% of immunoreactive VEGF was similarly released over 42 days, without significant losses in heparin binding affinity in the release medium.
Collapse
Affiliation(s)
- Ronak B Shah
- Department of Pharmaceutical Sciences, The Biointerfaces Institute, University of Michigan, 2800 Plymouth Rd., Ann Arbor, MI 48109, USA
| | - Steven P Schwendeman
- Department of Pharmaceutical Sciences, The Biointerfaces Institute, University of Michigan, 2800 Plymouth Rd., Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, 2800 Plymouth Rd., Ann Arbor, MI 48109, USA.
| |
Collapse
|
35
|
Besret S, Vicogne J, Dahmani F, Fafeur V, Desmet R, Drobecq H, Romieu A, Melnyk P, Melnyk O. Thiocarbamate-linked polysulfonate-peptide conjugates as selective hepatocyte growth factor receptor binders. Bioconjug Chem 2014; 25:1000-10. [PMID: 24749766 PMCID: PMC4064695 DOI: 10.1021/bc500137j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The capacity of many proteins to interact with natural or synthetic polyanions has been exploited for modulating their biological action. However, the polydispersity of these macromolecular polyanions as well as their poor specificity is a severe limitation to their use as drugs. An emerging trend in this field is the synthesis of homogeneous and well-defined polyanion-peptide conjugates, which act as bivalent ligands, with the peptide part bringing the selectivity of the scaffold. Alternately, this strategy can be used for improving the binding of short peptides to polyanion-binding protein targets. This work describes the design and first synthesis of homogeneous polysulfonate-peptide conjugates using thiocarbamate ligation for binding to the extracellular domain of MET tyrosine kinase receptor for hepatocyte growth factor.
Collapse
|
36
|
Minsky BB, Zheng B, Dubin PL. Inhibition of antithrombin and bovine serum albumin native state aggregation by heparin. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2014; 30:278-287. [PMID: 24313340 DOI: 10.1021/la4039232] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Protein native state aggregation, a major problem in pharmaceutical and biological processes, has been addressed pharmacologically by the addition of protein-binding excipients. Heparin (Hp), a highly sulfated polysaccharide, interacts with numerous proteins with moderate to high affinity, but reports about its effect on protein aggregation are contradictory. We studied the pH dependence of the aggregation of antithrombin (AT) and bovine serum albumin (BSA) in the presence and absence of heparin. High-precision turbidimetry showed strong aggregation for both AT and BSA in I = 10 mM NaCl, conditions at which electrostatically driven Hp binding and aggregation both occur, with more obvious aggregation of heparin-free AT appearing as larger aggregate size. Aggregation of AT was dramatically inhibited at Hp: protein 6:1 (mole ratio); however, the effect at 0.5:1 Hp:protein was greater for BSA. Frontal analysis capillary electrophoresis showed a much larger equilibrium association constant Kobs between Hp and AT, in accord with the onset of Hp binding at a higher pH; both effects are explained by the higher charge density of the positive domain for AT as revealed by modeling with DelPhi. The corresponding modeling images showed that these domains persist at high salt only for AT, consistent with the 160-fold drop in Kobs at 100 mM salt for BSA-Hp binding. The smaller inhibition effect for AT arises from the tendency of its uncomplexed monomer to form larger aggregates more rapidly, but the stronger binding of Hp to AT does not facilitate Hp-induced aggregate dissolution which occurs more readily for BSA. This can be attributed to the higher density of AT aggregates evidenced by higher fractal dimensions. Differences between inhibition and reversal by Hp arise because the former may depend on the stage at which Hp enters the aggregation process and the latter on aggregate size and morphology.
