1
|
Nicy, Morgan JWR, Wales DJ. Energy landscapes for clusters of hexapeptides. J Chem Phys 2024; 161:054112. [PMID: 39092941 DOI: 10.1063/5.0220652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 07/17/2024] [Indexed: 08/04/2024] Open
Abstract
We present the results for energy landscapes of hexapeptides obtained using interfaces to the Large-scale Atomic/Molecular Massively Parallel Simulator (LAMMPS) program. We have used basin-hopping global optimization and discrete path sampling to explore the landscapes of hexapeptide monomers, dimers, and oligomers containing 10, 100, and 200 monomers modeled using a residue-level coarse-grained potential, Mpipi, implemented in LAMMPS. We find that the dimers of peptides containing amino acid residues that are better at promoting phase separation, such as tyrosine and arginine, have melting peaks at higher temperature in their heat capacity compared to phenylalanine and lysine, respectively. This observation correlates with previous work on the same uncapped hexapeptide monomers modeled using atomistic potential. For oligomers, we compare the variation in monomer conformations with radial distance and observe trends for selected angles calculated for each monomer. The LAMMPS interfaces to the GMIN and OPTIM programs for landscape exploration offer new opportunities to investigate larger systems and provide access to the coarse-grained potentials implemented within LAMMPS.
Collapse
Affiliation(s)
- Nicy
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - John W R Morgan
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - David J Wales
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| |
Collapse
|
2
|
Lemche E, Killick R, Mitchell J, Caton PW, Choudhary P, Howard JK. Molecular mechanisms linking type 2 diabetes mellitus and late-onset Alzheimer's disease: A systematic review and qualitative meta-analysis. Neurobiol Dis 2024; 196:106485. [PMID: 38643861 DOI: 10.1016/j.nbd.2024.106485] [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: 06/30/2023] [Revised: 03/18/2024] [Accepted: 03/23/2024] [Indexed: 04/23/2024] Open
Abstract
Research evidence indicating common metabolic mechanisms through which type 2 diabetes mellitus (T2DM) increases risk of late-onset Alzheimer's dementia (LOAD) has accumulated over recent decades. The aim of this systematic review is to provide a comprehensive review of common mechanisms, which have hitherto been discussed in separate perspectives, and to assemble and evaluate candidate loci and epigenetic modifications contributing to polygenic risk linkages between T2DM and LOAD. For the systematic review on pathophysiological mechanisms, both human and animal studies up to December 2023 are included. For the qualitative meta-analysis of genomic bases, human association studies were examined; for epigenetic mechanisms, data from human studies and animal models were accepted. Papers describing pathophysiological studies were identified in databases, and further literature gathered from cited work. For genomic and epigenomic studies, literature mining was conducted by formalised search codes using Boolean operators in search engines, and augmented by GeneRif citations in Entrez Gene, and other sources (WikiGenes, etc.). For the systematic review of pathophysiological mechanisms, 923 publications were evaluated, and 138 gene loci extracted for testing candidate risk linkages. 3 57 publications were evaluated for genomic association and descriptions of epigenomic modifications. Overall accumulated results highlight insulin signalling, inflammation and inflammasome pathways, proteolysis, gluconeogenesis and glycolysis, glycosylation, lipoprotein metabolism and oxidation, cell cycle regulation or survival, autophagic-lysosomal pathways, and energy. Documented findings suggest interplay between brain insulin resistance, neuroinflammation, insult compensatory mechanisms, and peripheral metabolic dysregulation in T2DM and LOAD linkage. The results allow for more streamlined longitudinal studies of T2DM-LOAD risk linkages.
