1
|
Leimu L, Holm P, Gąciarz A, Haavisto O, Prince S, Pesonen U, Huovinen T, Lamminmäki U. Epitope-specific antibody fragments block aggregation of AGelD187N, an aberrant peptide in gelsolin amyloidosis. J Biol Chem 2024; 300:107507. [PMID: 38944121 PMCID: PMC11298591 DOI: 10.1016/j.jbc.2024.107507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 06/10/2024] [Accepted: 06/21/2024] [Indexed: 07/01/2024] Open
Abstract
Aggregation of aberrant fragment of plasma gelsolin, AGelD187N, is a crucial event underlying the pathophysiology of Finnish gelsolin amyloidosis, an inherited form of systemic amyloidosis. The amyloidogenic gelsolin fragment AGelD187N does not play any physiological role in the body, unlike most aggregating proteins related to other protein misfolding diseases. However, no therapeutic agents that specifically and effectively target and neutralize AGelD187N exist. We used phage display technology to identify novel single-chain variable fragments that bind to different epitopes in the monomeric AGelD187N that were further maturated by variable domain shuffling and converted to antigen-binding fragment (Fab) antibodies. The generated antibody fragments had nanomolar binding affinity for full-length AGelD187N, as evaluated by biolayer interferometry. Importantly, all four Fabs selected for functional studies efficiently inhibited the amyloid formation of full-length AGelD187N as examined by thioflavin fluorescence assay and transmission electron microscopy. Two Fabs, neither of which bound to the previously proposed fibril-forming region of AGelD187N, completely blocked the amyloid formation of AGelD187N. Moreover, no small soluble aggregates, which are considered pathogenic species in protein misfolding diseases, were formed after successful inhibition of amyloid formation by the most promising aggregation inhibitor, as investigated by size-exclusion chromatography combined with multiangle light scattering. We conclude that all regions of the full-length AGelD187N are important in modulating its assembly into fibrils and that the discovered epitope-specific anti-AGelD187N antibody fragments provide a promising starting point for a disease-modifying therapy for gelsolin amyloidosis, which is currently lacking.
Collapse
Affiliation(s)
- Laura Leimu
- R&D, Orion Pharma, Orion Corporation, Turku, Finland; Faculty of Medicine, Institute of Biomedicine, University of Turku, Turku, Finland.
| | - Patrik Holm
- R&D, Orion Pharma, Orion Corporation, Turku, Finland; Department of Life Technologies, University of Turku, Turku, Finland; Organon R&D Finland, Turku, Finland
| | - Anna Gąciarz
- R&D, Orion Pharma, Orion Corporation, Turku, Finland; Mobidiag, A Hologic Company, Espoo, Finland
| | - Oskar Haavisto
- Department of Life Technologies, University of Turku, Turku, Finland
| | - Stuart Prince
- R&D, Orion Pharma, Orion Corporation, Turku, Finland; MediCity Research Laboratory, University of Turku, Turku, Finland
| | - Ullamari Pesonen
- Faculty of Medicine, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Tuomas Huovinen
- Department of Life Technologies, University of Turku, Turku, Finland
| | - Urpo Lamminmäki
- Department of Life Technologies, University of Turku, Turku, Finland.
| |
Collapse
|
2
|
Kataria A, Srivastava A, Singh DD, Haque S, Han I, Yadav DK. Systematic computational strategies for identifying protein targets and lead discovery. RSC Med Chem 2024; 15:2254-2269. [PMID: 39026640 PMCID: PMC11253860 DOI: 10.1039/d4md00223g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 05/10/2024] [Indexed: 07/20/2024] Open
Abstract
Computational algorithms and tools have retrenched the drug discovery and development timeline. The applicability of computational approaches has gained immense relevance owing to the dramatic surge in the structural information of biomacromolecules and their heteromolecular complexes. Computational methods are now extensively used in identifying new protein targets, druggability assessment, pharmacophore mapping, molecular docking, the virtual screening of lead molecules, bioactivity prediction, molecular dynamics of protein-ligand complexes, affinity prediction, and for designing better ligands. Herein, we provide an overview of salient components of recently reported computational drug-discovery workflows that includes algorithms, tools, and databases for protein target identification and optimized ligand selection.
