1
|
Wang L, Li X, Men X, Liu X, Luo J. Research progress on antioxidants and protein aggregation inhibitors in cataract prevention and therapy (Review). Mol Med Rep 2025; 31:22. [PMID: 39513587 PMCID: PMC11574704 DOI: 10.3892/mmr.2024.13387] [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: 06/25/2024] [Accepted: 10/10/2024] [Indexed: 11/15/2024] Open
Abstract
Cataracts are primarily caused by aging or gene mutations and are the leading cause of blindness globally. As the older population increases, the number of patients with a cataract is expected to grow rapidly. At present, cataract surgery to replace the lens with an artificial intraocular lens is the principal treatment method. However, surgery has several drawbacks, including economic burdens and complications such as inflammation, xerophthalmia, macular edema and posterior capsular opacification. Thus, developing an effective non‑surgical treatment strategy is beneficial to both patients and public health. Mechanistically, cataract formation may be due to various reasons but is primarily initiated and promoted by oxidative stress and is closely associated with crystallin aggregation. In the present review, the current research progress on anti‑cataract drugs, including antioxidants and protein aggregation inhibitors is examined. It summarizes strategies for preventing and treating cataract through cell apoptosis and protein aggregation inhibition while discussing their limitations and further prospects.
Collapse
Affiliation(s)
- Ling Wang
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, Hunan 410219, P.R. China
| | - Xin Li
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, Hunan 410219, P.R. China
| | - Xiaoju Men
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, Hunan 410219, P.R. China
| | - Xiangyi Liu
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, Hunan 410219, P.R. China
| | - Jinque Luo
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, Hunan 410219, P.R. China
| |
Collapse
|
2
|
Zabcı S, Kocabıyık S. Anti-aggregation Properties of the Mini-Peptides Derived from Alpha Crystallin Domain of the Small Heat Shock Protein, Tpv HSP 14.3. Mol Biotechnol 2024:10.1007/s12033-024-01332-1. [PMID: 39645640 DOI: 10.1007/s12033-024-01332-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 11/15/2024] [Indexed: 12/09/2024]
Abstract
The highly conserved alpha crystallin domain of the small heat shock proteins is essential for dimerization and also implicated in substrate interaction. In this study, we designed four novel mini-peptides from alpha crystallin domain of archaeal Small Heat Shock Protein Tpv HSP 14.3. Among the peptide designs, the mini-peptides 38SDLVLEAEMAGFDKKNIKVS57 and 40LVLEAEMAGFD50 overlapped to the sequences of β3-β4 region. The other two peptides 77YIDQRVDKVYKVVKLPVE94 and 107GILTVRMK114 correspond to β6-β7 region and β9, respectively. Functional activity of the peptides was evaluated by monitoring heat-induced aggregation of the model substrates alcohol dehydrogenase at 43 °C and citrate synthase at 45 °C. Our results showed that the (38-57) and the (77-94) fragments exhibited chaperone activity with both of the substrate proteins. The (40-50) fragment while exhibiting a noticeable protective effect (> 90%) when tested with citrate synthase showed an anti-chaperone property toward alcohol dehydrogenase. Unlike the (40-50) fragment, the (107-114) fragment did not show any chaperone activity with citrate synthase but exhibited the highest chaperone efficiency among four mini-peptides with alcohol dehydrogenase. The selectivity of the (40-50) and the (107-114) fragments in targeting the client proteins is most likely dependent on their surface hydrophobicity and/or charge as revealed by the sequence and exposed surface analyses.
Collapse
Affiliation(s)
- Sema Zabcı
- Department of Biological Sciences, Faculty of Arts and Science, Middle East Technical University, 06800, Ankara, Türkiye.
- Department of Molecular Biology and Genetics, Faculty of Arts and Science, Baskent University, 06790, Ankara, Türkiye.
| | - Semra Kocabıyık
- Department of Biological Sciences, Faculty of Arts and Science, Middle East Technical University, 06800, Ankara, Türkiye
| |
Collapse
|
3
|
Johnson GA, Kodati B, Nahomi RB, Pham JH, Krishnamoorthy VR, Phillips NR, Krishnamoorthy RR, Nagaraj RH, Stankowska DL. Mechanisms contributing to inhibition of retinal ganglion cell death by cell permeable peptain-1 under glaucomatous stress. Cell Death Discov 2024; 10:305. [PMID: 38942762 PMCID: PMC11213865 DOI: 10.1038/s41420-024-02070-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 06/03/2024] [Accepted: 06/13/2024] [Indexed: 06/30/2024] Open
Abstract
This study assesses the neuroprotective potential of CPP-P1, a conjugate of an anti-apoptotic peptain-1 (P1) and a cell-penetrating peptide (CPP) in in vitro, in vivo, and ex vivo glaucoma models. Primary retinal ganglion cells (RGCs) were subjected to either neurotrophic factor (NF) deprivation for 48 h or endothelin-3 (ET-3) treatment for 24 h and received either CPP-P1 or vehicle. RGC survival was analyzed using a Live/Dead assay. Axotomized human retinal explants were treated with CPP-P1 or vehicle for seven days, stained with RGC marker RBPMS, and RGC survival was analyzed. Brown Norway (BN) rats with elevated intraocular pressure (IOP) received weekly intravitreal injections of CPP-P1 or vehicle for six weeks. RGC function was evaluated using a pattern electroretinogram (PERG). RGC and axonal damage were also assessed. RGCs from ocular hypertensive rats treated with CPP-P1 or vehicle for seven days were isolated for transcriptomic analysis. RGCs subjected to 48 h of NF deprivation were used for qPCR target confirmation. NF deprivation led to a significant loss of RGCs, which was markedly reduced by CPP-P1 treatment. CPP-P1 also decreased ET-3-mediated RGC death. In ex vivo human retinal explants, CPP-P1 decreased RGC loss. IOP elevation resulted in significant RGC loss in mid-peripheral and peripheral retinas compared to that in naive rats, which was significantly reduced by CPP-P1 treatment. PERG amplitude decline in IOP-elevated rats was mitigated by CPP-P1 treatment. Following IOP elevation in BN rats, the transcriptomic analysis showed over 6,000 differentially expressed genes in the CPP-P1 group compared to the vehicle-treated group. Upregulated pathways included CREB signaling and synaptogenesis. A significant increase in Creb1 mRNA and elevated phosphorylated Creb were observed in CPP-P1-treated RGCs. Our study showed that CPP-P1 is neuroprotective through CREB signaling enhancement in several settings that mimic glaucomatous conditions. The findings from this study are significant as they address the pressing need for the development of efficacious therapeutic strategies to maintain RGC viability and functionality associated with glaucoma.
Collapse
Affiliation(s)
- Gretchen A Johnson
- North Texas Eye Research Institute, School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, TX, USA
- Department of Microbiology, Immunology, and Genetics, School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Bindu Kodati
- North Texas Eye Research Institute, School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, TX, USA
- Department of Pharmacology and Neuroscience, School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Rooban B Nahomi
- Department of Ophthalmology, School of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO, USA
| | - Jennifer H Pham
- North Texas Eye Research Institute, School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, TX, USA
- Department of Microbiology, Immunology, and Genetics, School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, TX, USA
| | | | - Nicole R Phillips
- Department of Microbiology, Immunology, and Genetics, School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Raghu R Krishnamoorthy
- North Texas Eye Research Institute, School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, TX, USA
- Department of Pharmacology and Neuroscience, School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Ram H Nagaraj
- Department of Ophthalmology, School of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO, USA
| | - Dorota L Stankowska
- North Texas Eye Research Institute, School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, TX, USA.
- Department of Microbiology, Immunology, and Genetics, School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, TX, USA.
| |
Collapse
|
4
|
Ghosh D, Agarwal M, Radhakrishna M. Molecular Insights into the Inhibitory Role of α-Crystallin against γD-Crystallin Aggregation. J Chem Theory Comput 2024; 20:1740-1752. [PMID: 38078935 DOI: 10.1021/acs.jctc.3c00774] [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: 12/21/2023]
Abstract
Cataracts, a major cause of global blindness, contribute significantly to the overall prevalence of blindness. The opacification of the lens, resulting in cataract formation, primarily occurs due to the aggregation of crystallin proteins within the eye lens. Despite the high concentration of these crystallins, they remarkably maintain the lens transparency and refractive index. α-Crystallins (α-crys), acting as chaperones, play a crucial role in preventing crystallin aggregation, although the exact molecular mechanism remains uncertain. In this study, we employed a combination of molecular docking, all-atom molecular dynamics simulations, and advanced free energy calculations to investigate the interaction between γD-crystallin (γD-crys), a major structural protein of the eye lens, and α-crystallin proteins. Our findings demonstrate that α-crys exhibits an enhanced affinity for the NTD2 and CTD4 regions of γD-crys. The NTD2 and CTD4 regions form the interface between the N-terminal domain (NTD) and the C-terminal domain (CTD) of the γD-crys protein. By binding to the interface region between the NTD and CTD of the protein, α-crys effectively inhibits the formation of domain-swapped aggregates and mitigates protein aggregation. Analysis of the Markov state models using molecular dynamics trajectories confirms that minimum free energy conformations correspond to the binding of the α-crystallin domain (ACD) of α-crys to NTD2 and CTD4 that form the interdomain interface.
Collapse
Affiliation(s)
- Deepshikha Ghosh
- Department of Biological Sciences and Engineering, Indian Institute of Technology (IIT) Gandhinagar, Palaj, Gujarat 382355, India
| | - Manish Agarwal
- Computer Services Centre, Indian Institute of Technology (IIT) Delhi, Hauz Khas, New Delhi, Delhi 110016, India
| | - Mithun Radhakrishna
- Department of Chemical Engineering, Indian Institute of Technology (IIT) Gandhinagar, Palaj, Gujarat 382355, India
- Center for Biomedical Engineering, Indian Institute of Technology (IIT) Gandhinagar, Palaj, Gujarat 382355, India
| |
Collapse
|
5
|
Chernova LS, Vishnyakov IE, Börner J, Bogachev MI, Thormann KM, Kayumov AR. The Functionality of IbpA from Acholeplasma laidlawii Is Governed by Dynamic Rearrangement of Its Globular-Fibrillar Quaternary Structure. Int J Mol Sci 2023; 24:15445. [PMID: 37895124 PMCID: PMC10607609 DOI: 10.3390/ijms242015445] [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: 08/18/2023] [Revised: 10/18/2023] [Accepted: 10/20/2023] [Indexed: 10/29/2023] Open
Abstract
Small heat shock proteins (sHSPs) represent a first line of stress defense in many bacteria. The primary function of these molecular chaperones involves preventing irreversible protein denaturation and aggregation. In Escherichia coli, fibrillar EcIbpA binds unfolded proteins and keeps them in a folding-competent state. Further, its structural homologue EcIbpB induces the transition of EcIbpA to globules, thereby facilitating the substrate transfer to the HSP70-HSP100 system for refolding. The phytopathogenic Acholeplasma laidlawii possesses only a single sHSP, AlIbpA. Here, we demonstrate non-trivial features of the function and regulation of the chaperone-like activity of AlIbpA according to its interaction with other components of the mycoplasma multi-chaperone network. Our results show that the efficiency of the A. laidlawii multi-chaperone system is driven with the ability of AlIbpA to form both globular and fibrillar structures, thus combining functions of both IbpA and IbpB when transferring the substrate proteins to the HSP70-HSP100 system. In contrast to EcIbpA and EcIbpB, AlIbpA appears as an sHSP, in which the competition between the N- and C-terminal domains regulates the shift of the protein quaternary structure between a fibrillar and globular form, thus representing a molecular mechanism of its functional regulation. While the C-terminus of AlIbpA is responsible for fibrils formation and substrate capture, the N-terminus seems to have a similar function to EcIbpB through facilitating further substrate protein disaggregation using HSP70. Moreover, our results indicate that prior to the final disaggregation process, AlIbpA can directly transfer the substrate to HSP100, thereby representing an alternative mechanism in the HSP interaction network.