Collapse
Affiliation(s)
- Erwin Lemche
- Section of Cognitive Neuropsychiatry and Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, United Kingdom.
| | - Richard Killick
- Section of Old Age Psychiatry, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, United Kingdom
| | - Jackie Mitchell
- Department of Basic and Clinical Neurosciences, Maurice Wohl CIinical Neurosciences Institute, Institute of Psychiatry, Psychology & Neuroscience, King's College London, 125 Coldharbour Lane, London SE5 9NU, United Kingdom
| | - Paul W Caton
- Diabetes Research Group, School of Life Course Sciences, King's College London, Hodgkin Building, Guy's Campus, London SE1 1UL, United Kingdom
| | - Pratik Choudhary
- Diabetes Research Group, Weston Education Centre, King's College London, 10 Cutcombe Road, London SE5 9RJ, United Kingdom
| | - Jane K Howard
- School of Cardiovascular and Metabolic Medicine & Sciences, Hodgkin Building, Guy's Campus, King's College London, Great Maze Pond, London SE1 1UL, United Kingdom
| |
Collapse
|
3
|
Kaygisiz K, Rauch-Wirth L, Dutta A, Yu X, Nagata Y, Bereau T, Münch J, Synatschke CV, Weil T. Data-mining unveils structure-property-activity correlation of viral infectivity enhancing self-assembling peptides. Nat Commun 2023; 14:5121. [PMID: 37612273 PMCID: PMC10447463 DOI: 10.1038/s41467-023-40663-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 08/01/2023] [Indexed: 08/25/2023] Open
Abstract
Gene therapy via retroviral vectors holds great promise for treating a variety of serious diseases. It requires the use of additives to boost infectivity. Amyloid-like peptide nanofibers (PNFs) were shown to efficiently enhance retroviral gene transfer. However, the underlying mode of action of these peptides remains largely unknown. Data-mining is an efficient method to systematically study structure-function relationship and unveil patterns in a database. This data-mining study elucidates the multi-scale structure-property-activity relationship of transduction enhancing peptides for retroviral gene transfer. In contrast to previous reports, we find that not the amyloid fibrils themselves, but rather µm-sized β-sheet rich aggregates enhance infectivity. Specifically, microscopic aggregation of β-sheet rich amyloid structures with a hydrophobic surface pattern and positive surface charge are identified as key material properties. We validate the reliability of the amphiphilic sequence pattern and the general applicability of the key properties by rationally creating new active sequences and identifying short amyloidal peptides from various pathogenic and functional origin. Data-mining-even for small datasets-enables the development of new efficient retroviral transduction enhancers and provides important insights into the diverse bioactivity of the functional material class of amyloids.
Collapse
Affiliation(s)
- Kübra Kaygisiz
- Department Synthesis of Macromolecules, Max Planck Institute for Polymer Research, Ackermannweg 10, 55128, Mainz, Germany
| | - Lena Rauch-Wirth
- Institute of Molecular Virology, Ulm University Medical Center, Meyerhofstraße 1, 89081, Ulm, Germany
| | - Arghya Dutta
- Department Polymer Theory, Max Planck Institute for Polymer Research, Ackermannweg 10, 55128, Mainz, Germany
- Institute of Biochemistry II, Faculty of Medicine, Goethe University, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Xiaoqing Yu
- Department Molecular Spectroscopy, Max Planck Institute for Polymer Research, Ackermannweg 10, 55128, Mainz, Germany
| | - Yuki Nagata
- Department Molecular Spectroscopy, Max Planck Institute for Polymer Research, Ackermannweg 10, 55128, Mainz, Germany
| | - Tristan Bereau
- Department Polymer Theory, Max Planck Institute for Polymer Research, Ackermannweg 10, 55128, Mainz, Germany
- Institute for Theoretical Physics, Heidelberg University, Philosophenweg 19, 69120, Heidelberg, Germany
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, Meyerhofstraße 1, 89081, Ulm, Germany
| | - Christopher V Synatschke
- Department Synthesis of Macromolecules, Max Planck Institute for Polymer Research, Ackermannweg 10, 55128, Mainz, Germany.
| | - Tanja Weil
- Department Synthesis of Macromolecules, Max Planck Institute for Polymer Research, Ackermannweg 10, 55128, Mainz, Germany.