Collapse
Affiliation(s)
- Arti Kataria
- Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH) Hamilton MT 59840 USA
| | - Ankit Srivastava
- Laboratory of Neurological Infections and Immunity, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH) Hamilton MT 59840 USA
| | - Desh Deepak Singh
- Amity Institute of Biotechnology, Amity University Rajasthan Jaipur India
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Health Sciences, Jazan University Jazan-45142 Saudi Arabia
| | - Ihn Han
- Plasma Bioscience Research Center, Applied Plasma Medicine Center, Department of Electrical & Biological Physics, Kwangwoon University Seoul 01897 Republic of Korea +82 32 820 4948
| | - Dharmendra Kumar Yadav
- Department of Biologics, College of Pharmacy, Gachon University Hambakmoeiro 191, Yeonsu-gu Incheon 21924 Republic of Korea
| |
Collapse
|
3
|
Sharma S, Tomar VR, Deep S. Mechanism of the interaction of toxic SOD1 fibrils with two potent polyphenols: curcumin and quercetin. Phys Chem Chem Phys 2023; 25:23081-23091. [PMID: 37602388 DOI: 10.1039/d3cp02120c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/22/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a debilitating neurodegenerative disease commonly caused due to the aggregation of superoxide dismutase 1 (SOD1) protein. Finding inhibitors of SOD1 aggregation is of prime concern, but understanding the mechanistic action of inhibitors is equally important. Recent experiments found that two polyphenols, curcumin, and quercetin, have the ability to inhibit SOD1 aggregation. Quercetin was experimentally proven to break pre-formed fibrils into shorter segments, while curcumin did not significantly affect the pre-formed species. Here, we delve deeper into understanding the mechanism of action of quercetin and curcumin on pre-formed octameric fibrils of SOD1 (28PVKVWGSIKGL38: chains A-H) with the help of molecular dynamics (MD) simulations of a fibril docked polyphenol complex. Our results suggest that quercetin shows π-π stacking interaction with one of the key residues for toxic amyloid formation, Trp 32 of chains D, E, and F, and breaks the peptide chains G, and H from the rest of the fibril. On the other hand, curcumin binds to the hydrophobic amino acids of almost all the chains B-H and stabilizes the fibril rather than destabilizing it. Binding free energy calculations using MM/PBSA showed that curcumin binds more strongly to the SOD1 fibril due to greater van der Waals interactions compared to quercetin. These findings provide insights for the development of potential ALS treatments.
Collapse
Affiliation(s)
- Shilpa Sharma
- Department of Chemistry, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India.
| | - Vijay Raj Tomar
- Department of Chemistry, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India.
| | - Shashank Deep
- Department of Chemistry, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India.
| |
Collapse
|
4
|
Romero-Márquez JM, Forbes-Hernández TY, Navarro-Hortal MD, Quirantes-Piné R, Grosso G, Giampieri F, Lipari V, Sánchez-González C, Battino M, Quiles JL. Molecular Mechanisms of the Protective Effects of Olive Leaf Polyphenols against Alzheimer's Disease. Int J Mol Sci 2023; 24:ijms24054353. [PMID: 36901783 PMCID: PMC10001635 DOI: 10.3390/ijms24054353] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/14/2023] [Accepted: 02/16/2023] [Indexed: 02/25/2023] Open
Abstract
Alzheimer's Disease (AD) is the cause of around 60-70% of global cases of dementia and approximately 50 million people have been reported to suffer this disease worldwide. The leaves of olive trees (Olea europaea) are the most abundant by-products of the olive grove industry. These by-products have been highlighted due to the wide variety of bioactive compounds such as oleuropein (OLE) and hydroxytyrosol (HT) with demonstrated medicinal properties to fight AD. In particular, the olive leaf (OL), OLE, and HT reduced not only amyloid-β formation but also neurofibrillary tangles formation through amyloid protein precursor processing modulation. Although the isolated olive phytochemicals exerted lower cholinesterase inhibitory activity, OL demonstrated high inhibitory activity in the cholinergic tests evaluated. The mechanisms underlying these protective effects may be associated with decreased neuroinflammation and oxidative stress via NF-κB and Nrf2 modulation, respectively. Despite the limited research, evidence indicates that OL consumption promotes autophagy and restores loss of proteostasis, which was reflected in lower toxic protein aggregation in AD models. Therefore, olive phytochemicals may be a promising tool as an adjuvant in the treatment of AD.
Collapse
Affiliation(s)
- Jose M. Romero-Márquez
- Department of Physiology, Institute of Nutrition and Food Technology “José Mataix Verdú”, Biomedical Research Centre, University of Granada, 18016 Armilla, Spain
| | - Tamara Y. Forbes-Hernández
- Department of Physiology, Institute of Nutrition and Food Technology “José Mataix Verdú”, Biomedical Research Centre, University of Granada, 18016 Armilla, Spain
| | - María D. Navarro-Hortal
- Department of Physiology, Institute of Nutrition and Food Technology “José Mataix Verdú”, Biomedical Research Centre, University of Granada, 18016 Armilla, Spain
| | - Rosa Quirantes-Piné
- Research and Development Functional Food Centre (CIDAF), Health Science Technological Park, Avenida del Conocimiento 37, 18016 Granada, Spain
| | - Giuseppe Grosso
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
- Center for Human Nutrition and Mediterranean Foods (NUTREA), University of Catania, 95123 Catania, Italy
| | - Francesca Giampieri
- Research Group on Foods, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Isabel Torres 21, 39011 Santander, Spain
| | - Vivian Lipari
- Research Group on Foods, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Isabel Torres 21, 39011 Santander, Spain
- Department of Project Management, Universidad Internacional Iberoamericana, Campeche 24560, Mexico