Collapse
Affiliation(s)
- Burcu Baykal Minsky
- Department of Chemistry, University of Massachusetts , Amherst, Massachusetts, 01003, United States
| | | | | |
Collapse
|
37
|
Minsky BB, Nguyen TV, Peyton SR, Kaltashov IA, Dubin PL. Heparin decamer bridges a growth factor and an oligolysine by different charge-driven interactions. Biomacromolecules 2013; 14:4091-8. [PMID: 24107074 DOI: 10.1021/bm401227p] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Full-length heparin is widely used in tissue engineering applications due its multiple protein-binding sites that allow it to retain growth factor affinity while associating with oligopeptide components of the tissue scaffold. However, the extent to which oligopeptide coupling interferes with cognate protein binding is difficult to predict. To investigate such simultaneous interactions, we examined a well-defined ternary system comprised of acidic fibroblast growth factor (FGF), tetralysine (K4), with a heparin decamer (dp10) acting as a noncovalent coupler. Electrospray ionization mass spectrometry was used to assess binding affinities and complex stoichiometries as a function of ionic strength for dp10·K4 and FGF·dp10. The ionic strength dependence of K4·dp10 formation is qualitatively consistent with binding driven by the release of condensed counterions previously suggested for native heparin with divalent oligopeptides (Mascotti, D. P.; Lohman, T. M. Biochemistry 1995, 34, 2908-2915). On the other hand, FGF binding displays more complex ionic strength dependence, with higher salt resistance. Remarkably, dp10 that can bind two FGF molecules can only bind one tetralysine. The limited binding of K4 to dp10 suggests that the tetralysine might not block growth factor binding, and the 1:1:1 ternary complex is indeed observed. The analysis of mass distribution of the bound dp10 chains in FGF·dp10, FGF2·dp10, and FGF·dp10·K4 complexes indicated that higher degrees of dp10 sulfation promote the formation of FGF2·dp10 and FGF·dp10·K4. Thus, the selectivity of appropriately chosen short heparin chains could be used to modulate growth factor sequestration and release in a way not feasible with heterogeneous native heparin. In support of this, human hepatocellular carcinoma cells (HEP3Bs) treated with FGF·dp10·K4 were found to exhibit biological activity similar to cells treated with FGF.
Collapse
Affiliation(s)
- Burcu Baykal Minsky
- Departments of †Chemistry and ‡Chemical Engineering, University of Massachusetts , 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | | | | | | | | |
Collapse
|
38
|
Biological Water Dynamics and Entropy: A Biophysical Origin of Cancer and Other Diseases. ENTROPY 2013. [DOI: 10.3390/e15093822] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
39
|
McCullough RW. High-potency sucralfate prevents and rapidly reverses chemo-radiation mucositis in a patient with stage 4b head and neck cancer. World J Transl Med 2013; 2:13-21. [DOI: 10.5528/wjtm.v2.i2.13] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Accepted: 04/27/2013] [Indexed: 02/05/2023] Open
Abstract
AIM: To study usefulness of high-potency sucralfate (HPS) in a patient with chemoradiation mucositis and discuss its mechanism of action.
METHODS: HPS, a non-covalently cross-link of sucralfate, cations and bidentate anionic chelators, has a maintains a surface concentration of sucralfate 3 h following administration that is 7-23 fold that possible with standard-potency sucralfate. The accelerated mucosal healing and pain alleviation of HPS in patients with erosive esophageal reflux, prompted its use in this patient with chemoradiation mucositis of the oropharynx and alimentary tract. A literature-based review of the immuno-modulatory effects of sucralfate is discussed.
RESULTS: Within 48 h of intervention: (1) there was complete disappearance of oral mucositis lesions; tenderness with (2) patient-reported disappearance of pain, nausea and diarrhea; patient required (3) no opiate analgesia and (4) no tube-feeding supplements to regular diet. Dysgeusia and xerostomia persisted. A modified Naranjo Questionnaire score of 10 supported the likelihood that HPS intervention caused the observed clinical effects. No adverse reactions noted.
CONCLUSION: In this patient HPS was useful to treat chemo-radiation mucositis of the oropharynx and alimentary tract. HPS may directly or indirectly facilitate an immunomodulatory mechanism involving accelerated growth factor activation, which may be a new target for therapeutic intervention in such patients.
Collapse
|
40
|
Hadigal S, Shukla D. Exploiting herpes simplex virus entry for novel therapeutics. Viruses 2013; 5:1447-65. [PMID: 23752649 PMCID: PMC3717716 DOI: 10.3390/v5061447] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Revised: 05/25/2013] [Accepted: 05/31/2013] [Indexed: 12/23/2022] Open
Abstract
Herpes Simplex virus (HSV) is associated with a variety of diseases such as genital herpes and numerous ocular diseases. At the global level, high prevalence of individuals who are seropositive for HSV, combined with its inconspicuous infection, remains a cause for major concern. At the molecular level, HSV entry into a host cell involves multiple steps, primarily the interaction of viral glycoproteins with various cell surface receptors, many of which have alternate substitutes. The molecular complexity of the virus to enter a cell is also enhanced by the existence of different modes of viral entry. The availability of many entry receptors, along with a variety of entry mechanisms, has resulted in a virus that is capable of infecting virtually all cell types. While HSV uses a wide repertoire of viral and host factors in establishing infection, current therapeutics aimed against the virus are not as diversified. In this particular review, we will focus on the initial entry of the virus into the cell, while highlighting potential novel therapeutics that can control this process. Virus entry is a decisive step and effective therapeutics can translate to less virus replication, reduced cell death, and detrimental symptoms.