| |
Collapse
|
4
|
Wales DJ. Energy Landscapes and Heat Capacity Signatures for Monomers and Dimers of Amyloid-Forming Hexapeptides. Int J Mol Sci 2023; 24:10613. [PMID: 37445791 DOI: 10.3390/ijms241310613] [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: 05/18/2023] [Revised: 06/16/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
Amyloid formation is a hallmark of various neurodegenerative disorders. In this contribution, energy landscapes are explored for various hexapeptides that are known to form amyloids. Heat capacity (CV) analysis at low temperature for these hexapeptides reveals that the low energy structures contributing to the first heat capacity feature above a threshold temperature exhibit a variety of backbone conformations for amyloid-forming monomers. The corresponding control sequences do not exhibit such structural polymorphism, as diagnosed via end-to-end distance and a dihedral angle defined for the monomer. A similar heat capacity analysis for dimer conformations obtained using basin-hopping global optimisation shows clear features in end-to-end distance versus dihedral correlation plots, where amyloid-forming sequences exhibit a preference for larger end-to-end distances and larger positive dihedrals. These results hold true for sequences taken from tau, amylin, insulin A chain, a de novo designed peptide, and various control sequences. While there is a little overall correlation between the aggregation propensity and the temperature at which the low-temperature CV feature occurs, further analysis suggests that the amyloid-forming sequences exhibit the key CV feature at a lower temperature compared to control sequences derived from the same protein.
Collapse
Affiliation(s)
- David J Wales
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
| |
Collapse
|
5
|
Papiri G, D’Andreamatteo G, Cacchiò G, Alia S, Silvestrini M, Paci C, Luzzi S, Vignini A. Multiple Sclerosis: Inflammatory and Neuroglial Aspects. Curr Issues Mol Biol 2023; 45:1443-1470. [PMID: 36826039 PMCID: PMC9954863 DOI: 10.3390/cimb45020094] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/28/2023] [Accepted: 02/02/2023] [Indexed: 02/11/2023] Open
Abstract
Multiple sclerosis (MS) represents the most common acquired demyelinating disorder of the central nervous system (CNS). Its pathogenesis, in parallel with the well-established role of mechanisms pertaining to autoimmunity, involves several key functions of immune, glial and nerve cells. The disease's natural history is complex, heterogeneous and may evolve over a relapsing-remitting (RRMS) or progressive (PPMS/SPMS) course. Acute inflammation, driven by infiltration of peripheral cells in the CNS, is thought to be the most relevant process during the earliest phases and in RRMS, while disruption in glial and neural cells of pathways pertaining to energy metabolism, survival cascades, synaptic and ionic homeostasis are thought to be mostly relevant in long-standing disease, such as in progressive forms. In this complex scenario, many mechanisms originally thought to be distinctive of neurodegenerative disorders are being increasingly recognized as crucial from the beginning of the disease. The present review aims at highlighting mechanisms in common between MS, autoimmune diseases and biology of neurodegenerative disorders. In fact, there is an unmet need to explore new targets that might be involved as master regulators of autoimmunity, inflammation and survival of nerve cells.