Collapse
Affiliation(s)
- Liliya S. Chernova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kremlevskaya 18, 420008 Kazan, Russia;
- Institute of Cytology, Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 St. Petersburg, Russia;
- Institute of Microbiology and Molecular Biology, Justus Liebig University, Heinrich-Buff-Ring 26, 35392 Giessen, Germany; (J.B.); (K.M.T.)
| | - Innokentii E. Vishnyakov
- Institute of Cytology, Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 St. Petersburg, Russia;
| | - Janek Börner
- Institute of Microbiology and Molecular Biology, Justus Liebig University, Heinrich-Buff-Ring 26, 35392 Giessen, Germany; (J.B.); (K.M.T.)
| | - Mikhail I. Bogachev
- Centre for Digital Telecommunication Technologies, St. Petersburg Electrotechnical University, Professora Popova 5, 197376 St. Petersburg, Russia;
| | - Kai M. Thormann
- Institute of Microbiology and Molecular Biology, Justus Liebig University, Heinrich-Buff-Ring 26, 35392 Giessen, Germany; (J.B.); (K.M.T.)
| | - Airat R. Kayumov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kremlevskaya 18, 420008 Kazan, Russia;
| |
Collapse
|
6
|
Attia SA, Truong AT, Phan A, Lee SJ, Abanmai M, Markanovic M, Avila H, Luo H, Ali A, Sreekumar PG, Kannan R, MacKay JA. αB-Crystallin Peptide Fused with Elastin-like Polypeptide: Intracellular Activity in Retinal Pigment Epithelial Cells Challenged with Oxidative Stress. Antioxidants (Basel) 2023; 12:1817. [PMID: 37891896 PMCID: PMC10604459 DOI: 10.3390/antiox12101817] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 09/19/2023] [Accepted: 09/22/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND Oxidative stress-induced retinal degeneration is among the main contributing factors of serious ocular pathologies that can lead to irreversible blindness. αB-crystallin (cry) is an abundant component of the visual pathway in the vitreous humor, which modulates protein and cellular homeostasis. Within this protein exists a 20 amino acid fragment (mini-cry) with both chaperone and antiapoptotic activity. This study fuses this mini-cry peptide to two temperature-sensitive elastin-like polypeptides (ELP) with the goal of prolonging its activity in the retina. METHODS The biophysical properties and chaperone activity of cry-ELPs were confirmed by mass spectrometry, cloud-point determination, and dynamic light scattering 'DLS'. For the first time, this work compares a simpler ELP architecture, cry-V96, with a previously reported ELP diblock copolymer, cry-SI. Their relative mechanisms of cellular uptake and antiapoptotic potential were tested using retinal pigment epithelial cells (ARPE-19). Oxidative stress was induced with H2O2 and comparative internalization of both cry-ELPs was made using 2D and 3D culture models. We also explored the role of lysosomal membrane permeabilization by confocal microscopy. RESULTS The results indicated successful ELP fusion, cellular association with both 2D and 3D cultures, which were enhanced by oxidative stress. Both constructs suppressed apoptotic signaling (cleaved caspase-3); however, cry-V96 exhibited greater lysosomal escape. CONCLUSIONS ELP architecture is a critical factor to optimize delivery of therapeutic peptides, such as the anti-apoptotic mini-cry peptide; furthermore, the protection of mini-cry via ELPs is enhanced by lysosomal membrane permeabilization.
Collapse
Affiliation(s)
- Sara Aly Attia
- Department of Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA; (S.A.A.); (A.T.T.); (A.P.); (M.A.); (M.M.); (H.A.); (H.L.); (A.A.)
| | - Anh Tan Truong
- Department of Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA; (S.A.A.); (A.T.T.); (A.P.); (M.A.); (M.M.); (H.A.); (H.L.); (A.A.)
| | - Alvin Phan
- Department of Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA; (S.A.A.); (A.T.T.); (A.P.); (M.A.); (M.M.); (H.A.); (H.L.); (A.A.)
| | - Shin-Jae Lee
- Mann Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA;
| | - Manal Abanmai
- Department of Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA; (S.A.A.); (A.T.T.); (A.P.); (M.A.); (M.M.); (H.A.); (H.L.); (A.A.)
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam bin Abdulaziz University, Al Kharj 11942, Saudi Arabia
| | - Marinella Markanovic
- Department of Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA; (S.A.A.); (A.T.T.); (A.P.); (M.A.); (M.M.); (H.A.); (H.L.); (A.A.)
| | - Hugo Avila
- Department of Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA; (S.A.A.); (A.T.T.); (A.P.); (M.A.); (M.M.); (H.A.); (H.L.); (A.A.)
| | - Haozhong Luo
- Department of Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA; (S.A.A.); (A.T.T.); (A.P.); (M.A.); (M.M.); (H.A.); (H.L.); (A.A.)
| | - Atham Ali
- Department of Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA; (S.A.A.); (A.T.T.); (A.P.); (M.A.); (M.M.); (H.A.); (H.L.); (A.A.)
| | | | - Ram Kannan
- Doheny Eye Institute, Pasadena, CA 91103, USA; (P.G.S.); (R.K.)
- Stein Eye Institute, Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - J. Andrew MacKay
- Department of Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA; (S.A.A.); (A.T.T.); (A.P.); (M.A.); (M.M.); (H.A.); (H.L.); (A.A.)
- Mann Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA;
- Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| |
Collapse
|
7
|
Mini-αA-Crystallin Stifled Melittin-Induced Haemolysis and Lymphocyte Lysis. Int J Pept Res Ther 2023. [DOI: 10.1007/s10989-023-10502-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2023]
Abstract
AbstractMelittin, the most potent pharmacological ingredient of honey bee venom, induces haemolysis, lymphocyte lysis, long-term pain, localised inflammation, and hyperalgesia. In this study, efforts were made to subdue the melittin’s ill effects using a chaperone peptide called ‘mini-αA-crystallin’ (MAC) derived from eye lens αA-crystallin. Haemolytic test on human red blood cells, percentage viability, and DNA diffusion assay on Human peripheral blood lymphocytes (HPBLs) were performed with melittin in the presence or absence of MAC. Propidium iodide and Annexin V-FITC dual staining were performed to analyse quantitative levels of necrotic and apoptotic induction by melittin in the presence or absence of MAC on HPBLs using a flow cytometer. A computational study to find out the interactions between MAC and melittin was undertaken by modelling the structure of MAC using a PEP-FOLD server. The result showed that MAC inhibited melittin-induced lysis in nucleated (lymphocytes) and enucleated (RBC) cells. Flow cytometric analysis revealed a substantial increase in the necrotic and late apoptotic cells after treating HPBLs with melittin (4 µg/ml) for 24 h. Treatment with MAC at a 2:1 molar ratio prevented HPBLs from developing melittin-induced necrosis and late apoptosis. In the docking study, hydrogen, van der Waals, π-π stacking, and salt bridges were observed between the MAC and melittin complex, confirming a strong interaction between them. The MAC-melittin complex was stable during molecular dynamics simulation. These findings may be beneficial in developing a medication for treating severe cases of honeybee stings.
Collapse
|
8
|
Emerging therapeutic roles of small heat shock protein-derived mini-chaperones and their delivery strategies. Biochimie 2022; 208:56-65. [PMID: 36521577 DOI: 10.1016/j.biochi.2022.12.004] [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: 08/26/2022] [Revised: 11/28/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022]
Abstract
The small heat shock protein (sHsp) family is a group of proteins in which some are induced in response to external stimuli, such as environmental and pathological stresses, while others are constitutively expressed. They show chaperone-like activity, protect cells from apoptosis, and maintain cytoskeletal architecture. Short sequences or fragments ranging from approximately 19-20 residues in sHsps were shown to display chaperone activity in vitro. These sequences are termed sHsp-derived mini-peptides/mini-chaperones. These peptides offer an advantage in providing protective and therapeutic effects over full-length proteins owing to their small molecular weight and easy uptake into the cells. Research on sHsp mini-chaperone therapy has recently received attention and advanced tremendously. sHsp mini-chaperones have shown a wide range of therapeutic effects, such as anti-aggregation of proteins, anti-apoptotic, anti-inflammatory, anti-oxidant, senolytic, and anti-platelet activity. The administration of mini-chaperones into the several disease animal models, including experimental autoimmune encephalomyelitis, cataract, age-related macular degeneration, glaucoma, and thrombosis through various routes reduced symptoms or prevented the progression of the disease. However, it was found that the therapeutic potential of sHsp mini-chaperones is limited by their short turnover and enzymatic degradation in circulation. Nonetheless, carrier molecules approach such as nanoparticles, cell penetration peptides, and extracellular vesicles increased their efficacy by enhancing the uptake, retention time, protection from enzymatic degradation, and site-specific delivery without altering their biological activity. In this context, this review highlights the recent advances in the therapeutic potential of sHsp-derived mini-chaperones, their effect in experimental animal models, and approaches for increasing their efficacy.
Collapse
|
9
|
Sharma A, Rastogi H, Sundar Ghosh K. Enhancement in chaperone activity of human αA-crystallin by nanochaperone gold nanoparticles: Multispectroscopic studies on their molecular interactions. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2022; 279:121344. [PMID: 35605420 DOI: 10.1016/j.saa.2022.121344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 04/28/2022] [Accepted: 04/30/2022] [Indexed: 06/15/2023]
Abstract
The chaperone activity of human αA-crystallin (HAA) against aggregation of human γD-crystallin (HGD) was enhanced by gold nanoparticles (AuNPs). Chaperone activity of HAA was almost doubled in the presence of 5.5 nM gold nanoparticles (AuNPs). To decipher this effect at molecular level, interactions between HAA and AuNPs were studied using fluorescence and circular dichroism spectroscopic techniques. The nanoparticles were synthesized and characterized by using TEM and dynamic light scattering (DLS). TEM and DLS studies revealed that bioconjugation of AuNPs with HAA did not cause any significant change in the size of the nanoparticles. AuNPs had caused static quenching of tryptophan (Trp) fluorescence, which was confirmed through determination of excited state lifetime of Trp residue of HAA in absence and the presence of AuNPs. The association and quenching constant for HAA-AuNPs conjugation were ∼ 109 M-1. Hydrogen bonding and van der Waals interactions were found to be involved in HAA-AuNPs complex. Polarity of Trp microenvironment in HAA was not perturbed by AuNPs as supported by synchronous and three-dimensional fluorescence spectroscopy. Far-UV CD spectra suggested that the secondary structure of HAA was not significantly affected by AuNPs.
Collapse
Affiliation(s)
- Anchal Sharma
- Department of Chemistry, National Institute of Technology Hamirpur, Himachal Pradesh 177005, India
| | - Harshita Rastogi
- Department of Chemistry, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| | - Kalyan Sundar Ghosh
- Department of Chemistry, National Institute of Technology Hamirpur, Himachal Pradesh 177005, India.
| |
Collapse
|
10
|
Rajeswaren V, Wong JO, Yabroudi D, Nahomi RB, Rankenberg J, Nam MH, Nagaraj RH. Small Heat Shock Proteins in Retinal Diseases. Front Mol Biosci 2022; 9:860375. [PMID: 35480891 PMCID: PMC9035800 DOI: 10.3389/fmolb.2022.860375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 03/11/2022] [Indexed: 11/29/2022] Open
Abstract
This review summarizes the latest findings on small heat shock proteins (sHsps) in three major retinal diseases: glaucoma, diabetic retinopathy, and age-related macular degeneration. A general description of the structure and major cellular functions of sHsps is provided in the introductory remarks. Their role in specific retinal diseases, highlighting their regulation, role in pathogenesis, and possible use as therapeutics, is discussed.
Collapse
Affiliation(s)
- Vivian Rajeswaren
- Department of Ophthalmology, Sue Anschutz-Rodgers Eye Center, School of Medicine, Aurora, CO, United States
| | - Jeffrey O. Wong
- Department of Ophthalmology, Sue Anschutz-Rodgers Eye Center, School of Medicine, Aurora, CO, United States
| | - Dana Yabroudi
- Department of Ophthalmology, Sue Anschutz-Rodgers Eye Center, School of Medicine, Aurora, CO, United States
| | - Rooban B. Nahomi
- Department of Ophthalmology, Sue Anschutz-Rodgers Eye Center, School of Medicine, Aurora, CO, United States
| | - Johanna Rankenberg
- Department of Ophthalmology, Sue Anschutz-Rodgers Eye Center, School of Medicine, Aurora, CO, United States
| | - Mi-Hyun Nam
- Department of Ophthalmology, Sue Anschutz-Rodgers Eye Center, School of Medicine, Aurora, CO, United States
- *Correspondence: Mi-Hyun Nam, ; Ram H. Nagaraj,
| | - Ram H. Nagaraj
- Department of Ophthalmology, Sue Anschutz-Rodgers Eye Center, School of Medicine, Aurora, CO, United States
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, United States
- *Correspondence: Mi-Hyun Nam, ; Ram H. Nagaraj,
| |
Collapse
|
11
|
Timsina R, Trossi-Torres G, Thieme J, O'Dell M, Khadka NK, Mainali L. Alpha-Crystallin Association with the Model of Human and Animal Eye Lens-Lipid Membranes is Modulated by Surface Hydrophobicity of Membranes. Curr Eye Res 2022; 47:843-853. [PMID: 35179407 DOI: 10.1080/02713683.2022.2040539] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
PURPOSE This research aims to probe the interaction of α-crystallin with a model of human, porcine, and mouse lens-lipid membranes. METHODS Cholesterol/model of human lens-lipid (Chol/MHLL), cholesterol/model of porcine lens-lipid (Chol/MPLL), and cholesterol/model of mouse lens-lipid (Chol/MMLL) membranes with 0 to 60 mol% Chol were prepared using the rapid solvent exchange method and probe-tip sonication. The hydrophobicity near the surface of model lens-lipid membranes and α-crystallin association with these membranes were investigated using the electron paramagnetic resonance spin-labeling approach. RESULTS With increased Chol content, the hydrophobicity near the surface of Chol/MHLL, Chol/MPLL, and Chol/MMLL membranes, the maximum percentage of membrane surface occupied (MMSO) by α-crystallin, and the association constant (Ka) decreased, showing that surface hydrophobicity of model lens-lipid membranes modulated the α-crystallin association with these membranes. The different MMSO and Ka for different model lens-lipid membranes with different rates of decrease of MMSO and Ka with increased Chol content and decreased hydrophobicity near the surface of these membranes suggested that the lipid composition also modulates α-crystallin association with membranes. Despite different lipid compositions, complete inhibition of α-crystallin association with model lens-lipid membranes was observed at saturating Chol content forming cholesterol bilayer domains (CBDs) with the lowest hydrophobicity near the surface of these membranes. The decreased mobility parameter with increased α-crystallin concentration suggested that membranes near the surface became less mobile due to α-crystallin association. The decreased mobility parameter and increased maximum splitting with increased Chol content suggested that membranes became less mobile and more ordered near the surface with increased Chol content. CONCLUSIONS This study suggested that the interaction of α-crystallin with model lens-lipid membranes is hydrophobic. Furthermore, our data indicated that Chol and CBDs reduce α-crystallin association with lens membrane, likely increase α-crystallin concentration in lens cytoplasm, and possibly favor the chaperone-like activity of α-crystallin maintaining lens cytoplasm homeostasis.