Collapse
Affiliation(s)
- Satvik Hadigal
- Department of Ophthalmology and Visual Sciences, College of Medicine, University of Illinois at Chicago, 1855 West Taylor Street, m/c 648, Room 3.138, Chicago, IL 60612, USA; E-Mail:
| | - Deepak Shukla
- Department of Ophthalmology and Visual Sciences, College of Medicine, University of Illinois at Chicago, 1855 West Taylor Street, m/c 648, Room 3.138, Chicago, IL 60612, USA; E-Mail:
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, 835 S. Wolcott, Chicago, IL 60612, USA
- Lions of Illinois Eye Research Institute, University of Illinois at Chicago, 1905 West Taylor Street, Chicago, IL 606012, USA
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-312-355-0908; Fax: +1-312-996-7772
| |
Collapse
|
41
|
Uversky VN. The alphabet of intrinsic disorder: II. Various roles of glutamic acid in ordered and intrinsically disordered proteins. INTRINSICALLY DISORDERED PROTEINS 2013; 1:e24684. [PMID: 28516010 PMCID: PMC5424795 DOI: 10.4161/idp.24684] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Revised: 03/27/2013] [Accepted: 04/12/2013] [Indexed: 11/19/2022]
Abstract
The ability of a protein to fold into unique functional state or to stay intrinsically disordered is encoded in its amino acid sequence. Both ordered and intrinsically disordered proteins (IDPs) are natural polypeptides that use the same arsenal of 20 proteinogenic amino acid residues as their major building blocks. The exceptional structural plasticity of IDPs, their capability to exist as heterogeneous structural ensembles and their wide array of important disorder-based biological functions that complements functional repertoire of ordered proteins are all rooted within the peculiar differential usage of these building blocks by ordered proteins and IDPs. In fact, some residues (so-called disorder-promoting residues) are noticeably more common in IDPs than in sequences of ordered proteins, which, in their turn, are enriched in several order-promoting residues. Furthermore, residues can be arranged according to their “disorder promoting potencies,” which are evaluated based on the relative abundances of various amino acids in ordered and disordered proteins. This review continues a series of publications on the roles of different amino acids in defining the phenomenon of protein intrinsic disorder and concerns glutamic acid, which is the second most disorder-promoting residue.
Collapse
Affiliation(s)
- Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute; College of Medicine; University of South Florida; Tampa, FL USA.,Institute for Biological Instrumentation; Russian Academy of Sciences; Moscow, Russia
| |
Collapse
|
42
|
Minsky BB, Atmuri A, Kaltashov IA, Dubin PL. Counterion Condensation on Heparin Oligomers. Biomacromolecules 2013; 14:1113-21. [DOI: 10.1021/bm400006g] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Burcu Baykal Minsky
- Department
of Chemistry, University of Massachusetts, 710 North Pleasant Street,
Amherst, Massachusetts, 01003, United States
| | - Anand Atmuri
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts,
01003, United States
| | - Igor A. Kaltashov
- Department
of Chemistry, University of Massachusetts, 710 North Pleasant Street,
Amherst, Massachusetts, 01003, United States
| | - Paul L. Dubin
- Department
of Chemistry, University of Massachusetts, 710 North Pleasant Street,
Amherst, Massachusetts, 01003, United States
| |
Collapse
|
43
|
Esue O, Xie AX, Kamerzell TJ, Patapoff TW. Thermodynamic and structural characterization of an antibody gel. MAbs 2013; 5:323-34. [PMID: 23425660 DOI: 10.4161/mabs.23183] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Although extensively studied, protein-protein interactions remain highly elusive and are of increasing interest in drug development. We show the assembly of a monoclonal antibody, using multivalent carboxylate ions, into highly-ordered structures. While the presence and function of similar structures in vivo are not known, the results may present a possible unexplored area of antibody structure-function relationships. Using a variety of tools (e.g., mechanical rheology, electron microscopy, isothermal calorimetry, Fourier transform infrared spectroscopy), we characterized the physical, biochemical, and thermodynamic properties of these structures and found that citrate may interact directly with the amino acid residue histidine, after which the individual protein units assemble into a filamentous network gel exhibiting high elasticity and interfilament interactions. Citrate interacts exothermically with the monoclonal antibody with an association constant that is highly dependent on solution pH and temperature. Secondary structure analysis also reveals involvement of hydrophobic and aromatic residues.