- Department of Prohect Management, Universidade Internacional do Cuanza, Cuito 250, Bié, Angola
| | - Cristina Sánchez-González
- Department of Physiology, Institute of Nutrition and Food Technology “José Mataix Verdú”, Biomedical Research Centre, University of Granada, 18016 Armilla, Spain
- Sport and Health Research Centre, University of Granada, C/Menéndez Pelayo 32, 18016 Granada, Spain
| | - Maurizio Battino
- Department of Clinical Sciences, Polytechnic University of Marche, 60131 Ancona, Italy
- International Joint Research Laboratory of Intelligent Agriculture and Agri-Products Processing, Jiangsu University, Zhenjiang 212013, China
| | - José L. Quiles
- Department of Physiology, Institute of Nutrition and Food Technology “José Mataix Verdú”, Biomedical Research Centre, University of Granada, 18016 Armilla, Spain
- Research and Development Functional Food Centre (CIDAF), Health Science Technological Park, Avenida del Conocimiento 37, 18016 Granada, Spain
- Research Group on Foods, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Isabel Torres 21, 39011 Santander, Spain
- Correspondence:
| |
Collapse
|
5
|
Sanfilippo C, Castrogiovanni P, Imbesi R, Musumeci G, Vecchio M, Li Volti G, Tibullo D, Broggi G, Caltabiano R, Ulivieri M, Kazakova M, Parenti R, Vicario N, Fazio F, Di Rosa M. Sex-dependent neuro-deconvolution analysis of Alzheimer's disease brain transcriptomes according to CHI3L1 expression levels. J Neuroimmunol 2022; 373:577977. [PMID: 36228382 DOI: 10.1016/j.jneuroim.2022.577977] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 08/29/2022] [Accepted: 10/01/2022] [Indexed: 11/07/2022]
Abstract
Glial activation and related neuroinflammatory processes play a key role in the aging and progression of Alzheimer's disease (AD). CHI3L1/ YKL40 is a widely investigated chitinase in neurodegenerative diseases and recent studies have shown its involvement in aging and AD. Nevertheless, the biological function of CHI3L1 in AD is still unknown. Here, we collected microarray datasets from the National Center for Biotechnology Information (NCBI) brain samples of not demented healthy controls (NDHC) who died from causes not attributable to neurodegenerative disorders (n = 460), and of deceased patients suffering from Alzheimer's disease (AD) (n = 697). The NDHC and AD patients were stratified according to CHI3L1 expression levels as a cut-off. We identified two groups both males and females, subsequently used for our statistical comparisons: the high CHI3L1 expression group (HCEG) and the low CHI3L1 expression group (LCEG). Comparing HCEG to LCEG, we attained four signatures according to the sex of patients, in order to identify the healthy and AD brain cellular architecture, performing a genomic deconvolution analysis. We used neurological signatures (NS) belonging to six neurological cells populations and nine signatures that included the main physiological neurological processes. We discovered that, in the brains of NDHC the high expression levels of CHI3L1 were associated with astrocyte activation profile, while in AD males and females we showed an inflammatory profile microglia-mediated. The low CHI3L1 brain expression levels in NDHC and AD patients highlighted a neuronal activation profile. Furthermore, using drugs opposing CHI3L1 transcriptomic signatures, we found a specific drug profile for AD males and females characterized by high levels of CHI3L1 composed of fostamatinib, rucaparib, cephaeline, prednisolone, and dinoprostone. Brain levels of CHI3L1 in AD patients represent a biological signature that allows distinguishing between males and females and their likely cellular brain architecture.
Collapse
Affiliation(s)
- Cristina Sanfilippo
- Neurologic Unit, AOU "Policlinico-San Marco", Department of Medical, Surgical Sciences and Advanced Technologies, GF, Ingrassia, University of Catania, Via Santa Sofia n.78, 95100 Catania, Sicily, Italy
| | - Paola Castrogiovanni
- Department of Biomedical and Biotechnological Sciences, Anatomy, Histology and Movement Sciences Section, School of Medicine, University of Catania, 95125 Catania, Italy
| | - Rosa Imbesi
- Department of Biomedical and Biotechnological Sciences, Anatomy, Histology and Movement Sciences Section, School of Medicine, University of Catania, 95125 Catania, Italy
| | - Giuseppe Musumeci
- Department of Biomedical and Biotechnological Sciences, Anatomy, Histology and Movement Sciences Section, School of Medicine, University of Catania, 95125 Catania, Italy
| | - Michele Vecchio
- Rehabilitation Unit, "AOU Policlinico Vittorio Emanuele", Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania 95123, Italy
| | - Giovanni Li Volti
- Department of Biomedical and Biotechnological Sciences, Section of Biochemistry, University of Catania, 95123, Catania, Italy
| | - Daniele Tibullo
- Department of Biomedical and Biotechnological Sciences, Section of Biochemistry, University of Catania, 95123, Catania, Italy
| | - Giuseppe Broggi
- Department of Medical and Surgical Sciences and Advanced Technologies "G. F. Ingrassia", Anatomic Pathology, University of Catania, 95123, Catania, Italy
| | - Rosario Caltabiano
- Department of Medical and Surgical Sciences and Advanced Technologies "G. F. Ingrassia", Anatomic Pathology, University of Catania, 95123, Catania, Italy
| | - Martina Ulivieri
- University of California San Diego, Department of Psychiatry, Health Science, San Diego, La Jolla, CA, USA
| | - Maria Kazakova
- Department of Medical Biology, Medical University, Plovdiv, 4002 Plovdiv, Bulgaria; Research Institute, Medical University-, Plovdiv, 4002 Plovdiv, Bulgaria
| | - Rosalba Parenti
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Nunzio Vicario
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Francesco Fazio
- University of California San Diego, Department of Psychiatry, Health Science, San Diego, La Jolla, CA, USA
| | - Michelino Di Rosa
- Department of Biomedical and Biotechnological Sciences, Anatomy, Histology and Movement Sciences Section, School of Medicine, University of Catania, 95125 Catania, Italy.