Collapse
Affiliation(s)
- Raju Timsina
- Department of Physics, Boise State University, Boise, ID 83725, USA
| | | | - Jackson Thieme
- Department of Physics, Boise State University, Boise, ID 83725, USA
| | - Matthew O'Dell
- Biomolecular Sciences Graduate Program, Boise State University, Boise, ID 83725, USA
| | - Nawal K Khadka
- Department of Physics, Boise State University, Boise, ID 83725, USA
| | - Laxman Mainali
- Department of Physics, Boise State University, Boise, ID 83725, USA.,Biomolecular Sciences Graduate Program, Boise State University, Boise, ID 83725, USA
| |
Collapse
|
12
|
Muranov KO, Ostrovsky MA. Biochemistry of Eye Lens in the Norm and in Cataractogenesis. BIOCHEMISTRY. BIOKHIMIIA 2022; 87:106-120. [PMID: 35508906 DOI: 10.1134/s0006297922020031] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/30/2021] [Accepted: 09/30/2021] [Indexed: 06/14/2023]
Abstract
The absence of cellular organelles in fiber cells and very high cytoplasmic protein concentration (up to 900 mg/ml) minimize light scattering in the lens and ensure its transparency. Low oxygen concentration, powerful defense systems (antioxidants, antioxidant enzymes, chaperone-like protein alpha-crystallin, etc.) maintain lens transparency. On the other hand, the ability of crystallins to accumulate age-associated post-translational modifications, which reduce the resistance of lens proteins to oxidative stress, is an important factor contributing to the cataract formation. Here, we suggest a mechanism of cataractogenesis common for the action of different cataractogenic factors, such as age, radiation, ultraviolet light, diabetes, etc. Exposure to these factors leads to the damage and death of lens epithelium, which allows oxygen to penetrate into the lens through the gaps in the epithelial layer and cause oxidative damage to crystallins, resulting in protein denaturation, aggregation, and formation of multilamellar bodies (the main cause of lens opacification). The review discusses various approaches to the inhibition of lens opacification (cataract development), in particular, a combined use of antioxidants and compounds enhancing the chaperone-like properties of alpha-crystallin. We also discuss the paradox of high efficiency of anti-cataract drugs in laboratory settings with the lack of their clinical effect, which might be due to the late use of the drugs at the stage, when the opacification has already formed. A probable solution to this situation will be development of new diagnostic methods that will allow to predict the emergence of cataract long before the manifestation of its clinical signs and to start early preventive treatment.
Collapse
Affiliation(s)
- Konstantin O Muranov
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Moscow, 119334, Russia.
| | - Mikhail A Ostrovsky
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Moscow, 119334, Russia
| |
Collapse
|
13
|
Montepietra D, Cecconi C, Brancolini G. Combining enhanced sampling and deep learning dimensionality reduction for the study of the heat shock protein B8 and its pathological mutant K141E. RSC Adv 2022; 12:31996-32011. [PMID: 36380940 PMCID: PMC9641792 DOI: 10.1039/d2ra04913a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 10/28/2022] [Indexed: 11/11/2022] Open
Abstract
The biological functions of proteins closely depend on their conformational dynamics. This aspect is especially relevant for intrinsically disordered proteins (IDP) for which structural ensembles often offer more useful representations than individual conformations. Here we employ extensive enhanced sampling temperature replica-exchange atomistic simulations (TREMD) and deep learning dimensionality reduction to study the conformational ensembles of the human heat shock protein B8 and its pathological mutant K141E, for which no experimental 3D structures are available. First, we combined homology modelling with TREMD to generate high-dimensional data sets of 3D structures. Then, we employed a recently developed machine learning based post-processing algorithm, EncoderMap, to project the large conformational data sets into meaningful two-dimensional maps that helped us interpret the data and extract the most significant conformations adopted by both proteins during TREMD. These studies provide the first 3D structural characterization of HSPB8 and reveal the effects of the pathogenic K141E mutation on its conformational ensembles. In particular, this missense mutation appears to increase the compactness of the protein and its structural variability, at the same time rearranging the hydrophobic patches exposed on the protein surface. These results offer the possibility of rationalizing the pathogenic effects of the K141E mutation in terms of conformational changes. The study provides the first 3D structural characterization of HSPB8 and its K141E mutant: extensive TREMD are combined with a deep learning algorithm to rationalize the disordered ensemble of structures adopted by each variant.![]()
Collapse
Affiliation(s)
- Daniele Montepietra
- Department of Physics, Computer Science and Mathematics, University of Modena and Reggio Emilia, Via Campi 213/A, 41100 Modena, Italy
- Istituto Nanoscienze – CNR-NANO, Center S3, Via G. Campi 213/A, 41100 Modena, Italy
| | - Ciro Cecconi
- Department of Physics, Computer Science and Mathematics, University of Modena and Reggio Emilia, Via Campi 213/A, 41100 Modena, Italy
- Istituto Nanoscienze – CNR-NANO, Center S3, Via G. Campi 213/A, 41100 Modena, Italy
| | - Giorgia Brancolini
- Istituto Nanoscienze – CNR-NANO, Center S3, Via G. Campi 213/A, 41100 Modena, Italy
| |
Collapse
|
14
|
Fu X, Ezemaduka AN, Lu X, Chang Z. The Caenorhabditis elegans 12-kDa small heat shock proteins with little in vitro chaperone activity play crucial roles for its dauer formation, longevity, and reproduction. Protein Sci 2021; 30:2170-2182. [PMID: 34272907 DOI: 10.1002/pro.4160] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 07/15/2021] [Accepted: 07/16/2021] [Indexed: 01/31/2023]
Abstract
Small heat shock proteins (sHSPs) are known to exhibit in vitro chaperone activity by suppressing the aggregation of misfolded proteins. The 12-kDa sHSPs (Hsp12s) subfamily members from Caenorhabditis elegans, including Hsp12.2, Hsp12.3, and Hsp12.6, however, are devoid of such chaperone activity, and their in vivo functions are poorly understood. Here we verified that Hsp12.1, similar to its homologs Hsp12.2, Hsp12.3, and Hsp12.6, hardly exhibited any chaperone activity. Strikingly, we demonstrated that these Hsp12s seem to play crucial physiological roles in C. elegans, for suppressing dauer formation and promoting both longevity and reproduction. A unique sHSP gene from Filarial nematode worm Brugia malayi was identified such that it encodes two products, one as a full-length Hsp12.6 protein and the other one having an N-terminal arm of normal length but lacks the C-terminal extension. This gene may represent an intermediate form in evolution from a common sHSP to a Hsp12. Together, our study offers insights on what biological functions the chaperone-defective sHSPs may exhibit and also implicates an evolutionary scenario for the unique Hsp12s subfamily.
Collapse
Affiliation(s)
- Xinmiao Fu
- Provincial University Key Laboratory of Cellular Stress Response and Metabolic Regulation, College of Life Sciences, Fujian Normal University, Fuzhou City, Fujian Province, China.,School of Life Sciences, Peking University, Beijing, China
| | - Anastasia N Ezemaduka
- School of Life Sciences, Peking University, Beijing, China.,Key Laboratory of Wetland Ecology and Environment, Northeast institute of Geography and Agroecology, Chinese Academy of Sciences, Changchun, China
| | - Xinping Lu
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Zengyi Chang
- School of Life Sciences, Peking University, Beijing, China
| |
Collapse
|
15
|
Phadte AS, Sluzala ZB, Fort PE. Therapeutic Potential of α-Crystallins in Retinal Neurodegenerative Diseases. Antioxidants (Basel) 2021; 10:1001. [PMID: 34201535 PMCID: PMC8300683 DOI: 10.3390/antiox10071001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 06/12/2021] [Accepted: 06/15/2021] [Indexed: 11/18/2022] Open
Abstract
The chaperone and anti-apoptotic activity of α-crystallins (αA- and αB-) and their derivatives has received increasing attention due to their tremendous potential in preventing cell death. While originally known and described for their role in the lens, the upregulation of these proteins in cells and animal models of neurodegenerative diseases highlighted their involvement in adaptive protective responses to neurodegeneration associated stress. However, several studies also suggest that chronic neurodegenerative conditions are associated with progressive loss of function of these proteins. Thus, while external supplementation of α-crystallin shows promise, their potential as a protein-based therapeutic for the treatment of chronic neurodegenerative diseases remains ambiguous. The current review aims at assessing the current literature supporting the anti-apoptotic potential of αA- and αB-crystallins and its potential involvement in retinal neurodegenerative diseases. The review further extends into potentially modulating the chaperone and the anti-apoptotic function of α-crystallins and the use of such functionally enhanced proteins for promoting neuronal viability in retinal neurodegenerative disease.
Collapse
Affiliation(s)
- Ashutosh S. Phadte
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA; (A.S.P.); (Z.B.S.)
| | - Zachary B. Sluzala
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA; (A.S.P.); (Z.B.S.)
| | - Patrice E. Fort
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA; (A.S.P.); (Z.B.S.)
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48105, USA
| |
Collapse
|
16
|
Sprague-Piercy MA, Rocha MA, Kwok AO, Martin RW. α-Crystallins in the Vertebrate Eye Lens: Complex Oligomers and Molecular Chaperones. Annu Rev Phys Chem 2021; 72:143-163. [PMID: 33321054 PMCID: PMC8062273 DOI: 10.1146/annurev-physchem-090419-121428] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
α-Crystallins are small heat-shock proteins that act as holdase chaperones. In humans, αA-crystallin is expressed only in the eye lens, while αB-crystallin is found in many tissues. α-Crystallins have a central domain flanked by flexible extensions and form dynamic, heterogeneous oligomers. Structural models show that both the C- and N-terminal extensions are important for controlling oligomerization through domain swapping. α-Crystallin prevents aggregation of damaged β- and γ-crystallins by binding to the client protein using a variety of binding modes. α-Crystallin chaperone activity can be compromised by mutation or posttranslational modifications, leading to protein aggregation and cataract. Because of their high solubility and their ability to form large, functional oligomers, α-crystallins are particularly amenable to structure determination by solid-state nuclear magnetic resonance (NMR) and solution NMR, as well as cryo-electron microscopy.
Collapse
Affiliation(s)
- Marc A Sprague-Piercy
- Department of Molecular Biology and Biochemistry, University of California, Irvine, California 92697, USA;
| | - Megan A Rocha
- Department of Chemistry, University of California, Irvine, California 92697, USA
| | - Ashley O Kwok
- Department of Chemistry, University of California, Irvine, California 92697, USA
| | - Rachel W Martin
- Department of Molecular Biology and Biochemistry, University of California, Irvine, California 92697, USA;
- Department of Chemistry, University of California, Irvine, California 92697, USA
| |
Collapse
|
17
|
Augusteyn RC. α‐crystallin: a review of its structure and function. Clin Exp Optom 2021; 87:356-66. [PMID: 15575808 DOI: 10.1111/j.1444-0938.2004.tb03095.x] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2004] [Revised: 06/23/2004] [Accepted: 07/18/2004] [Indexed: 11/28/2022] Open
Abstract
alpha-crystallin, the major protein of the mammalian lens in most species, is an aggregate assembled from two polypeptides, each with a molecular weight around 20,000 Da. It is polydisperse and can be isolated in a variety of forms, including spherical particles with molecular weights ranging upwards from about 200 kDa. Sequence comparisons reveal that it is a member of the small heat shock protein (shsp) family. These proteins are aggregates assembled from polypeptides of 10 to 25 kDa that share a common central domain of about 90 residues (the 'alpha-crystallin domain') with variable N- and C-terminal extensions. alpha-crystallin has been intensively studied for more than 50 years but its three-dimensional structure remains unknown because it has not been possible to obtain crystals for X-ray studies and it is too large for NMR measurements. Structural information has been derived from a variety of solution studies. Because of the protein's polydispersity, interpretation of data has been difficult. This led to different viewpoints and vigorous debate on its structure and properties. Recently, the crystal structures of two closely-related small heat shock proteins have been determined. These have provided some insight into the structure of a-crystallin and explanations of previous observations. Like many other heat shock proteins, alpha-crystallin exhibits chaperone-like properties, including the ability to prevent the precipitation of denatured proteins and to increase cellular tolerance to stress. It has been suggested that these functions are important for the maintenance of lens transparency and the prevention of cataract.