Collapse
Affiliation(s)
- Osigwe Esue
- Pharmaceutical Development, Genentech, South San Francisco, CA, USA.
| | | | | | | |
Collapse
|
44
|
Xu Y, Engel Y, Yan Y, Chen K, Moyano DF, Dubin PL, Rotello VM. Enhanced Electrostatic Discrimination of Proteins on Nanoparticle-Coated Surfaces. J Mater Chem B 2013; 1. [PMID: 24273645 DOI: 10.1039/c3tb20377h] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Two β-lactoglobulin (BLG) isoforms, BLGA and BLGB, were used a test bed for the differentiation of proteins using electrostatics. In these studies, the BLGA and BLGB binding to a highly charged, cationic gold nanoparticle (GNP) modified surface was investigated by atomic force microscopy (AFM) and surface plasmon resonance (SPR) spectroscopy The binding affinity, and more importantly, the selectivity of this surface towards these two almost identical protein isoforms were both significantly increased on the cationic GNP surface array relative to the values measured with the same free cationic GNP in solution. While protein recognition is traditionally achieved almost exclusively via orientation dependent short-range interactions such as hydrogen bonds and hydrophobic interactions, our results show the potential of protein recognition platforms based on enhanced electrostatic interactions.
Collapse
Affiliation(s)
- Yisheng Xu
- Department of Chemistry, University of Massachusetts Amherst, MA 01003, USA ; State Key Laboratory of Chemical Engineering, East China University of Science and Technology, Shanghai 200237, P.R. China
| | | | | | | | | | | | | |
Collapse
|
45
|
Kayitmazer AB, Seeman D, Minsky BB, Dubin PL, Xu Y. Protein–polyelectrolyte interactions. SOFT MATTER 2013; 9:2553. [DOI: 10.1039/c2sm27002a] [Citation(s) in RCA: 306] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
46
|
Bechara C, Pallerla M, Zaltsman Y, Burlina F, Alves ID, Lequin O, Sagan S. Tryptophan within basic peptide sequences triggers glycosaminoglycan‐dependent endocytosis. FASEB J 2012; 27:738-49. [DOI: 10.1096/fj.12-216176] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Chérine Bechara
- Université Pierre et Marie Curie (UPMC), Université Paris 6Unité Mixte de Recherche (UMR) 7203, Laboratoire des BioMolécules (LBM)ParisFrance
- Centre National de la Recherche Scientifique (CNRS)UMR 7203, Laboratoire des BioMolécules (LBM)ParisFrance
- Ecole Normale Supérieure (ENS)UMR 7203, Laboratoire des BioMolécules (LBM)ParisFrance
| | - Manjula Pallerla
- Université Pierre et Marie Curie (UPMC), Université Paris 6Unité Mixte de Recherche (UMR) 7203, Laboratoire des BioMolécules (LBM)ParisFrance
- Centre National de la Recherche Scientifique (CNRS)UMR 7203, Laboratoire des BioMolécules (LBM)ParisFrance
- Ecole Normale Supérieure (ENS)UMR 7203, Laboratoire des BioMolécules (LBM)ParisFrance
| | - Yefim Zaltsman
- Université Pierre et Marie Curie (UPMC), Université Paris 6Unité Mixte de Recherche (UMR) 7203, Laboratoire des BioMolécules (LBM)ParisFrance
- Centre National de la Recherche Scientifique (CNRS)UMR 7203, Laboratoire des BioMolécules (LBM)ParisFrance
- Ecole Normale Supérieure (ENS)UMR 7203, Laboratoire des BioMolécules (LBM)ParisFrance
| | - Fabienne Burlina
- Université Pierre et Marie Curie (UPMC), Université Paris 6Unité Mixte de Recherche (UMR) 7203, Laboratoire des BioMolécules (LBM)ParisFrance
- Centre National de la Recherche Scientifique (CNRS)UMR 7203, Laboratoire des BioMolécules (LBM)ParisFrance
- Ecole Normale Supérieure (ENS)UMR 7203, Laboratoire des BioMolécules (LBM)ParisFrance
| | - Isabel D. Alves
- Chimie et Biologie Des Membranes et Des Nanoobjets (CBMN)UMR 5248, CNRSPessacFrance
| | - Olivier Lequin
- Université Pierre et Marie Curie (UPMC), Université Paris 6Unité Mixte de Recherche (UMR) 7203, Laboratoire des BioMolécules (LBM)ParisFrance
- Centre National de la Recherche Scientifique (CNRS)UMR 7203, Laboratoire des BioMolécules (LBM)ParisFrance
- Ecole Normale Supérieure (ENS)UMR 7203, Laboratoire des BioMolécules (LBM)ParisFrance
| | - Sandrine Sagan
- Université Pierre et Marie Curie (UPMC), Université Paris 6Unité Mixte de Recherche (UMR) 7203, Laboratoire des BioMolécules (LBM)ParisFrance