| |
Collapse
|
6
|
Shahbaz SK, Koushki K, Sathyapalan T, Majeed M, Sahebkar A. PLGA-Based Curcumin Delivery System: An Interesting Therapeutic Approach in the Treatment of Alzheimer's Disease. Curr Neuropharmacol 2022; 20:309-323. [PMID: 34429054 PMCID: PMC9413791 DOI: 10.2174/1570159x19666210823103020] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 06/23/2021] [Accepted: 07/24/2021] [Indexed: 11/22/2022] Open
Abstract
Progressive degeneration and dysfunction of the nervous system because of oxidative stress, aggregations of misfolded proteins, and neuroinflammation are the key pathological features of neurodegenerative diseases. Alzheimer's disease is a chronic neurodegenerative disorder driven by uncontrolled extracellular deposition of β-amyloid (Aβ) in the amyloid plaques and intracellular accumulation of hyperphosphorylated tau protein. Curcumin is a hydrophobic polyphenol with noticeable neuroprotective and anti-inflammatory effects that can cross the blood-brain barrier. Therefore, it is widely studied for the alleviation of inflammatory and neurological disorders. However, the clinical application of curcumin is limited due to its low aqueous solubility and bioavailability. Recently, nano-based curcumin delivery systems are developed to overcome these limitations effectively. This review article discusses the effects and potential mechanisms of curcumin-loaded PLGA nanoparticles in Alzheimer's disease.
Collapse
Affiliation(s)
- Sanaz Keshavarz Shahbaz
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Disease, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Khadijeh Koushki
- Hepatitis Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Thozhukat Sathyapalan
- Department of Academic Diabetes, Endocrinology and Metabolism, Hull York Medical School, University of Hull, Hull HU3 2JZ, UK
| | | | - Amirhossein Sahebkar
- BARUiotechnol Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Medicine, The University of Western Australia, Perth, Australia
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
7
|
Aggregation of gelsolin wild-type and G167K/R, N184K, and D187N/Y mutant peptides and inhibition. Mol Cell Biochem 2021; 476:2393-2408. [PMID: 33598831 DOI: 10.1007/s11010-021-04085-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 01/25/2021] [Indexed: 10/22/2022]
Abstract
Gelsolin, an actin-binding protein, is localized intra- and extracellularly in the bloodstream and throughout the body. Gelsolin amyloidosis is a disease characterized by several point mutations that lead to cleavage and fibrillization of gelsolin. The D187 mutation to N or Y leads to aggregation of peptide fragments with shortest aggregating peptide identified as 182SFNNGDCFILD192. Recently, G167 has also been identified as relevant gelsolin mutation, which leads to gelsolin deposits in kidneys, but its aggregation is much less understood. Hence, we systematically investigated in vitro the aggregation propensities of the following gelsolin peptides: 167GRRVV171 (1), 161RLFQVKG167 (2), 184NNGDCFILDL193 (3), 188CFILDL193 (4), 187DCFILDL193 (5), and their respective mutants (G167K, G167R, N184K, D187Y, D187N), by using spectroscopic methods [fluorescence Proteostat, Thioflavin T (ThT), turbidity assay, and Dynamic Light Scattering (DLS)], and Transmission Electron Microscopy (TEM). The (non) mutant peptides containing CFILDL sequence aggregated into fibrillar networks, while G167R mutation promoted aggregation compared to the wild-type sequence. In the presence of inhibitors, Methylene Blue (MB) and epigallocatechin gallate (EGCG), the gelsolin peptide (3-5) aggregation was reduced with the IC50 values in the 2-13 µM range. We discovered that inhibitors have dual functionality, as aggregation inhibitors and disaggregation promoters, potentially allowing for the prevention and reversal of gelsolin amyloidosis. Such therapeutic strategies may improve outcomes related to other amyloidogenic diseases of the heart, brain, and eye.
Collapse
|
8
|
Admane N, Srivastava A, Jamal S, Kundu B, Grover A. Protective Effects of a Neurohypophyseal Hormone Analogue on Prion Aggregation, Cellular Internalization, and Toxicity. ACS Chem Neurosci 2020; 11:2422-2430. [PMID: 31407881 DOI: 10.1021/acschemneuro.9b00299] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Herein, we report novel neuroprotective activity of the neurohypophyseal hormone analogue desmopressin (DDAVP) against toxic conformations of human prion protein. Systematic analysis using biophysical techniques in conjunction with surface plasmon resonance, high-end microscopy, conformational antibodies, and cell-based assays demonstrated DDAVP's specific binding and potent antiaggregating effects on prion protein (rPrPres). In addition to subjugating conformational conversion of rPrPres into oligomeric forms, DDAVP also exhibits potent fibril modulatory effects. It eventually ameliorated neuronal toxicity of rPrPres oligomers by significantly reducing their cellular internalization. Molecular dynamics simulations showed that DDAVP prevents β-sheet transitions in the N-terminal amyloidogenic region of prion and induces antagonistic mobilities in its α2-α3 and β2-α2 loop regions. Collectively, our data proposes DDAVP as a new structural motif for rational drug discovery against prion diseases.