Collapse
Affiliation(s)
- Robert C Augusteyn
- Vision Cooperative Research Centre, University of NSW, Sydney, Australia
| |
Collapse
|
18
|
Tender T, Rahangdale RR, Balireddy S, Nampoothiri M, Sharma KK, Raghu Chandrashekar H. Melittin, a honeybee venom derived peptide for the treatment of chemotherapy-induced peripheral neuropathy. Med Oncol 2021; 38:52. [PMID: 33796975 PMCID: PMC8016801 DOI: 10.1007/s12032-021-01496-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 03/09/2021] [Indexed: 12/15/2022]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is the most prevalent neurological complication of cancer treatment which involves sensory and motor nerve dysfunction. Severe CIPN has been reported in around 5% of patients treated with single and up to 38% of patients treated with multiple chemotherapeutic agents. Present medications available for CIPN are the use of opioids, nonsteroidal anti-inflammatory agents, and tricyclic antidepressants, which are only marginally effective in treating neuropathic symptoms. In reality, symptom reappears after these drugs are discontinued. The pathogenesis of CIPN has not been sufficiently recognized and methods for the prevention and treatment of CIPN remain vulnerable to therapeutic problems. It has witnessed that the present medicines available for the disease offer only symptomatic relief for the short term and have severe adverse side effects. There is no standard treatment protocol for preventing, reducing, and treating CIPN. Therefore, there is a need to develop curative therapy that can be used to treat this complication. Melittin is the main pharmacological active constituent of honeybee venom and has therapeutic values including in chemotherapeutic-induced peripheral neuropathy. It has been shown that melittin and whole honey bee venom are effective in treating paclitaxel and oxaliplatin-induced peripheral neuropathy. The use of melittin against peripheral neuropathy caused by chemotherapy has been limited despite having strong therapeutic efficacy against the disease. Melittin mediated haemolysis is the key reason to restrict its use. In our study, it is found that α-Crystallin (an eye lens protein) is capable of inhibiting melittin-induced haemolysis which gives hope of using an appropriate combination of melittin and α-Crystallin in the treatment of CIPN. The review summarizes the efforts made by different research groups to address the concern with melittin in the treatment of chemotherapeutic-induced neuropathy. It also focuses on the possible approaches to overcome melittin-induced haemolysis.
Collapse
Affiliation(s)
- Tenzin Tender
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Rakesh Ravishankar Rahangdale
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Sridevi Balireddy
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Madhavan Nampoothiri
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - K Krishna Sharma
- Department of Ophthalmology and Biochemistry, University of Missouri - Columbia School of Medicine, Columbia, MO, 65211, USA
| | - Hariharapura Raghu Chandrashekar
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India.
| |
Collapse
|
19
|
Clinical characteristics of congenital lamellar cataract and myopia in a Chinese family. Biosci Rep 2021; 40:222026. [PMID: 32010934 PMCID: PMC7024846 DOI: 10.1042/bsr20191349] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 01/03/2020] [Accepted: 01/20/2020] [Indexed: 11/24/2022] Open
Abstract
To investigate the clinical characteristics and the genetic defect in a Chinese family with congenital lamellar cataract with myopia. Three generations of a single family were recruited in the present study. A detailed family history and clinical data were recorded. A total of 100 unrelated ethnically matched controls without family history of congenital cataracts and myopia were also recruited. Genomic DNA was extracted from peripheral blood leukocytes. The sequencing of candidate genes was performed to screen out the disease-causing mutation. The effects of amino acid changes on the structure of proteins were predicted by bioinformatics analysis. Affected individuals presented lamellar lens opacities and myopia. Direct sequencing revealed a heterozygous c. 34 C>T variation in the αA-crystallin protein (CRYAA) gene, which resulted in the replacement of a highly conserved arginine by cystine at codon 12 (p.R12C). This mutation co-segregated with all affected individuals and was not observed in unaffected members or the 100 normal controls. Bioinformatic analysis showed that a highly conserved region was located around Arg12, an increase in local hydrophobicity was shown around the substitution site and the secondary structure of the mutant CRYAA protein has been changed. This is the case of a congenital lamellar cataract phenotype with myopia associated with the mutation of Arg12Cys (p.R12C) in CRYAA. Our finding confirms the high rate of mutations at this dinucleotide. In addition, these results demonstrate a myopia susceptibility locus in this region, which might also be associated with the mutation in CRYAA.
Collapse
|
20
|
Schimansky A, Yadav JK. Amyloid cross-sequence interaction between Aβ(1-40) and αA(66-80) in relation to the pathogenesis of cataract. Int J Biol Macromol 2021; 179:61-70. [PMID: 33626371 DOI: 10.1016/j.ijbiomac.2021.02.111] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 01/29/2021] [Accepted: 02/15/2021] [Indexed: 10/22/2022]
Abstract
Alzheimer's disease (AD) and cataract represent two common protein misfolding diseases closely associated with aging. Growing evidence suggests that these two diseases may be interrelated with each other through cross-sequence interactions between β-amyloid (Aβ) peptide and the short aggregating peptides derived from proteolytic breakdown of α-crystallin. αΑ(66-80) is one of several peptides produced by the proteolytic breakdown of α-crystallin in aged eye lens. Although it is evident that the Aβ(1-40) and αΑ(66-80) coexist in aged eye lenses and both the peptides are known to form macromolecular assemblies, their cross-sequence interaction and the seeding behavior are not known. In this study, the aggregation behavior of αΑ(66-80) has been examined in the presence of Aβ(1-40) on using thioflavin T (ThT) based aggregation kinetics. The presence of monomeric Aβ(1-40) augmented the aggregation kinetics of αΑ(66-80) and reduced the lag time of αΑ(66-80) aggregation. However, the addition of Aβ(1-40) or αΑ(66-80) fibrils (seeds) didn't result in any change in the rate of αΑ(66-80) aggregation. In this in vitro study, we could show that the presence Aβ(1-40) has substantial effect on the aggregation of αΑ(66-80), which suggests a possible interaction between AD and cataract pathologies.
Collapse
Affiliation(s)
- Anna Schimansky
- Ulm University, Institute of Protein Biochemistry, Helmholtzstraße 8/1, 89081 Ulm, Germany
| | - Jay Kant Yadav
- Ulm University, Institute of Protein Biochemistry, Helmholtzstraße 8/1, 89081 Ulm, Germany; Department of Biotechnology, Central University of Rajasthan, NH-8 Bandarsindri, Kishangarh, Ajmer 305817, Rajasthan, India.
| |
Collapse
|
21
|
PV1 Protein from Plasmodium falciparum Exhibits Chaperone-Like Functions and Cooperates with Hsp100s. Int J Mol Sci 2020; 21:ijms21228616. [PMID: 33207549 PMCID: PMC7697860 DOI: 10.3390/ijms21228616] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 01/17/2023] Open
Abstract
Plasmodium falciparum parasitophorous vacuolar protein 1 (PfPV1), a protein unique to malaria parasites, is localized in the parasitophorous vacuolar (PV) and is essential for parasite growth. Previous studies suggested that PfPV1 cooperates with the Plasmodium translocon of exported proteins (PTEX) complex to export various proteins from the PV. However, the structure and function of PfPV1 have not been determined in detail. In this study, we undertook the expression, purification, and characterization of PfPV1. The tetramer appears to be the structural unit of PfPV1. The activity of PfPV1 appears to be similar to that of molecular chaperones, and it may interact with various proteins. PfPV1 could substitute CtHsp40 in the CtHsp104, CtHsp70, and CtHsp40 protein disaggregation systems. Based on these results, we propose a model in which PfPV1 captures various PV proteins and delivers them to PTEX through a specific interaction with HSP101.
Collapse
|
22
|
Development of a brain-permeable peptide nanofiber that prevents aggregation of Alzheimer pathogenic proteins. PLoS One 2020; 15:e0235979. [PMID: 32706773 PMCID: PMC7380640 DOI: 10.1371/journal.pone.0235979] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 06/25/2020] [Indexed: 01/09/2023] Open
Abstract
Alzheimer's disease (AD) is proposed to be induced by abnormal aggregation of amyloidβ in the brain. Here, we designed a brain-permeable peptide nanofiber drug from a fragment of heat shock protein to suppress aggregation of the pathogenic proteins. To facilitate delivery of the nanofiber into the brain, a protein transduction domain from Drosophila Antennapedia was incorporated into the peptide sequence. The resulting nanofiber efficiently suppressed the cytotoxicity of amyloid βby trapping amyloid β onto its hydrophobic nanofiber surface. Moreover, the intravenously or intranasally injected nanofiber was delivered into the mouse brain, and improved the cognitive function of an Alzheimer transgenic mouse model. These results demonstrate the potential therapeutic utility of nanofibers for the treatment of AD.
Collapse
|
23
|
Roskamp KW, Paulson CN, Brubaker WD, Martin RW. Function and Aggregation in Structural Eye Lens Crystallins. Acc Chem Res 2020; 53:863-874. [PMID: 32271004 DOI: 10.1021/acs.accounts.0c00014] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Crystallins are transparent, refractive proteins that contribute to the focusing power of the vertebrate eye lens. These proteins are extremely soluble and resist aggregation for decades, even under crowded conditions. Crystallins have evolved to avoid strong interprotein interactions and have unusual hydration properties. Crystallin aggregation resulting from mutation, damage, or aging can lead to cataract, a disease state characterized by opacity of the lens.Different aggregation mechanisms can occur, following multiple pathways and leading to aggregates with varied morphologies. Studies of variant proteins found in individuals with childhood-onset cataract have provided insight into the molecular factors underlying crystallin stability and solubility. Modulation of exposed hydrophobic surface is critical, as is preventing specific intermolecular interactions that could provide nucleation sites for aggregation. Biophysical measurements and structural biology techniques are beginning to provide a detailed picture of how crystallins crowd into the lens, providing high refractivity while avoiding excessively tight binding that would lead to aggregation.Despite the central biological importance of refractivity, relatively few experimental measurements have been made for lens crystallins. Our work and that of others have shown that hydration is important to the high refractive index of crystallin proteins, as are interactions between pairs of aromatic residues and potentially other specific structural features.This Account describes our efforts to understand both the functional and disease states of vertebrate eye lens crystallins, particularly the γ-crystallins. We use a variety of biophysical techniques, notably NMR spectroscopy, to investigate crystallin stability and solubility. In the first section, we describe efforts to understand the relative stability and aggregation propensity of different γS-crystallin variants. The second section focuses on interactions of these proteins with the holdase chaperone αB-crystallin. The third, fourth, and fifth sections explore different modes of aggregation available to crystallin proteins, and the final section highlights the importance of refractive index and the sometimes conflicting demands of selection for refractivity and solubility.
Collapse
Affiliation(s)
- Kyle W. Roskamp
- Department of Chemistry, University of California, Irvine, California 92697-2025, United States
| | - Carolyn N. Paulson
- Institute for Therapeutics Discovery and Development, University of Minnesota, Minneapolis, Minnesota 55414, United States
| | - William D. Brubaker
- SRI International, 333 Ravenswood Avenue, Menlo Park, California 94025, United States
| | - Rachel W. Martin
- Department of Chemistry, University of California, Irvine, California 92697-2025, United States
- Department of Molecular Biology and Biochemistry, University of California, Irvine, California 92697-3900, United States
| |
Collapse
|
24
|
Magami K, Kim I, Fujii N. A single Asp isomer substitution in an αA-crystallin-derived peptide induces a large change in peptide properties. Exp Eye Res 2020; 192:107930. [DOI: 10.1016/j.exer.2020.107930] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 01/07/2020] [Accepted: 01/08/2020] [Indexed: 10/25/2022]
|
25
|
Ramirez LM, Shekhtman A, Pande J. Hydrophobic residues of melittin mediate its binding to αA-crystallin. Protein Sci 2019; 29:572-588. [PMID: 31762096 DOI: 10.1002/pro.3792] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 11/13/2019] [Accepted: 11/15/2019] [Indexed: 01/01/2023]
Abstract
The molecular chaperone αA-crystallin, mainly localized in the human ocular lens, is believed to protect the lens from opacification and cataract, by suppressing the aggregation of the other lens proteins. The present study provides structural and thermodynamic insights into the ability of human αA-crystallin (HAA) to bind to its partially unfolded clients in the lens, using a small peptide, melittin from bee venom, as a model client. We characterized the thermodynamic parameters of the binding process between melittin and HAA through isothermal titration calorimetry (ITC), and found the binding to be endothermic and entropy-driven. We identified the amino acids in melittin important for binding to HAA by saturation-transfer difference (STD) nuclear magnetic resonance (NMR) experiments, and analysis of NMR line broadening upon titration of melittin with HAA. Our results suggest that hydrophobic residues Ile17 and Ile20 on the C-terminal region of melittin are in close contact with HAA in the melittin-HAA complex. Information obtained from NMR experiments was used to generate structural models of the melittin-HAA complex by molecular docking with high-ambiguity driven docking (HADDOCK). Structural models of the melittin-HAA complex reveal important principles underlying the interaction of HAA with its clients.