- Centre National de la Recherche Scientifique (CNRS)UMR 7203, Laboratoire des BioMolécules (LBM)ParisFrance
- Ecole Normale Supérieure (ENS)UMR 7203, Laboratoire des BioMolécules (LBM)ParisFrance
| |
Collapse
|
47
|
Santner AA, Croy CH, Vasanwala FH, Uversky VN, Van YYJ, Dunker AK. Sweeping away protein aggregation with entropic bristles: intrinsically disordered protein fusions enhance soluble expression. Biochemistry 2012; 51:7250-62. [PMID: 22924672 DOI: 10.1021/bi300653m] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Intrinsically disordered, highly charged protein sequences act as entropic bristles (EBs), which, when translationally fused to partner proteins, serve as effective solubilizers by creating both a large favorable surface area for water interactions and large excluded volumes around the partner. By extending away from the partner and sweeping out large molecules, EBs can allow the target protein to fold free from interference. Using both naturally occurring and artificial polypeptides, we demonstrate the successful implementation of intrinsically disordered fusions as protein solubilizers. The artificial fusions discussed herein have a low level of sequence complexity and a high net charge but are diversified by means of distinctive amino acid compositions and lengths. Using 6xHis fusions as controls, soluble protein expression enhancements from 65% (EB60A) to 100% (EB250) were observed for a 20-protein portfolio. Additionally, these EBs were able to more effectively solubilize targets compared to frequently used fusions such as maltose-binding protein, glutathione S-transferase, thioredoxin, and N utilization substance A. Finally, although these EBs possess very distinct physiochemical properties, they did not perturb the structure, conformational stability, or function of the green fluorescent protein or the glutathione S-transferase protein. This work thus illustrates the successful de novo design of intrinsically disordered fusions and presents a promising technology and complementary resource for researchers attempting to solubilize recalcitrant proteins.
Collapse
Affiliation(s)
- Aaron A Santner
- Molecular Kinetics Inc., Indianapolis, Indiana 46268, United States
| | | | | | | | | | | |
Collapse
|
48
|
Dannies PS. Prolactin and growth hormone aggregates in secretory granules: the need to understand the structure of the aggregate. Endocr Rev 2012; 33:254-70. [PMID: 22357343 DOI: 10.1210/er.2011-1002] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Prolactin and GH form reversible aggregates in the trans-Golgi lumen that become the dense cores of secretory granules. Aggregation is an economical means of sorting, because self-association removes the hormones from other possible pathways. Secretory granules containing different aggregates show different behavior, such as the reduction in stimulated release of granules containing R183H-GH compared with release of those containing wild-type hormone. Aggregates may facilitate localization of membrane proteins necessary for transport and exocytosis of secretory granules, and therefore understanding their properties is important. Three types of self-association have been characterized: dimers of human GH that form with Zn(2+), low-affinity self-association of human prolactin caused by acidic pH and Zn(2+) with macromolecular crowding, and amyloid fibers of prolactin. The best candidate for the form in most granules may be low-affinity self-association because it occurs rapidly at Zn(2+) concentrations that are likely to be in granules and reverses rapidly in neutral pH. Amyloid may form in older granules. Determining differences between aggregates of wild type and those of R183H-GH should help to understand why granules containing the mutant behave differently from those containing wild-type hormone. If reversible aggregation of other hormones, including those that are proteolytically processed, is the crucial act in forming granules, rather than use of a sorting signal, then prohormones should form reversible aggregates in solution in conditions that resemble those of the trans-Golgi lumen, including macromolecular crowding.