Collapse
Affiliation(s)
- Nikita Admane
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India − 110067
| | - Ankit Srivastava
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, India − 110016
| | - Salma Jamal
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India − 110067
| | - Bishwajit Kundu
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, India − 110016
| | - Abhinav Grover
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India − 110067
| |
Collapse
|
9
|
Mohammadi G, Fathian-Kolahkaj M, Mohammadi P, Adibkia K, Fattahi A. Preparation, Physicochemical Characterization and Anti-Fungal Evaluation of Amphotericin B-Loaded PLGA-PEG-Galactosamine Nanoparticles. Adv Pharm Bull 2020; 11:311-317. [PMID: 33880353 PMCID: PMC8046404 DOI: 10.34172/apb.2021.044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 07/11/2020] [Accepted: 07/15/2020] [Indexed: 12/19/2022] Open
Abstract
Purpose: The present study aimed to formulate PLGA and PLGA-PEG-galactosamine nanoparticles (NPs) loaded with amphotericin B with appropriate physicochemical properties and antifungal activity. PLGA was functionalized with GalN to increase the adhesion and antifungal activity of NPs against Candida albicans. Methods: The physicochemical properties of NPs were characterized by particle size determination, zeta potential, drug crystallinity, loading efficiency, dissolution studies, differential scanning calorimeter (DSC), X-ray powder diffraction (XRPD), and Fourier transform infrared (FT-IR). Antifungal activity of the NPs at different drug/polymer ratios was examined by determining minimum inhibitory concentrations (MICs). Results: the FT-IR and 1 HNMR analysis successfully confirmed the formation of PLGA- PEG-GalN NPs. The PLGA NPs were in the size range of 174.1 ± 3.49 to 238.2±7.59 nm while PLGA-GalN NPs were 255.6 ±4.08 nm in size , respectively. Loading efficiency was in the range of 67%±2.4 to 77%±1.6, and entrapment efficiency in the range of 68.185%±1.9 to 73.05%±0.6. Zeta potential and loading efficiency for PLGA-GalN NPs were –0.456, 71%. The NPs indicated an amorphous status according to XRPD patterns and DSC thermograms. The PLGA-PEG-GalN NPs showed higher fungistatic activity than PLGA NPs. Conclusion: the results demonstrated that the antifungal activity of PLGA-PEG-GalN NPs was higher than pure amphotericin B and PLGA NPs.
Collapse
Affiliation(s)
- Ghobad Mohammadi
- Pharmaceutical Sciences Research Center Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | | | - Pardis Mohammadi
- Pharmaceutical Sciences Research Center Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Khosro Adibkia
- Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Fattahi
- Pharmaceutical Sciences Research Center Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
10
|
Wilson AC, Chou SF, Lozano R, Chen JY, Neuenschwander PF. Thermal and Physico-Mechanical Characterizations of Thromboresistant Polyurethane Films. Bioengineering (Basel) 2019; 6:bioengineering6030069. [PMID: 31416139 PMCID: PMC6783839 DOI: 10.3390/bioengineering6030069] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 07/19/2019] [Accepted: 08/07/2019] [Indexed: 01/27/2023] Open
Abstract
Hemocompatibility remains a challenge for injectable and/or implantable medical devices, and thromboresistant coatings appear to be one of the most attractive methods to down-regulate the unwanted enzymatic reactions that promote the formation of blood clots. Among all polymeric materials, polyurethanes (PUs) are a class of biomaterials with excellent biocompatibility and bioinertness that are suitable for the use of thromboresistant coatings. In this work, we investigated the thermal and physico-mechanical behaviors of ester-based and ether-based PU films for potential uses in thromboresistant coatings. Our results show that poly(ester urethane) and poly(ether urethane) films exhibited characteristic peaks corresponding to their molecular configurations. Thermal characterizations suggest a two-step decomposition process for the poly(ether urethane) films. Physico-mechanical characterizations show that the surfaces of the PU films were hydrophobic with minimal weight changes in physiological conditions over 14 days. All PU films exhibited high tensile strength and large elongation to failure, attributed to their semi-crystalline structure. Finally, the in vitro clotting assays confirmed their thromboresistance with approximately 1000-fold increase in contact time with human blood plasma as compared to the glass control. Our work correlates the structure-property relationships of PU films with their excellent thromboresistant ability.
Collapse
Affiliation(s)
- Aaron C Wilson
- Department of Mechanical Engineering, College of Engineering, The University of Texas at Tyler, 3900 University Blvd, Tyler, TX 75799, USA
| | - Shih-Feng Chou
- Department of Mechanical Engineering, College of Engineering, The University of Texas at Tyler, 3900 University Blvd, Tyler, TX 75799, USA.
| | - Roberto Lozano
- School of Human Ecology, College of Natural Sciences, The University of Texas at Austin, Austin, TX 78712, USA
| | - Jonathan Y Chen
- School of Human Ecology, College of Natural Sciences, The University of Texas at Austin, Austin, TX 78712, USA
| | - Pierre F Neuenschwander
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX 75708, USA
| |
Collapse
|
11
|
Specific keratinase derived designer peptides potently inhibit Aβ aggregation resulting in reduced neuronal toxicity and apoptosis. Biochem J 2019; 476:1817-1841. [DOI: 10.1042/bcj20190183] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 05/22/2019] [Accepted: 05/27/2019] [Indexed: 11/17/2022]
Abstract
Abstract
Compelling evidence implicates self-assembly of amyloid-β (Aβ1–42) peptides into soluble oligomers and fibrils as a major underlying event in Alzheimer's disease (AD) pathogenesis. Herein, we employed amyloid-degrading keratinase (kerA) enzyme as a key Aβ1–42-binding scaffold to identify five keratinase-guided peptides (KgPs) capable of interacting with and altering amyloidogenic conversion of Aβ1–42. The KgPs showed micromolar affinities with Aβ1–42 and abolished its sigmoidal amyloidogenic transition, resulting in abrogation of fibrillogenesis. Comprehensive assessment using dynamic light scattering (DLS), atomic force microscopy (AFM) and Fourier-transform infrared (FTIR) spectroscopy showed that KgPs induced the formation of off-pathway oligomers comparatively larger than the native Aβ1–42 oligomers but with a significantly reduced cross-β signature. These off-pathway oligomers exhibited low immunoreactivity against oligomer-specific (A11) and fibril-specific (OC) antibodies and rescued neuronal cells from Aβ1–42 oligomer toxicity as well as neuronal apoptosis. Structural analysis using molecular docking and molecular dynamics (MD) simulations showed two preferred KgP binding sites (Lys16–Phe20 and Leu28–Val39) on the NMR ensembles of monomeric and fibrillar Aβ1–42, indicating an interruption of crucial hydrophobic and aromatic interactions. Overall, our results demonstrate a new approach for designing potential anti-amyloid molecules that could pave way for developing effective therapeutics against AD and other amyloid diseases.