Collapse
Affiliation(s)
- Lisa M Ramirez
- Department of Chemistry, University at Albany, State University of New York, Albany, New York
| | - Alexander Shekhtman
- Department of Chemistry, University at Albany, State University of New York, Albany, New York
| | - Jayanti Pande
- Department of Chemistry, University at Albany, State University of New York, Albany, New York
| |
Collapse
|
26
|
Chauhan P, Ghosh KS. Inhibition of copper-induced aggregation of human γD-crystallin by rutin and studies on its role in molecular level for enhancing the chaperone activity of human αA-crystallin by using multi-spectroscopic techniques. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2019; 218:229-236. [PMID: 31003047 DOI: 10.1016/j.saa.2019.04.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 04/07/2019] [Accepted: 04/09/2019] [Indexed: 06/09/2023]
Abstract
Oxidative aggregation of γ-crystallins induced by copper in aged lens increases the lens opacity and causes cataract formation. Therefore, chelation of free Cu2+ by small molecules can inhibit metal-mediated aggregation of γ-crystallin. In this work, the inhibition potency of several naturally occurring flavonoid compounds was studied against aggregation of human γD-crystallin (HGD) mediated by copper ions. Among them, rutin demonstrated ~20% inhibition of HGD aggregation induced by Cu2+ through its metal chelation ability. Not only that, the chaperone activity of lens chaperone, human αA-crystallin (HAA) was found to be enhanced in the presence of rutin. Subsequently, the molecular interactions between HAA and rutin were investigated using fluorescence and CD spectroscopy to understand the molecular basis of the chaperone activity enhancement by rutin. Quenching of HAA fluorescence by rutin with a quenching constant in the order of ~105 M-1 depicts a complexation between them. Entropy driven process of complexation between HAA and rutin suggests significant involvement of hydrophobic interactions. Fluorescence resonance energy transfer between protein and ligand can occur at a distance of 2.73 nm. Synchronous fluorescence and circular dichroism spectroscopy revealed that protein-ligand interaction does not cause any notable conformational changes in HAA. Experimental observations have been well substantiated by docking.
Collapse
Affiliation(s)
- Priyanka Chauhan
- Department of Chemistry, National Institute of Technology Hamirpur, Himachal Pradesh 177005, India
| | - Kalyan S Ghosh
- Department of Chemistry, National Institute of Technology Hamirpur, Himachal Pradesh 177005, India.
| |
Collapse
|
27
|
Karmakar S, Biswas S, Das KP, Tripathy U. Surface plasmon resonance study of the interaction of 4,4′-dianilino-1,1′-binaphthyl-5,5′-disulfonic acid dipotassium salt (bis-ANS) and adenosine triphosphate (ATP) with oligomeric recombinant human lens αA-crystallin. CAN J CHEM 2019. [DOI: 10.1139/cjc-2018-0412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
α-Crystallin, an abundant mammalian lens protein made up of two subunits (αA- and αB-crystallin), is involved in the maintenance of the optimal refractive index in the lens. The protein is implicated in the pathophysiology of a large number of retinal diseases including cataract, age-related macular degeneration, diabetic retinopathy, and uveitis. α-Crystallin belongs to the small heat shock protein (sHSP) family, forms large oligomeric structures, and functions as a molecular chaperone appearing very early during embryonic development. To gain mechanistic insight into the structural and functional role of α-crystallin and its alterations in various retinal diseases, it is important to study the interaction chemistry with its known partners. The hydrophobic sites in α-crystallin have been studied extensively using environmentally sensitive fluorescent probes such as 4,4′-dianilino-1,1′-binaphthyl-5,5′-disulfonic acid dipotassium salt (bis-ANS) that interacts with both subunits of α-cystallin in 1:1 stoichiometry at 37 °C and diminishes the chaperone-like activity of the protein. Furthermore, it has been shown that ATP plays a crucial role in the association of α-crystallin with substrate proteins. We use surface plasmon resonance (SPR) to monitor the interactions of immobilized oligomeric recombinant αA subunit of human α-crystallin protein with bis-ANS and ATP. We assess the thermodynamic parameters and kinetics of such interactions at various temperatures. Our results indicate that bis-ANS binds to αA-crystallin with higher affinity when compared with ATP, although both αA-crystallin and αB-crystallin display fast interaction kinetics.
Collapse
Affiliation(s)
- Srabani Karmakar
- Department of Biotechnology, Techno India University, EM-4/1, Sector V, Salt Lake, Kolkata-700091, India
| | - Shrutidhara Biswas
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Guwahati-781029, India
| | - Kali P. Das
- Protein Chemistry Laboratory, Department of Chemistry, Bose Institute, 93/1 A.P.C. Road, Kolkata-700009, India
| | - Umakanta Tripathy
- Department of Applied Physics, Indian Institute of Technology (Indian School of Mines), Dhanbad-826004, India
| |
Collapse
|
28
|
A monoclonal antibody targeted to the functional peptide of αB-crystallin inhibits the chaperone and anti-apoptotic activities. J Immunol Methods 2019; 467:37-47. [PMID: 30738041 DOI: 10.1016/j.jim.2019.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 01/24/2019] [Accepted: 02/05/2019] [Indexed: 01/18/2023]
Abstract
αB-Crystallin is a member of the small heat shock protein family. It is a molecular chaperone and an anti-apoptotic protein. Previous studies have shown that the peptide (73DRFSVNLDVKHFSPEELKVKV93, hereafter referred to as peptain-1) from the core domain of αB-crystallin exhibits both chaperone and anti-apoptotic properties similar to the parent protein. We developed a mouse monoclonal antibody against peptain-1 with the aim of blocking the functions of αB-crystallin. The antibody reacted with peptain-1, it did not react with the chaperone peptide of αA-crystallin. The antibody strongly reacted with human recombinant αB-crystallin but weakly with Hsp20; it did not react with αA-crystallin or Hsp27. The antibody specifically reacted with αB-crystallin in human and mouse lens proteins but not with αA-crystallin. The antibody reacted with αB-crystallin in human lens epithelial cells, human retinal endothelial cells, and with peptain-1 in peptain-1-transduced cells. Unlike the commercial antibodies against αB-crystallin, the antibody against peptain-1 inhibited the chaperone and anti-apoptotic activities of peptain-1. The antibody might find use in inhibiting αB-crystallin's chaperone and anti-apoptotic activities in diseases where αB-crystallin is a causative or contributing factor.
Collapse
|
29
|
Gliniewicz EF, Chambers KM, De Leon ER, Sibai D, Campbell HC, McMenimen KA. Chaperone-like activity of the N-terminal region of a human small heat shock protein and chaperone-functionalized nanoparticles. Proteins 2019; 87:401-415. [PMID: 30684363 DOI: 10.1002/prot.25662] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 01/03/2019] [Accepted: 01/22/2019] [Indexed: 11/10/2022]
Abstract
Small heat shock proteins (sHsps) are molecular chaperones employed to interact with a diverse range of substrates as the first line of defense against cellular protein aggregation. The N-terminal region (NTR) is implicated in defining features of sHsps; notably in their ability to form dynamic and polydisperse oligomers, and chaperone activity. The physiological relevance of oligomerization and chemical-scale mode(s) of chaperone function remain undefined. We present novel chemical tools to investigate chaperone activity and substrate specificity of human HspB1 (B1NTR), through isolation of B1NTR and development of peptide-conjugated gold nanoparticles (AuNPs). We demonstrate that B1NTR exhibits chaperone capacity for some substrates, determined by anti-aggregation assays and size-exclusion chromatography. The importance of protein dynamics and multivalency on chaperone capacity was investigated using B1NTR-conjugated AuNPs, which exhibit concentration-dependent chaperone activity for some substrates. Our results implicate sHsp NTRs in chaperone activity, and demonstrate the therapeutic potential of sHsp-AuNPs in rescuing aberrant protein aggregation.
Collapse
Affiliation(s)
- Emily F Gliniewicz
- Department of Chemistry, Mount Holyoke College, South Hadley, Massachusetts
| | - Kelly M Chambers
- Department of Chemistry, Mount Holyoke College, South Hadley, Massachusetts
| | | | - Diana Sibai
- Department of Chemistry, Mount Holyoke College, South Hadley, Massachusetts
| | - Helen C Campbell
- Department of Chemistry, Mount Holyoke College, South Hadley, Massachusetts
| | | |
Collapse
|
30
|
Abstract
Melittin is an extensively studied, 26-residue toxic peptide from honey bee venom. Because of its versatility in adopting a variety of secondary (helix or coil) and quaternary (monomer or tetramer) structures in various environments, melittin has been the focus of numerous investigations as a model peptide in protein folding studies as well as in studies involving binding to proteins, lipids, and polysaccharides. A significant body of evidence supports the view that melittin binds to these macromolecules in a predominantly helical conformation, but detailed structural knowledge of this conformation is lacking. In this report, we present nuclear magnetic resonance (NMR)-based structural insights into the helix formation of recombinant melittin in the presence of trifluoroethanol (TFE): a known secondary structure inducer in peptides. These studies were performed at neutral pH, with micromolar amounts of the peptide. Using nuclear Overhauser effect (NOE)-derived distance restraints from three-dimensional NMR spectra, we determined the atomic resolution solution NMR structure of recombinant melittin bearing a TFE-stabilized helix. To circumvent the complications with structure determination of small peptides with high conformational flexibility, we developed a workflow for enhancing proton NOEs by increasing the viscosity of the medium. In the TFE-containing medium, recombinant monomeric melittin forms a long, continuous helical structure, which consists of the N- and C-terminal α-helices and the noncanonical 310-helix in the middle. The noncanonical 310-helix is missing in the previously solved X-ray structure of tetrameric melittin and the NMR structure of melittin in methanol. Melittin's structure in TFE-containing medium provides insights into melittin's conformational transitions, which are relevant to the peptide's interactions with its biological targets.
Collapse
Affiliation(s)
- Lisa S Ramirez
- Department of Chemistry , State University of New York at Albany , Albany , New York 12222 , United States
| | - Jayanti Pande
- Department of Chemistry , State University of New York at Albany , Albany , New York 12222 , United States
| | - Alexander Shekhtman
- Department of Chemistry , State University of New York at Albany , Albany , New York 12222 , United States
| |
Collapse
|
31
|
RETRACTED: Peptide-induced formation of protein aggregates and amyloid fibrils in human and guinea pig αA-crystallins under physiological conditions of temperature and pH. Exp Eye Res 2018; 179:193-205. [PMID: 30448341 DOI: 10.1016/j.exer.2018.11.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 10/17/2018] [Accepted: 11/14/2018] [Indexed: 02/07/2023]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (https://www.elsevier.com/about/our-business/policies/article-withdrawal).
This article has been retracted at the request of the authors. The senior author contacted the journal in a forthright manner, in an effort to preserve the scientific integrity of the literature, after discovering a significant error in the results reported in the article. The authors were recently made aware of a paper by Kim et al. (Nature Commun. 2019) which shows a spirosome structure (the enzyme aldehyde-alcohol dehydrogenase) present in E. coli (Fig. 5a) that is very similar to the structure the authors thought formed when synthetic alpha A crystallin (66-80) peptide was incubated for 24 h with recombinant guinea pig alpha A insert crystallin (see Kumarasamy et al., Figs. 7C and F, and Fig. 9). Subsequent to publication of their report, the authors later found a number of images that showed what appeared to be the same structure present in samples of their presumably purified recombinant guinea pig alpha A insert crystallin which had been incubated without peptide for 24 h. Hence, the authors now conclude that the structures shown in Figs. 7C and F, and Fig. 9 of their article published in this journal are actually due to E. coli contaminant aldehyde-alcohol dehydrogenase. The authors deeply regret this error and any inconvenience it may have caused.
Collapse
|
32
|
Phadte AS, Santhoshkumar P, Sharma KK. Characterization of an N-terminal mutant of αA-crystallin αA-R21Q associated with congenital cataract. Exp Eye Res 2018; 174:185-195. [PMID: 29782825 DOI: 10.1016/j.exer.2018.05.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 04/06/2018] [Accepted: 05/16/2018] [Indexed: 01/20/2023]
Abstract
Several mutations associated with congenital cataracts in human beings target conserved arginine residues in αA-crystallin. The N-terminal region of αA-crystallin is a "mutational hotspot," with multiple cataract-related mutations reported in this region. Two mutations at arginine 21 in the N-terminal domain of αA-crystallin - αA-R21L and αA-R21W have been associated with congenital cataract. A third mutant of R21, αA-R21Q, was recently identified to be associated with congenital cataract in a South Australian family. The point mutation was reported to compromise the quaternary structure of αA-crystallin by preventing its assembly into higher ordered oligomers. To assess the effect of the αA-R21Q mutation on αA-crystallin function, recombinant αA-R21Q was expressed, purified and characterized in vitro. Compared to wild-type αA-crystallin, the recombinant αA-R21Q exhibits enhanced chaperone-like activity, increased surface hydrophobicity, lesser stability in urea and increased susceptibility to digestion by trypsin. αA-R21Q demonstrated increased binding affinity towards unfolding ADH and bovine lens fiber cell membranes. αA-R21Q homo-oligomers and hetero-oligomers also prevented H2O2-induced apoptosis in ARPE-19 cells. Taken together, αA-R21Q exhibited a gain of function despite subtle structural differences as compared to wild-type αA-crystallin. This study further validates the involvement of arginine 21 in regulating αA-crystallin structure and function.