Collapse
Affiliation(s)
- Priscilla S Dannies
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut 06520-8066, USA.
| |
Collapse
|
49
|
Noinaj N, Song ES, Bhasin S, Alper BJ, Schmidt WK, Hersh LB, Rodgers DW. Anion activation site of insulin-degrading enzyme. J Biol Chem 2011; 287:48-57. [PMID: 22049080 DOI: 10.1074/jbc.m111.264614] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Insulin-degrading enzyme (IDE) (insulysin) is a zinc metallopeptidase that metabolizes several bioactive peptides, including insulin and the amyloid β peptide. IDE is an unusual metallopeptidase in that it is allosterically activated by both small peptides and anions, such as ATP. Here, we report that the ATP-binding site is located on a portion of the substrate binding chamber wall arising largely from domain 4 of the four-domain IDE. Two variants having residues in this site mutated, IDEK898A,K899A,S901A and IDER429S, both show greatly decreased activation by the polyphosphate anions ATP and PPPi. IDEK898A,K899A,S901A is also deficient in activation by small peptides, suggesting a possible mechanistic link between the two types of allosteric activation. Sodium chloride at high concentrations can also activate IDE. There are no observable differences in average conformation between the IDE-ATP complex and unliganded IDE, but regions of the active site and C-terminal domain do show increased crystallographic thermal factors in the complex, suggesting an effect on dynamics. Activation by ATP is shown to be independent of the ATP hydrolysis activity reported for the enzyme. We also report that IDEK898A,K899A,S901A has reduced intracellular function relative to unmodified IDE, consistent with a possible role for anion activation of IDE activity in vivo. Together, the data suggest a model in which the binding of anions activates by reducing the electrostatic attraction between the two halves of the enzyme, shifting the partitioning between open and closed conformations of IDE toward the open form.
Collapse
Affiliation(s)
- Nicholas Noinaj
- Department of Molecular and Cellular Biochemistry and the Center for Structural Biology, University of Kentucky, Lexington, Kentucky 40536
| | - Eun Suk Song
- Department of Molecular and Cellular Biochemistry and the Center for Structural Biology, University of Kentucky, Lexington, Kentucky 40536
| | - Sonia Bhasin
- Department of Molecular and Cellular Biochemistry and the Center for Structural Biology, University of Kentucky, Lexington, Kentucky 40536
| | - Benjamin J Alper
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia 30602
| | - Walter K Schmidt
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia 30602
| | - Louis B Hersh
- Department of Molecular and Cellular Biochemistry and the Center for Structural Biology, University of Kentucky, Lexington, Kentucky 40536.
| | - David W Rodgers
- Department of Molecular and Cellular Biochemistry and the Center for Structural Biology, University of Kentucky, Lexington, Kentucky 40536.
| |
Collapse
|
50
|
Dunn IS. RNA templating of molecular assembly and covalent modification patterning in early molecular evolution and modern biosystems. J Theor Biol 2011; 284:32-41. [PMID: 21703277 DOI: 10.1016/j.jtbi.2011.06.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2011] [Revised: 05/23/2011] [Accepted: 06/08/2011] [Indexed: 10/18/2022]
Abstract
The Direct RNA Template (DRT) hypothesis proposes that an early stage of genetic code evolution involved RNA molecules acting as stereochemical recognition templates for assembly of specific amino acids in sequence-ordered arrays, providing a framework for directed covalent peptide bond formation. It is hypothesized here that modern biological precedents may exist for RNA-based structural templating with functional analogies to hypothetical DRT systems. Beyond covalent molecular assembly, an extension of the DRT concept can include RNA molecules acting as dynamic structural template guides for the specific non-covalent assembly of multi-subunit complexes, equivalent to structural assembly chaperones. However, despite numerous precedents for RNA molecules acting as scaffolds for protein complexes, true RNA-mediated assembly chaperoning appears to be absent in modern biosystems. Another level of function with parallels to a DRT system is possible if RNA structural motifs dynamically guided specific patterns of catalytic modifications within multiple target sites in a pre-formed polymer or macromolecular complex. It is suggested that this type of structural RNA templating could logically play a functional role in certain areas of biology, one of which is the glycome of complex organisms. If any such RNA templating processes are shown to exist, they would share no necessary evolutionary relationships with events during early molecular evolution, but may promote understanding of the practical limits of biological RNA functions now and in the ancient RNA World. Awareness of these formal possibilities may also assist in the current search for functions of extensive non-coding RNAs in complex organisms, or for efforts towards artificial rendering of DRT systems.
Collapse
Affiliation(s)
- Ian S Dunn
- CytoCure LLC, 100 Cummings Center, Beverly, MA 01915, USA.
| |
Collapse
|