Collapse
|
12
|
Thymoquinone loaded mesoporous silica nanoparticles retard cell invasion and enhance in vitro cytotoxicity due to ROS mediated apoptosis in HeLa and MCF-7 cell lines. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 104:109881. [PMID: 31499940 DOI: 10.1016/j.msec.2019.109881] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 06/10/2019] [Accepted: 06/10/2019] [Indexed: 12/14/2022]
Abstract
Thymoquinone (TQ) loaded monodispersed mesoporous silica nanoparticles (TQ-MSNPs) with size of 188 ± 3 nm were prepared and characterized using DLS, TEM and FTIR. These TQ-MSNPs overcome the limitations of free TQ like hydrophobicity, low aqueous and photo stability and thus enhance its anticancer activity. In vitro release kinetics showed biphasic drug release where up to 50% was released in first 8 h and subsequently 98% released after 48 h. Enhanced cytotoxicity of TQ-MSNPs was observed against MCF-7 and HeLa cell lines as compared to free TQ. DAPI and Annexin V-FITC/PI staining confirmed the induction of apoptosis in cancer cells following treatment with TQ-MSNPs. Also, TQ-MSNPs exhibited enhanced anti-invasion properties against both cell lines as very low concentration of loaded TQ imparts similar benefits as free TQ. Both TQ and TQ-MSNPs exerted their cytotoxicity via reactive oxygen species (ROS) generation, as addition of an antioxidant NAC attenuated their killing activity.
Collapse
|
13
|
Bonsai Gelsolin Survives Heat Induced Denaturation by Forming β-Amyloids which Leach Out Functional Monomer. Sci Rep 2018; 8:12602. [PMID: 30135452 PMCID: PMC6105678 DOI: 10.1038/s41598-018-30951-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 07/31/2018] [Indexed: 12/13/2022] Open
Abstract
Here, we report that minimal functional gelsolin i.e. fragment 28–161 can display F-actin depolymerizing property even after heating the protein to 80 °C. Small angle X-ray scattering (SAXS) data analysis confirmed that under Ca2+-free conditions, 28–161 associates into monomer to dimer and tetramer, which later forms β-amyloids, but in presence of Ca2+, it forms dimers which proceed to non-characterizable aggregates. The dimeric association also explained the observed decrease in ellipticity in circular dichroism experiments with increase in temperature. Importantly, SAXS data based models correlated well with our crystal structure of dimeric state of 28–161. Characterization of higher order association by electron microscopy, Congo red and ThioflavinT staining assays further confirmed that only in absence of Ca2+ ions, heating transforms 28–161 into β-amyloids. Gel filtration and other experiments showed that β-amyloids keep leaching out the monomer, and the release rates could be enhanced by addition of L-Arg to the amyloids. F-actin depolymerization showed that addition of Ca2+ ions to released monomer initiated the depolymerization activity. Overall, we propose a way to compose a supramolecular assembly which releases functional protein in sustained manner which can be applied for varied potentially therapeutic interventions.
Collapse
|
14
|
Co-delivery of curcumin and serratiopeptidase in HeLa and MCF-7 cells through nanoparticles show improved anti-cancer activity. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 92:673-684. [PMID: 30184794 DOI: 10.1016/j.msec.2018.07.025] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 06/27/2018] [Accepted: 07/09/2018] [Indexed: 11/21/2022]
Abstract
Curcumin was employed to prepare anticancer nanoparticles (size 175 ± 15 nm) using anti-inflammatory enzyme serratiopeptidase by desolvation method. Here serratiopeptidase acted as a carrier as well as bioactive molecule in the nanoformulations. The Cur-SPD NPs (curcumin loaded serratiopeptidase nanoparticles) were characterized using DLS, FESEM and FTIR. The in vitro release behavior depicted biphasic pattern at 37 °C (pH 7.4) and release of 95% of both molecules occurred in 24 h. Serratiopeptidase not only provided stability to curcumin but also increased its effectiveness against cancer cells. These nanoparticles had anti-cancer activity in MCF-7 and HeLa cell lines as shown by cytotoxicity assay, DAPI nuclear staining, ROS production and DNA damage. The immunomodulatory tests showed that Cur-SPD NPs reduce level of IL-6 but increase TNFα level in THP1 cell lines. Structural similarity of serratiopeptidase to matrix metallo proteases (MMPs), particularly MMP8, have been found (based on low RMSD values) to induce TNFα production and play tumour suppressive role in certain cancers. Thus anti-cancer properties of Cur-SPD NPs may be attributed to synergistic effect of curcumin and serratiopeptidase. Thus results in present investigation provide an insight on role of serratiopeptidase in development of co-delivery of multifunctional nanoparticles with anti-cancer properties introduction.