Collapse
Affiliation(s)
- Ashutosh S Phadte
- Department of Ophthalmology, University of Missouri School of Medicine, Columbia, MO 65212, United States; Department of Biochemistry, University of Missouri, Columbia, MO 65211, United States
| | - Puttur Santhoshkumar
- Department of Ophthalmology, University of Missouri School of Medicine, Columbia, MO 65212, United States
| | - K Krishna Sharma
- Department of Ophthalmology, University of Missouri School of Medicine, Columbia, MO 65212, United States; Department of Biochemistry, University of Missouri, Columbia, MO 65211, United States.
| |
Collapse
|
33
|
Ramirez L, Shekhtman A, Pande J. Nuclear Magnetic Resonance-Based Structural Characterization and Backbone Dynamics of Recombinant Bee Venom Melittin. Biochemistry 2018; 57:2775-2785. [PMID: 29668274 DOI: 10.1021/acs.biochem.8b00156] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In recent years, there has been a resurgence of interest in melittin and its variants as their therapeutic potential has become increasingly evident. Melittin is a 26-residue peptide and a toxic component of honey bee venom. The versatility of melittin in interacting with various biological substrates, such as membranes, glycosaminoglycans, and a variety of proteins, has inspired a slew of studies that aim to improve our understanding of the structural basis of such interactions. However, these studies have largely focused on melittin solutions at high concentrations (>1 mM), even though melittin is generally effective at lower (micromolar) concentrations. Here we present high-resolution nuclear magnetic resonance studies in the lower-concentration regime using a novel method to produce isotope-labeled (15N and 13C) recombinant melittin. We provide residue-specific structural characterization of melittin in dilute aqueous solution and in 2,2,2-trifluoroethanol/water mixtures, which mimic melittin structure-function and interactions in aqueous and membrane-like environments, respectively. We find that the cis-trans isomerization of Pro14 is key to changes in the secondary structure of melittin. Thus, this study provides residue-specific structural information about melittin in the free state and in a model of the substrate-bound state. These results, taken together with published work from other laboratories, reveal the peptide's structural versatility that resembles that of intrinsically disordered proteins and peptides.
Collapse
Affiliation(s)
- Lisa Ramirez
- Department of Chemistry , University at Albany, State University of New York , Albany , New York 12222 , United States
| | - Alexander Shekhtman
- Department of Chemistry , University at Albany, State University of New York , Albany , New York 12222 , United States
| | - Jayanti Pande
- Department of Chemistry , University at Albany, State University of New York , Albany , New York 12222 , United States
| |
Collapse
|
34
|
Raju M, Santhoshkumar P, Sharma KK. Cell-penetrating Chaperone Peptide Prevents Protein Aggregation And Protects Against Cell Apoptosis. ACTA ACUST UNITED AC 2017; 2. [PMID: 30271873 DOI: 10.1002/adbi.201700095] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Many of the newly discovered therapeutic peptides and molecules are limited by their inability to cross the cell membrane. In the present study we employed a cell penetrating peptide (CPP), VPTLK, derived from Ku70 protein, to facilitate the entry of a mini-chaperone across the cell membrane. Our previous studies suggest that the mini-chaperone peptide representing the chaperone site in αA-crystallin, which can inhibit protein aggregation associated with proteopathies, has therapeutic potential. We have prepared a synthetic mini-chaperone by fusing the VPTLK sequence to N-terminus of mini-chaperone (FVIFLDVKHFSPEDLTVKGRD) to get VPTLKFVIFLDVKHFSPEDLTVKGRD peptide, which we call "CPPGRD." The amino acids, GRD, were added to increase the solubility of the peptide. The chaperone-like function of CPPGRD was measured using unfolding conditions for alcohol dehydrogenase and α-lactalbumin. The anti-apoptotic action of the peptide chaperone was evaluated using H2O2-induced Cos-7 and ARPE-19 cell apoptosis assays. The results show that the CPPGRD has both chaperone function and anti-apoptotic activity. Additionally, the CPPGRD was found to prevent β-amyloid fibril formation and suppress β-amyloid toxicity. The present study demonstrates that the CPPGRD protects unfolding proteins from aggregation and prevents cellular apoptosis. Therefore, the CPPGRD is a mini-chaperone with potential to become a therapeutic agent for protein aggregation diseases.
Collapse
Affiliation(s)
- Murugesan Raju
- Department of Ophthalmology, University of Missouri School of Medicine, Columbia, Missouri 65212
| | - Puttur Santhoshkumar
- Department of Ophthalmology, University of Missouri School of Medicine, Columbia, Missouri 65212
| | - K Krishna Sharma
- Department of Ophthalmology, University of Missouri School of Medicine, Columbia, Missouri 65212.,Department of Biochemistry, University of Missouri School of Medicine, Columbia, Missouri 65212
| |
Collapse
|
35
|
Garvey M, Ecroyd H, Ray NJ, Gerrard JA, Carver JA. Functional Amyloid Protection in the Eye Lens: Retention of α-Crystallin Molecular Chaperone Activity after Modification into Amyloid Fibrils. Biomolecules 2017; 7:biom7030067. [PMID: 28895938 PMCID: PMC5618248 DOI: 10.3390/biom7030067] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 08/21/2017] [Accepted: 09/07/2017] [Indexed: 11/16/2022] Open
Abstract
Amyloid fibril formation occurs from a wide range of peptides and proteins and is typically associated with a loss of protein function and/or a gain of toxic function, as the native structure of the protein undergoes major alteration to form a cross β-sheet array. It is now well recognised that some amyloid fibrils have a biological function, which has led to increased interest in the potential that these so-called functional amyloids may either retain the function of the native protein, or gain function upon adopting a fibrillar structure. Herein, we investigate the molecular chaperone ability of α-crystallin, the predominant eye lens protein which is composed of two related subunits αA- and αB-crystallin, and its capacity to retain and even enhance its chaperone activity after forming aggregate structures under conditions of thermal and chemical stress. We demonstrate that both eye lens α-crystallin and αB-crystallin (which is also found extensively outside the lens) retain, to a significant degree, their molecular chaperone activity under conditions of structural change, including after formation into amyloid fibrils and amorphous aggregates. The results can be related directly to the effects of aging on the structure and chaperone function of α-crystallin in the eye lens, particularly its ability to prevent crystallin protein aggregation and hence lens opacification associated with cataract formation.
Collapse
Affiliation(s)
- Megan Garvey
- CSL Limited, 45 Poplar Road, Parkville, VIC 3052, Australia.
| | - Heath Ecroyd
- School of Biological Sciences and the Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong NSW 2522, Australia.
| | - Nicholas J Ray
- Research School of Chemistry, The Australian National University, Acton ACT 2601, Australia.
| | - Juliet A Gerrard
- School of Biological Science and School of Chemical Science, University of Auckland, Auckland 1010, New Zealand.
| | - John A Carver
- Research School of Chemistry, The Australian National University, Acton ACT 2601, Australia.
| |
Collapse
|
36
|
Mallik PK, Shi H, Pande J. RNA aptamers targeted for human αA-crystallin do not bind αB-crystallin, and spare the α-crystallin domain. Biochem Biophys Res Commun 2017; 491:423-428. [PMID: 28720498 DOI: 10.1016/j.bbrc.2017.07.085] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 07/14/2017] [Indexed: 11/27/2022]
Abstract
The molecular chaperones, α-crystallins, belong to the small heat shock protein (sHSP) family and prevent the aggregation and insolubilization of client proteins. Studies in vivo have shown that the chaperone activity of the α-crystallins is raised or lowered in various disease states. Therefore, the development of tools to control chaperone activity may provide avenues for therapeutic intervention, as well as enable a molecular understanding of chaperone function. The major human lens α-crystallins, αA- (HAA) and αB- (HAB), share 57% sequence identity and show similar activity towards some clients, but differing activities towards others. Notably, both crystallins contain the "α-crystallin domain" (ACD, the primary client binding site), like all other members of the sHSP family. Here we show that RNA aptamers selected for HAA, in vitro, exhibit specific affinity to HAA but do not bind HAB. Significantly, these aptamers also exclude the ACD. This study thus demonstrates that RNA aptamers against sHSPs can be designed that show high affinity and specificity - yet exclude the primary client binding region - thereby facilitating the development of RNA aptamer-based therapeutic intervention strategies.
Collapse
Affiliation(s)
- Prabhat K Mallik
- Department of Chemistry, University at Albany, State University of New York, 1400 Washington Avenue, Albany 12222, N.Y, United States
| | - Hua Shi
- Department of Biological Sciences, University at Albany, State University of New York, 1400 Washington Avenue, Albany 12222, N.Y, United States
| | - Jayanti Pande
- Department of Chemistry, University at Albany, State University of New York, 1400 Washington Avenue, Albany 12222, N.Y, United States.
| |
Collapse
|
37
|
Carver JA, Grosas AB, Ecroyd H, Quinlan RA. The functional roles of the unstructured N- and C-terminal regions in αB-crystallin and other mammalian small heat-shock proteins. Cell Stress Chaperones 2017; 22:627-638. [PMID: 28391594 PMCID: PMC5465038 DOI: 10.1007/s12192-017-0789-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 03/06/2017] [Accepted: 03/16/2017] [Indexed: 01/18/2023] Open
Abstract
Small heat-shock proteins (sHsps), such as αB-crystallin, are one of the major classes of molecular chaperone proteins. In vivo, under conditions of cellular stress, sHsps are the principal defence proteins that prevent large-scale protein aggregation. Progress in determining the structure of sHsps has been significant recently, particularly in relation to the conserved, central and β-sheet structured α-crystallin domain (ACD). However, an understanding of the structure and functional roles of the N- and C-terminal flanking regions has proved elusive mainly because of their unstructured and dynamic nature. In this paper, we propose functional roles for both flanking regions, based around three properties: (i) they act in a localised crowding manner to regulate interactions with target proteins during chaperone action, (ii) they protect the ACD from deleterious amyloid fibril formation and (iii) the flexibility of these regions, particularly at the extreme C-terminus in mammalian sHsps, provides solubility for sHsps under chaperone and non-chaperone conditions. In the eye lens, these properties are highly relevant as the crystallin proteins, in particular the two sHsps αA- and αB-crystallin, are present at very high concentrations.
Collapse
Affiliation(s)
- John A Carver
- Research School of Chemistry, The Australian National University, Acton, ACT, 2601, Australia.
| | - Aidan B Grosas
- Research School of Chemistry, The Australian National University, Acton, ACT, 2601, Australia
| | - Heath Ecroyd
- School of Biological Sciences and the Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, 2522, Australia
| | - Roy A Quinlan
- Department of Biosciences, Durham University, Durham, DH1 3LE, UK
| |
Collapse
|
38
|
Mymrikov EV, Daake M, Richter B, Haslbeck M, Buchner J. The Chaperone Activity and Substrate Spectrum of Human Small Heat Shock Proteins. J Biol Chem 2016; 292:672-684. [PMID: 27909051 DOI: 10.1074/jbc.m116.760413] [Citation(s) in RCA: 110] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2016] [Revised: 11/12/2016] [Indexed: 11/06/2022] Open
Abstract
Small heat shock proteins (sHsps) are a ubiquitous family of molecular chaperones that suppress the unspecific aggregation of miscellaneous proteins. Multicellular organisms contain a large number of different sHsps, raising questions as to whether they function redundantly or are specialized in terms of substrates and mechanism. To gain insight into this issue, we undertook a comparative analysis of the eight major human sHsps on the aggregation of both model proteins and cytosolic lysates under standardized conditions. We discovered that sHsps, which form large oligomers (HspB1/Hsp27, HspB3, HspB4/αA-crystallin, and HspB5/αB-crystallin) are promiscuous chaperones, whereas the chaperone activity of the other sHsps is more substrate-dependent. However, all human sHsps analyzed except HspB7 suppressed the aggregation of cytosolic proteins of HEK293 cells. We identified ∼1100 heat-sensitive HEK293 proteins, 12% of which could be isolated in complexes with sHsps. Analysis of their biochemical properties revealed that most of the sHsp substrates have a molecular mass from 50 to 100 kDa and a slightly acidic pI (5.4-6.8). The potency of the sHsps to suppress aggregation of model substrates is correlated with their ability to form stable substrate complexes; especially HspB1 and HspB5, but also B3, bind tightly to a variety of proteins, whereas fewer substrates were detected in complex with the other sHsps, although these were also efficient in preventing the aggregation of cytosolic proteins.