Collapse
|
15
|
Pradhan N, Debnath K, Mandal S, Jana NR, Jana NR. Antiamyloidogenic Chemical/Biochemical-Based Designed Nanoparticle as Artificial Chaperone for Efficient Inhibition of Protein Aggregation. Biomacromolecules 2018; 19:1721-1731. [DOI: 10.1021/acs.biomac.8b00671] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Nibedita Pradhan
- Centre for Advanced Materials, Indian Association for the Cultivation of Science, Kolkata 700032, India
| | - Koushik Debnath
- Centre for Advanced Materials, Indian Association for the Cultivation of Science, Kolkata 700032, India
| | - Suman Mandal
- Centre for Advanced Materials, Indian Association for the Cultivation of Science, Kolkata 700032, India
| | - Nihar R. Jana
- Cellular and Molecular Neuroscience Laboratory, National Brain Research Centre, Manesar, Gurgaon 122051, India
| | - Nikhil R. Jana
- Centre for Advanced Materials, Indian Association for the Cultivation of Science, Kolkata 700032, India
| |
Collapse
|
16
|
Srivastava A, Singh J, Singh Yadav SP, Arya P, Kalim F, Rose P, Ashish, Kundu B. The Gelsolin Pathogenic D187N Mutant Exhibits Altered Conformational Stability and Forms Amyloidogenic Oligomers. Biochemistry 2018; 57:2359-2372. [DOI: 10.1021/acs.biochem.8b00039] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Ankit Srivastava
- Kusuma School of Biological Sciences, IIT Delhi, New Delhi 110016, India
| | - Jasdeep Singh
- Kusuma School of Biological Sciences, IIT Delhi, New Delhi 110016, India
| | | | - Prabha Arya
- Kusuma School of Biological Sciences, IIT Delhi, New Delhi 110016, India
| | - Fouzia Kalim
- Kusuma School of Biological Sciences, IIT Delhi, New Delhi 110016, India
| | - Pooja Rose
- Kusuma School of Biological Sciences, IIT Delhi, New Delhi 110016, India
| | - Ashish
- CSIR-Institute of Microbial Technology, Chandigarh 160036, India
| | - Bishwajit Kundu
- Kusuma School of Biological Sciences, IIT Delhi, New Delhi 110016, India
| |
Collapse
|
17
|
Pillarisetti S, Maya S, Sathianarayanan S, Jayakumar R. Tunable pH and redox-responsive drug release from curcumin conjugated γ-polyglutamic acid nanoparticles in cancer microenvironment. Colloids Surf B Biointerfaces 2017; 159:809-819. [DOI: 10.1016/j.colsurfb.2017.08.057] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 07/27/2017] [Accepted: 08/28/2017] [Indexed: 01/27/2023]
|
18
|
Verhelle A, Van Overbeke W, Peleman C, De Smet R, Zwaenepoel O, Lahoutte T, Van Dorpe J, Devoogdt N, Gettemans J. Non-Invasive Imaging of Amyloid Deposits in a Mouse Model of AGel Using 99mTc-Modified Nanobodies and SPECT/CT. Mol Imaging Biol 2017; 18:887-897. [PMID: 27130233 DOI: 10.1007/s11307-016-0960-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
PURPOSE Gelsolin amyloidosis (AGel), also known as familial amyloidosis, Finnish type (FAF), is an autosomal, dominant, incurable disease caused by a point mutation (G654A/T) in the gelsolin (GSN) gene. The mutation results in loss of a Ca2+-binding site in the second gelsolin domain. Subsequent incorrect folding exposes a cryptic furin cleavage site, leading to the formation of a 68-kDa C-terminal cleavage product (C68) in the trans-Golgi network. This C68 fragment is cleaved by membrane type 1-matrix metalloproteinase (MT1-MMP) during secretion into the extracellular environment, releasing 8- and 5-kDa amyloidogenic peptides. These peptides aggregate and cause disease-associated symptoms. We set out to investigate whether AGel-specific nanobodies could be used to monitor amyloidogenic gelsolin buildup. PROCEDURES Three nanobodies (FAF Nb1-3) raised against the 8-kDa fragment were screened as AGel amyloid imaging agents in WT and AGel mice using 99mTc-based single-photon emission computed tomography (SPECT)/X-ray tomography (CT), biodistribution analysis, and immunofluorescence (IF). The quantitative characteristics were analyzed in a follow-up study with a Nb11-expressing mouse model. RESULTS All three nanobodies possess the characteristics desired for a 99mTc-based SPECT/CT imaging agent, high specificity and a low background signal. FAF Nb1 was identified as the most potent, based on its superior signal-to-noise ratio and signal specificity. As a proof of concept, we implemented 99mTc-FAF Nb1 in a follow-up study of the Nb11-expressing AGel mouse model. Using biodistribution analysis and immunofluorescence, we demonstrated the validity of the data acquired via 99mTc-FAF Nb1 SPECT/CT. CONCLUSION These findings demonstrate the potential of this nanobody as a non-invasive tool to image amyloidogenic gelsolin deposition and assess the therapeutic capacity of AGel therapeutics currently under development. We propose that this approach can be extended to other amyloid diseases, thereby contributing to the development of specific therapies.
Collapse
Affiliation(s)
- Adriaan Verhelle
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University, Albert Baertsoenkaai 3, B-9000, Ghent, Belgium
| | - Wouter Van Overbeke
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University, Albert Baertsoenkaai 3, B-9000, Ghent, Belgium
| | - Cindy Peleman
- In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, Brussels, Belgium
| | - Rebecca De Smet
- Department of Medical and Forensic Pathology, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Olivier Zwaenepoel
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University, Albert Baertsoenkaai 3, B-9000, Ghent, Belgium
| | - Tony Lahoutte
- In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, Brussels, Belgium
| | - Jo Van Dorpe
- Department of Medical and Forensic Pathology, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Nick Devoogdt
- In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, Brussels, Belgium
| | - Jan Gettemans
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University, Albert Baertsoenkaai 3, B-9000, Ghent, Belgium.
| |
Collapse
|
19
|
Singh J, Srivastava A, Sharma P, Pradhan P, Kundu B. DNA intercalators as amyloid assembly modulators: mechanistic insights. RSC Adv 2017. [DOI: 10.1039/c6ra26313e] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
DNA intercalators modulate amyloid assembly of proteins through specific hetero-aromatic interactions diverting them to form amorphous aggregates.
Collapse
Affiliation(s)
- Jasdeep Singh
- Kusuma School of Biological Sciences
- Indian Institute of Technology Delhi
- New Delhi
- India
| | - Ankit Srivastava
- Kusuma School of Biological Sciences
- Indian Institute of Technology Delhi
- New Delhi
- India
| | - Pankaj Sharma
- Kusuma School of Biological Sciences
- Indian Institute of Technology Delhi
- New Delhi
- India
| | - Prashant Pradhan
- Kusuma School of Biological Sciences
- Indian Institute of Technology Delhi
- New Delhi
- India
| | - Bishwajit Kundu
- Kusuma School of Biological Sciences
- Indian Institute of Technology Delhi
- New Delhi
- India
| |
Collapse
|
20
|
Modulation of prion polymerization and toxicity by rationally designed peptidomimetics. Biochem J 2016; 474:123-147. [DOI: 10.1042/bcj20160737] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Revised: 10/25/2016] [Accepted: 10/31/2016] [Indexed: 11/17/2022]
Abstract
Misfolding and aggregation of cellular prion protein is associated with a large array of neurological disorders commonly called the transmissible spongiform encephalopathies. Designing inhibitors against prions has remained a daunting task owing to limited information about mechanism(s) of their pathogenic self-assembly. Here, we explore the anti-prion properties of a combinatorial library of bispidine-based peptidomimetics (BPMs) that conjugate amino acids with hydrophobic and aromatic side chains. Keeping the bispidine unit unaltered, a series of structurally diverse BPMs were synthesized and tested for their prion-modulating properties. Administration of Leu- and Trp-BPMs delayed and completely inhibited the amyloidogenic conversion of human prion protein (HuPrP), respectively. We found that each BPM induced the HuPrP to form unique oligomeric nanostructures differing in their biophysical properties, cellular toxicities and response to conformation-specific antibodies. While Leu-BPMs were found to stabilize the oligomers, Trp-BPMs effected transient oligomerization, resulting in the formation of non-toxic, non-fibrillar aggregates. Yet another aromatic residue, Phe, however, accelerated the aggregation process in HuPrP. Molecular insights obtained through MD (molecular dynamics) simulations suggested that each BPM differently engages a conserved Tyr 169 residue at the α2–β2 loop of HuPrP and affects the stability of α2 and α3 helices. Our results demonstrate that this new class of molecules having chemical scaffolds conjugating hydrophobic/aromatic residues could effectively modulate prion aggregation and toxicity.
Collapse
|
21
|
Siddiqi MK, Alam P, Chaturvedi SK, Khan RH. Anti-amyloidogenic behavior and interaction of Diallylsulfide with Human Serum Albumin. Int J Biol Macromol 2016; 92:1220-1228. [DOI: 10.1016/j.ijbiomac.2016.08.035] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Revised: 08/10/2016] [Accepted: 08/11/2016] [Indexed: 11/30/2022]
|
22
|
Dutta C, Yang M, Long F, Shahbazian-Yassar R, Tiwari A. Preformed Seeds Modulate Native Insulin Aggregation Kinetics. J Phys Chem B 2015; 119:15089-99. [DOI: 10.1021/acs.jpcb.5b07221] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Colina Dutta
- Department
of Chemistry, Michigan Technological University, Houghton, Michigan 49931, United States
| | - Mu Yang
- Department
of Chemistry, Michigan Technological University, Houghton, Michigan 49931, United States
| | - Fei Long
- Department
of Mechanical Engineering, Michigan Technological University, Houghton, Michigan 49931, United States
| | - Reza Shahbazian-Yassar
- Department
of Mechanical Engineering, Michigan Technological University, Houghton, Michigan 49931, United States
- Department
of Mechanical and Industrial Engineering, University of Illinois at Chicago, Chicago, Illinois 60607, United States
| | - Ashutosh Tiwari
- Department
of Chemistry, Michigan Technological University, Houghton, Michigan 49931, United States
| |
Collapse
|