Collapse
Affiliation(s)
- Evgeny V Mymrikov
- From the Center for Integrated Protein Science at the Department Chemie, Technische Universität München, Lichtenbergstrasse 4, 85748 Garching, Germany
| | - Marina Daake
- From the Center for Integrated Protein Science at the Department Chemie, Technische Universität München, Lichtenbergstrasse 4, 85748 Garching, Germany
| | - Bettina Richter
- From the Center for Integrated Protein Science at the Department Chemie, Technische Universität München, Lichtenbergstrasse 4, 85748 Garching, Germany
| | - Martin Haslbeck
- From the Center for Integrated Protein Science at the Department Chemie, Technische Universität München, Lichtenbergstrasse 4, 85748 Garching, Germany
| | - Johannes Buchner
- From the Center for Integrated Protein Science at the Department Chemie, Technische Universität München, Lichtenbergstrasse 4, 85748 Garching, Germany
| |
Collapse
|
39
|
Waku T, Tanaka N. Recent advances in nanofibrous assemblies based on β-sheet-forming peptides for biomedical applications. POLYM INT 2016. [DOI: 10.1002/pi.5195] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Tomonori Waku
- Faculty of Molecular Chemistry and Engineering; Kyoto Institute of Technology; Gosyokaido-cho, Matsugasaki Sakyo-ku Kyoto 606-8585 Japan
| | - Naoki Tanaka
- Faculty of Molecular Chemistry and Engineering; Kyoto Institute of Technology; Gosyokaido-cho, Matsugasaki Sakyo-ku Kyoto 606-8585 Japan
| |
Collapse
|
40
|
Santhoshkumar P, Karmakar S, Sharma KK. Structural and functional consequences of chaperone site deletion in αA-crystallin. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2016; 1864:1529-38. [PMID: 27524665 DOI: 10.1016/j.bbapap.2016.08.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 08/05/2016] [Accepted: 08/09/2016] [Indexed: 01/29/2023]
Abstract
The chaperone-like activity of αA-crystallin has an important role in maintaining lens transparency. Previously we identified residues 70-88 as a chaperone site in αA-crystallin. In this study, we deleted the chaperone site residues to generate αAΔ70-76 and αAΔ70-88 mutants and investigated if there are additional substrate-binding sites in αA-crystallin. Both mutant proteins when expressed in E. coli formed inclusion bodies, and on solubilizing and refolding, they exhibited similar structural properties, with a 2- to 3-fold increase in molar mass compared to the molar mass of wild-type protein. The deletion mutants were less stable than the wild-type αA-crystallin. Functionally αAΔ70-88 was completely inactive as a chaperone, while αAΔ70-76 demonstrated a 40-50% reduction in anti-aggregation activity against alcohol dehydrogenase (ADH). Deletion of residues 70-88 abolished the ADH binding sites in αA-crystallin at physiological temperature. At 45°C, cryptic ADH binding site(s) became exposed, which contributed subtly to the chaperone-like activity of αAΔ70-88. Both of the deletion mutants were completely inactive in suppressing aggregation of βL-crystallin at 53°C. The mutants completely lost the anti-apoptotic property that αA-crystallin exhibits while they protected ARPE-19 (a human retinal pigment epithelial cell line) and primary human primary lens epithelial (HLE) cells from oxidative stress. Our studies demonstrate that residues 70-88 in αA-crystallin act as a primary substrate binding site and account for the bulk of the total chaperone activity. The β3 and β4 strands in αA-crystallin comprising 70-88 residues play an important role in maintenance of the structure and in preventing aggregation of denaturing proteins.
Collapse
Affiliation(s)
- Puttur Santhoshkumar
- Department of Ophthalmology, University of Missouri School of Medicine, Columbia, MO 65212, United States.
| | - Srabani Karmakar
- Department of Ophthalmology, University of Missouri School of Medicine, Columbia, MO 65212, United States
| | - Krishna K Sharma
- Department of Ophthalmology, University of Missouri School of Medicine, Columbia, MO 65212, United States; Department of Biochemistry, University of Missouri School of Medicine, Columbia, MO 65212, United States.
| |
Collapse
|
41
|
Quintanar L, Domínguez-Calva JA, Serebryany E, Rivillas-Acevedo L, Haase-Pettingell C, Amero C, King JA. Copper and Zinc Ions Specifically Promote Nonamyloid Aggregation of the Highly Stable Human γ-D Crystallin. ACS Chem Biol 2016; 11:263-72. [PMID: 26579725 DOI: 10.1021/acschembio.5b00919] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Cataract is the leading cause of blindness in the world. It results from aggregation of eye lens proteins into high-molecular-weight complexes, causing light scattering and lens opacity. Copper and zinc concentrations in cataractous lens are increased significantly relative to a healthy lens, and a variety of experimental and epidemiological studies implicate metals as potential etiological agents for cataract. The natively monomeric, β-sheet rich human γD (HγD) crystallin is one of the more abundant proteins in the core of the lens. It is also one of the most thermodynamically stable proteins in the human body. Surprisingly, we found that both Cu(II) and Zn(II) ions induced rapid, nonamyloid aggregation of HγD, forming high-molecular-weight light-scattering aggregates. Unlike Zn(II), Cu(II) also substantially decreased the thermal stability of HγD and promoted the formation of disulfide-bridged dimers, suggesting distinct aggregation mechanisms. In both cases, however, metal-induced aggregation depended strongly on temperature and was suppressed by the human lens chaperone αB-crystallin (HαB), implicating partially folded intermediates in the aggregation process. Consistently, distinct site-specific interactions of Cu(II) and Zn(II) ions with the protein and conformational changes in specific hinge regions were identified by nuclear magnetic resonance. This study provides insights into the mechanisms of metal-induced aggregation of one of the more stable proteins in the human body, and it reveals a novel and unexplored bioinorganic facet of cataract disease.
Collapse
Affiliation(s)
- Liliana Quintanar
- Departamento
de Química, Centro de Investigación y de Estudios Avanzados (Cinvestav), 07360 Mexico City, México
| | - José A. Domínguez-Calva
- Departamento
de Química, Centro de Investigación y de Estudios Avanzados (Cinvestav), 07360 Mexico City, México
| | - Eugene Serebryany
- Department
of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Lina Rivillas-Acevedo
- Centro
de Investigaciones Químicas, Instituto de Investigación
en Ciencias Básicas y Aplicadas, Universidad Autónoma del Estado de Morelos, 62209 Cuernavaca, México
| | - Cameron Haase-Pettingell
- Department
of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Carlos Amero
- Centro
de Investigaciones Químicas, Instituto de Investigación
en Ciencias Básicas y Aplicadas, Universidad Autónoma del Estado de Morelos, 62209 Cuernavaca, México
| | - Jonathan A. King
- Department
of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
42
|
Tiwary E, Hegde S, Purushotham S, Deivanayagam C, Srivastava O. Interaction of βA3-Crystallin with Deamidated Mutants of αA- and αB-Crystallins. PLoS One 2015; 10:e0144621. [PMID: 26657544 PMCID: PMC4691197 DOI: 10.1371/journal.pone.0144621] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 10/22/2015] [Indexed: 11/18/2022] Open
Abstract
Interaction among crystallins is required for the maintenance of lens transparency. Deamidation is one of the most common post-translational modifications in crystallins, which results in incorrect interaction and leads to aggregate formation. Various studies have established interaction among the α- and β-crystallins. Here, we investigated the effects of the deamidation of αA- and αB-crystallins on their interaction with βA3-crystallin using surface plasmon resonance (SPR) and fluorescence lifetime imaging microscopy-fluorescence resonance energy transfer (FLIM-FRET) methods. SPR analysis confirmed adherence of WT αA- and WT αB-crystallins and their deamidated mutants with βA3-crystallin. The deamidated mutants of αA–crystallin (αA N101D and αA N123D) displayed lower adherence propensity for βA3-crystallin relative to the binding affinity shown by WT αA-crystallin. Among αB-crystallin mutants, αB N78D displayed higher adherence propensity whereas αB N146D mutant showed slightly lower binding affinity for βA3-crystallin relative to that shown by WT αB-crystallin. Under the in vivo condition (FLIM-FRET), both αA-deamidated mutants (αA N101D and αA N123D) exhibited strong interaction with βA3-crystallin (32±4% and 36±4% FRET efficiencies, respectively) compared to WT αA-crystallin (18±4%). Similarly, the αB N78D and αB N146D mutants showed strong interaction (36±4% and 22±4% FRET efficiencies, respectively) with βA3-crystallin compared to 18±4% FRET efficiency of WT αB-crystallin. Further, FLIM-FRET analysis of the C-terminal domain (CTE), N-terminal domain (NTD), and core domain (CD) of αA- and αB-crystallins with βA3-crystallin suggested that interaction sites most likely reside in the αA CTE and αB NTD regions, respectively, as these domains showed the highest FRET efficiencies. Overall, results suggest that similar to WT αA- and WTαB-crystallins, the deamidated mutants showed strong interactionfor βA3-crystallin. Variable in vitro and in vivo interactions are most likely due to the mutant’s large size oligomers, reduced hydrophobicity, and altered structures. Together, the results suggest that deamidation of α-crystallin may facilitate greater interaction and the formation of large oligomers with other crystallins, and this may contribute to the cataractogenic mechanism.
Collapse
Affiliation(s)
- Ekta Tiwary
- Department of Vision Sciences, School of Optometry, University of Alabama at Birmingham, Birmingham, Alabama, 35294, United States of America
| | - Shylaja Hegde
- Department of Vision Sciences, School of Optometry, University of Alabama at Birmingham, Birmingham, Alabama, 35294, United States of America
| | - Sangeetha Purushotham
- Department of Vision Sciences/Centre for Structural Biology, University of Alabama at Birmingham, Birmingham, Alabama, 35294, United States of America
| | - Champion Deivanayagam
- Department of Vision Sciences/Centre for Structural Biology, University of Alabama at Birmingham, Birmingham, Alabama, 35294, United States of America
| | - Om Srivastava
- Department of Vision Sciences, School of Optometry, University of Alabama at Birmingham, Birmingham, Alabama, 35294, United States of America
- * E-mail:
| |
Collapse
|
43
|
Cetinel S, Montemagno C. Nanotechnology for the Prevention and Treatment of Cataract. Asia Pac J Ophthalmol (Phila) 2015; 4:381-7. [PMID: 26716434 DOI: 10.1097/apo.0000000000000156] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
PURPOSE The purpose of this article was to review recent advances in the applications of nanotechnology in cataract treatment and prevention strategies. DESIGN A literature review on the use of nanotechnology for the prevention and treatment of cataract was done. METHODS Research articles about nanotechnology-based treatments and prevention technologies for cataract were searched on Web of Science, and the most recent advances were reported. RESULTS Nonsteroid anti-inflammatory drugs, natural antioxidants, biologic and chemical chaperones, and chaperones such as molecules have found great application in preventing and treating cataracts. Current scientific research on new treatment strategies, which focuses on the biochemical basis of the disease, will likely result in new anticataract agents. However, none of the drug formulations will be approved for use unless efficient delivery is promised. Nanoparticle engineering together with biomimetic strategies enable the development of next-generation, more efficient, less complex, and personalized treatments. CONCLUSIONS The only currently available treatment for cataracts, surgical replacement of the opacified lens, is not an easily accessible option in developing countries. New treatment strategies based on topical drugs would enable treatment to reach massive populations facing the threat of blindness and more effectively deal with the postsurgical complications. Nanotechnology plays a key role in improving drug delivery systems with enhanced controlled release, targeted delivery, and bioavailability to overcome diffusion limitations in the eye.
Collapse
Affiliation(s)
- Sibel Cetinel
- From the *Chemical and Materials Engineering and †Ingenuity Lab, University of Alberta, Edmonton, Alberta, Canada
| | | |
Collapse
|
44
|
Bakthisaran R, Akula KK, Tangirala R, Rao CM. Phosphorylation of αB-crystallin: Role in stress, aging and patho-physiological conditions. Biochim Biophys Acta Gen Subj 2015; 1860:167-82. [PMID: 26415747 DOI: 10.1016/j.bbagen.2015.09.017] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 09/22/2015] [Accepted: 09/23/2015] [Indexed: 01/18/2023]
Abstract
BACKGROUND αB-crystallin, once thought to be a lenticular protein, is ubiquitous and has critical roles in several cellular processes that are modulated by phosphorylation. Serine residues 19, 45 and 59 of αB-crystallin undergo phosphorylation. Phosphorylation of S45 is mediated by p44/42 MAP kinase, whereas S59 phosphorylation is mediated by MAPKAP kinase-2. Pathway involved in S19 phosphorylation is not known. SCOPE OF REVIEW The review highlights the role of phosphorylation in (i) oligomeric structure, stability and chaperone activity, (ii) cellular processes such as apoptosis, myogenic differentiation, cell cycle regulation and angiogenesis, and (iii) aging, stress, cardiomyopathy-causing αB-crystallin mutants, and in other diseases. MAJOR CONCLUSIONS Depending on the context and extent of phosphorylation, αB-crystallin seems to confer beneficial or deleterious effects. Phosphorylation alters structure, stability, size distribution and dynamics of the oligomeric assembly, thus modulating chaperone activity and various cellular processes. Phosphorylated αB-crystallin has a tendency to partition to the cytoskeleton and hence to the insoluble fraction. Low levels of phosphorylation appear to be protective, while hyperphosphorylation has negative implications. Mutations in αB-crystallin, such as R120G, Q151X and 464delCT, associated with inherited myofibrillar myopathy lead to hyperphosphorylation and intracellular inclusions. An ongoing study in our laboratory with phosphorylation-mimicking mutants indicates that phosphorylation of R120GαB-crystallin increases its propensity to aggregate. GENERAL SIGNIFICANCE Phosphorylation of αB-crystallin has dual role that manifests either beneficial or deleterious consequences depending on the extent of phosphorylation and interaction with cytoskeleton. Considering that disease-causing mutants of αB-crystallin are hyperphosphorylated, moderation of phosphorylation may be a useful strategy in disease management. This article is part of a Special Issue entitled Crystallin Biochemistry in Health and Disease.
Collapse
Affiliation(s)
- Raman Bakthisaran
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | - Kranthi Kiran Akula
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | - Ramakrishna Tangirala
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | - Ch Mohan Rao
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India.
| |
Collapse
|
45
|
Mueller NH, Fogueri U, Pedler MG, Montana K, Petrash JM, Ammar DA. Impact of Subunit Composition on the Uptake of α-Crystallin by Lens and Retina. PLoS One 2015; 10:e0137659. [PMID: 26355842 PMCID: PMC4565700 DOI: 10.1371/journal.pone.0137659] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 08/19/2015] [Indexed: 11/30/2022] Open
Abstract
Misfolded protein aggregation, including cataract, cause a significant amount of blindness worldwide. α-Crystallin is reported to bind misfolded proteins and prevent their aggregation. We hypothesize that supplementing retina and lens with α-crystallin may help to delay disease onset. The purpose of this study was to determine if αB-crystallin subunits containing a cell penetration peptide (gC-tagged αB-crystallin) facilitate the uptake of wild type αA-crystallin (WT-αA) in lens and retina. Recombinant human αB-crystallin was modified by the addition of a novel cell penetration peptide derived from the gC gene product of herpes simplex virus (gC-αB). Recombinant gC-αB and wild-type αA-crystallin (WT-αA) were purified from E. coli over-expression cultures. After Alexa-labeling of WT-αA, these proteins were mixed at ratios of 1:2, 1:5 and 1:10, respectively, and incubated at 37°C for 4 hours to allow for subunit exchange. Mixed oligomers were subsequently incubated with tissue culture cells or mouse organ cultures. Similarly, crystallin mixtures were injected into the vitreous of rat eyes. At various times after exposure, tissues were harvested and analyzed for protein uptake by confocal microscopy or flow cytometry. Chaperone-like activity assays were performed on α-crystallins ratios showing optimal uptake using chemically-induced or heat induced substrate aggregation assays. As determined by flow cytometry, a ratio of 1:5 for gC-αB to WT-αA was found to be optimal for uptake into retinal pigmented epithelial cells (ARPE-19). Chaperone-like activity assays demonstrated that hetero-oligomeric complex of gC-αB to WT-αA (in 1:5 ratio) retained protein aggregation protection. We observed a significant increase in protein uptake when optimized (gC-αB to WT-αA (1:5 ratio)) hetero-oligomers were used in mouse lens and retinal organ cultures. Increased levels of α-crystallin were found in lens and retina following intravitreal injection of homo- and hetero-oligomers in rats.
Collapse
Affiliation(s)
- Niklaus H. Mueller
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
- * E-mail:
| | - Uma Fogueri
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy, University of Colorado, Aurora, Colorado, United States of America
| | - Michelle G. Pedler
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Kameron Montana
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - J. Mark Petrash
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy, University of Colorado, Aurora, Colorado, United States of America
| | - David A. Ammar
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| |
Collapse
|
46
|
Anbarasu K, Sivakumar J. Multidimensional significance of crystallin protein-protein interactions and their implications in various human diseases. Biochim Biophys Acta Gen Subj 2015; 1860:222-33. [PMID: 26365509 DOI: 10.1016/j.bbagen.2015.09.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 08/28/2015] [Accepted: 09/08/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND Crystallins are the important structural and functional proteins in the eye lens responsible for refractive index. Post-translational modifications (PTMs) and mutations are major causative factors that affect crystallin structural conformation and functional characteristics thus playing a vital role in the etiology of cataractogenesis. SCOPE OF REVIEW The significance of crystallin protein-protein interactions (PPIs) in the lens and non-lenticular tissues is summarized. MAJOR CONCLUSIONS Aberrancy of PPIs between crystallin, its associated protein and metal ions has been accomplished in various human diseases including cataract. A detailed account on multidimensional structural and functional significance of crystallin PPI in humans must be brought into limelight, in order to understand the biochemical and molecular basis augmenting the aberrancies of such interaction. In this scenario, the present review is focused to shed light on studies which will aid to expand our present understanding on disease pathogenesis related to loss of PPI thereby paving the way for putative future therapeutic targets to curb such diseases. GENERAL SIGNIFICANCE The interactions with α-crystallins always aid to protect their structural and functional characteristics. The up-regulation of αB-crystallin in the non-lenticular tissues always decodes as biomarker for various stress related disorders. For better understanding and treatment of various diseases, PPI studies provide overall outline about the structural and functional characteristics of the proteins. This information not only helps to find out the route of cataractogenesis but also aid to identify potential molecules to inhibit/prevent the further development of such complicated phenomenon. This article is part of a Special Issue entitled Crystallin Biochemistry in Health and Disease.
Collapse
Affiliation(s)
- Kumarasamy Anbarasu
- Department of Marine Biotechnology, Bharathidasan University, Tiruchirapalli 620024, Tamil Nadu, India.
| | - Jeyarajan Sivakumar
- Department of Marine Biotechnology, Bharathidasan University, Tiruchirapalli 620024, Tamil Nadu, India
| |
Collapse
|
47
|
Clark JI. Functional sequences in human alphaB crystallin. Biochim Biophys Acta Gen Subj 2015; 1860:240-5. [PMID: 26341790 DOI: 10.1016/j.bbagen.2015.08.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 07/20/2015] [Accepted: 08/24/2015] [Indexed: 12/17/2022]
Abstract
BACKGROUND Human alphaB crystallin (HspB5) contains the alpha crystallin core domain, a series of antiparallel beta-strands organized into the characteristic beta sandwich of small heat shock proteins (sHsps). The full 3-dimensional structure for alpha crystallin has not been determined and the mechanism for the biological activity remains elusive because sHsps participate in multiple interactions with a broad range of target proteins that favor self-assembly of polydisperse fibrils and complexes. We selected human alphaB crystallin to study interactive sequences because it is involved in many human condensation, amyloid, and aggregation diseases and it is very sensitive to the destabilization of unfolding proteins. Sophisticated methods are being used to analyze and complete the structure of alphaB crystallin with the expectation of understanding sHsp function. This review considers the identification of interactive sites on the surface of the alphaB crystallin, which may be the key to understanding the multifunctional activity of human alphaB crystallin. SCOPE OF REVIEW This review summarizes the research on the identification of the bioactive interactive sequences responsible for the function of human alphaB crystallin, an sHsp with chaperone-like activity. MAJOR CONCLUSIONS The multifunctional activity of human alphaB crystallin results from the interactive peptide sequences exposed on the surface of the molecule. The multiple, non-covalent, interactive sequences can account for the selectivity and sensitivity of alphaB crystallin to the initiation of protein unfolding. GENERAL SIGNIFICANCE Human alphaB crystallin may be an important part of an endogenous protective mechanism in aging cells and tissues. This article is part of a Special Issue entitled Crystallin Biochemistry in Health and Disease.
Collapse
Affiliation(s)
- John I Clark
- Departments of Biological Structure and Ophthalmology, University of Washington, Seattle, WA 98195-7420, USA.
| |
Collapse
|
48
|
Su SP, Song X, Xavier D, Aquilina JA. Age-related cleavages of crystallins in human lens cortical fiber cells generate a plethora of endogenous peptides and high molecular weight complexes. Proteins 2015; 83:1878-86. [PMID: 26238763 DOI: 10.1002/prot.24872] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 07/16/2015] [Accepted: 07/29/2015] [Indexed: 11/07/2022]
Abstract
Low molecular weight peptides derived from the breakdown of crystallins have been reported in adult human lenses. The proliferation of these LMW peptides coincides with the earliest stages of cataract formation, suggesting that the protein cleavages involved may contribute to the aggregation and insolubilization of crystallins. This study reports the identification of 238 endogenous LMW crystallin peptides from the cortical extracts of four human lenses representing young, middle and old-age human lenses. Analysis of the peptide terminal amino acids showed that Lys and Arg were situated at the C-terminus with significantly higher frequency compared to other residues, suggesting that trypsin-like proteolysis may be active in the lens cortical fiber cells. Selected reaction monitoring analysis of an endogenous αA-crystallin peptide (αA(57-65)) showed that the concentration of this peptide in the human lens increased gradually to middle age, after which the rate of αA(57-65) formation escalated significantly. Using 2D gel electrophoresis/nanoLC-ESI-MS/MS, 12 protein complexes of 40-150 kDa consisting of multiple crystallin components were characterized from the water soluble cortical extracts of an adult human lens. The detection of these protein complexes suggested the possibility of crystallin cross-linking, with these complexes potentially acting to stabilize degraded crystallins by sequestration into water soluble complexes.
Collapse
Affiliation(s)
- Shih-Ping Su
- Illawarra Health and Medical Research Institute, School of Biological Sciences, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, New South Wales, 2522, Australia
| | - Xiaomin Song
- Australian Proteome Analysis Facility, Macquarie University, Sydney, New South Wales, 2109, Australia
| | - Dylan Xavier
- Australian Proteome Analysis Facility, Macquarie University, Sydney, New South Wales, 2109, Australia
| | - J Andrew Aquilina
- Illawarra Health and Medical Research Institute, School of Biological Sciences, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, New South Wales, 2522, Australia
| |
Collapse
|
49
|
Raju M, Santhoshkumar P, Krishna Sharma K. Alpha-crystallin-derived peptides as therapeutic chaperones. Biochim Biophys Acta Gen Subj 2015; 1860:246-51. [PMID: 26141743 DOI: 10.1016/j.bbagen.2015.06.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 06/19/2015] [Accepted: 06/26/2015] [Indexed: 11/16/2022]
Abstract
BACKGROUND The demonstration of chaperone-like activity in peptides (mini-chaperones) derived from α-crystallin's chaperone region has generated significant interest in exploring the therapeutic potential of peptide chaperones in diseases of protein aggregation. Recent studies in experimental animals show that mini-chaperones could reach intended targets and alter the disease phenotype. Although mini-chaperones show potential benefits against protein aggregation diseases, they do tend to form aggregates on storage. There is thus a need to fine-tune peptide chaperones to increase their solubility, pharmacokinetics, and biological efficacy. SCOPE OF REVIEW This review summarizes the properties and the potential therapeutic roles of mini-chaperones in protein aggregation diseases and highlights some of the refinements needed to increase the stability and biological efficacy of mini-chaperones while maintaining or enhancing their chaperone-like activity against precipitation of unfolding proteins. MAJOR CONCLUSIONS Mini-chaperones suppress the aggregation of proteins, block amyloid fibril formation, stabilize mutant proteins, sequester metal ions, and exhibit antiapoptotic properties. Much work must be done to fine-tune mini-chaperones and increase their stability and biological efficacy. Peptide chaperones could have a great therapeutic value in diseases associated with protein aggregation and apoptosis. GENERAL SIGNIFICANCE Accumulation of misfolded proteins is a primary cause for many age-related diseases, including cataract, macular degeneration, and various neurological diseases. Stabilization of native proteins is a logical therapeutic approach for such diseases. Mini-chaperones, with their inherent antiaggregation and antiapoptotic properties, may represent an effective therapeutic molecule to prevent the cascade of protein conformational disorders. Future studies will further uncover the therapeutic potential of mini-chaperones. This article is part of a Special Issue entitled Crystallin Biochemistry in Health and Disease.
Collapse
Affiliation(s)
- Murugesan Raju
- Department of Ophthalmology, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Puttur Santhoshkumar
- Department of Ophthalmology, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - K Krishna Sharma
- Department of Ophthalmology, University of Missouri School of Medicine, Columbia, MO 65212, USA; Department of Biochemistry, University of Missouri School of Medicine, Columbia, MO 65212, USA.
| |
Collapse
|
50
|
Haslbeck M, Peschek J, Buchner J, Weinkauf S. Structure and function of α-crystallins: Traversing from in vitro to in vivo. Biochim Biophys Acta Gen Subj 2015; 1860:149-66. [PMID: 26116912 DOI: 10.1016/j.bbagen.2015.06.008] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Revised: 06/10/2015] [Accepted: 06/22/2015] [Indexed: 12/11/2022]
Abstract
BACKGROUND The two α-crystallins (αA- and αB-crystallin) are major components of our eye lenses. Their key function there is to preserve lens transparency which is a challenging task as the protein turnover in the lens is low necessitating the stability and longevity of the constituent proteins. α-Crystallins are members of the small heat shock protein family. αB-crystallin is also expressed in other cell types. SCOPE OF THE REVIEW The review summarizes the current concepts on the polydisperse structure of the α-crystallin oligomer and its chaperone function with a focus on the inherent complexity and highlighting gaps between in vitro and in vivo studies. MAJOR CONCLUSIONS Both α-crystallins protect proteins from irreversible aggregation in a promiscuous manner. In maintaining eye lens transparency, they reduce the formation of light scattering particles and balance the interactions between lens crystallins. Important for these functions is their structural dynamics and heterogeneity as well as the regulation of these processes which we are beginning to understand. However, currently, it still remains elusive to which extent the in vitro observed properties of α-crystallins reflect the highly crowded situation in the lens. GENERAL SIGNIFICANCE Since α-crystallins play an important role in preventing cataract in the eye lens and in the development of diverse diseases, understanding their mechanism and substrate spectra is of importance. To bridge the gap between the concepts established in vitro and the in vivo function of α-crystallins, the joining of forces between different scientific disciplines and the combination of diverse techniques in hybrid approaches are necessary. This article is part of a Special Issue entitled Crystallin Biochemistry in Health and Disease.
Collapse
Affiliation(s)
- Martin Haslbeck
- Center for Integrated Protein Science at the Department Chemie, Technische Universität München, Lichtenbergstr. 4, D-85747 Garching, Germany
| | - Jirka Peschek
- Center for Integrated Protein Science at the Department Chemie, Technische Universität München, Lichtenbergstr. 4, D-85747 Garching, Germany
| | - Johannes Buchner
- Center for Integrated Protein Science at the Department Chemie, Technische Universität München, Lichtenbergstr. 4, D-85747 Garching, Germany.
| | - Sevil Weinkauf
- Center for Integrated Protein Science at the Department Chemie, Technische Universität München, Lichtenbergstr. 4, D-85747 Garching, Germany.
| |
Collapse
|