Collapse
Affiliation(s)
- Giulio Papiri
- Neurology Unit, Ospedale Provinciale “Madonna del Soccorso”, 63074 San Benedetto del Tronto, Italy
| | - Giordano D’Andreamatteo
- Neurology Unit, Ospedale Provinciale “Madonna del Soccorso”, 63074 San Benedetto del Tronto, Italy
| | - Gabriella Cacchiò
- Neurology Unit, Ospedale Provinciale “Madonna del Soccorso”, 63074 San Benedetto del Tronto, Italy
| | - Sonila Alia
- Section of Biochemistry, Biology and Physics, Department of Clinical Sciences, Università Politecnica delle Marche, 60100 Ancona, Italy
| | - Mauro Silvestrini
- Neurology Unit, Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60100 Ancona, Italy
| | - Cristina Paci
- Neurology Unit, Ospedale Provinciale “Madonna del Soccorso”, 63074 San Benedetto del Tronto, Italy
| | - Simona Luzzi
- Neurology Unit, Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60100 Ancona, Italy
| | - Arianna Vignini
- Section of Biochemistry, Biology and Physics, Department of Clinical Sciences, Università Politecnica delle Marche, 60100 Ancona, Italy
- Correspondence:
| |
Collapse
|
6
|
Li YL, Xie JY, Lu B, Sun XD, Chen FF, Tong ZJ, Sai WW, Zhang W, Wang ZH, Zhong M. β-sheets in serum protein are independent risk factors for coronary lesions besides LDL-C in coronary heart disease patients. Front Cardiovasc Med 2022; 9:911358. [PMID: 36017095 PMCID: PMC9395970 DOI: 10.3389/fcvm.2022.911358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
Background Coronary heart disease (CHD) patients with standard low-density lipoprotein cholesterol (LDL-C) remain at risk of cardiovascular events, making it critical to explore new targets to reduce the residual risk. The relationship between β-sheet conformation and CHD is gaining attention. This study was designed to compare the coronary lesions in CHD patients with varying LDL-C and evaluate whether serum β-sheets are associated with coronary damage. Methods Two hundred and one patients diagnosed with stable CHD were recruited and divided into four groups according to LDL-C. Baseline information, coronary lesion-related indicators, and peripheral blood samples were collected. Serum β-sheet content was determined by thioflavin T fluorescence. Results The baseline information was comparable in CHD patients with different LDL-C. No difference was found in indicators relevant to coronary lesions among groups. The content of β-sheet was negatively correlated with LDL-C. Multiple linear regression revealed that serum β-sheet was positively correlated with coronary lesion when risk factors such as age, smoking, and LDL-C were controlled. Conclusions This is the first study that reports the serum β-sheet levels of CHD patients being gradually increased with decreasing LDL-C when coronary lesions were comparable. Serum β-sheet might exacerbate the coronary lesions in CHD patients independent of known risk factors such as LDL-C.
Collapse
Affiliation(s)
- Yu-lin Li
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jia-ying Xie
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Bin Lu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiao-di Sun
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Geriatric Medicine, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Fang-fang Chen
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zhou-jie Tong
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wen-wen Sai
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wei Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zhi-hao Wang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Geriatric Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Shandong Key Laboratory of Cardiovascular Proteomics, Jinan, China
| | - Ming Zhong
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
7
|
Abstract
Amyloids are organized suprastructural polypeptide arrangements. The prevalence of amyloid-related processes of pathophysiological relevance has been linked to aging-related degenerative diseases. Besides the role of genetic polymorphisms on the relative risk of amyloid diseases, the contributions of nongenetic ontogenic cluster of factors remain elusive. In recent decades, mounting evidences have been suggesting the role of essential micronutrients, in particular transition metals, in the regulation of amyloidogenic processes, both directly (such as binding to amyloid proteins) or indirectly (such as regulating regulatory partners, processing enzymes, and membrane transporters). The features of transition metals as regulatory cofactors of amyloid proteins and the consequences of metal dyshomeostasis in triggering amyloidogenic processes, as well as the evidences showing amelioration of symptoms by dietary supplementation, suggest an exaptative role of metals in regulating amyloid pathways. The self- and cross-talk replicative nature of these amyloid processes along with their systemic distribution support the concept of their metastatic nature. The role of amyloidosis as nutrient sensors would act as intra- and transgenerational epigenetic metabolic programming factors determining health span and life span, viability, which could participate as an evolutive selective pressure.
Collapse
Affiliation(s)
- Luís Maurício T R Lima
- Laboratory for Pharmaceutical Biotechnology - pbiotech, Faculty of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
- Laboratory for Macromolecules (LAMAC-DIMAV), National Institute of Metrology, Quality and Technology - INMETRO, Duque de Caxias, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Tháyna Sisnande
- Laboratory for Pharmaceutical Biotechnology - pbiotech, Faculty of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
8
|
Nian Y, Zhang Y, Ruan C, Hu B. Update of the interaction between polyphenols and amyloid fibrils. Curr Opin Food Sci 2022. [DOI: 10.1016/j.cofs.2021.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
9
|
Hemed-Shaked M, Cowman MK, Kim JR, Huang X, Chau E, Ovadia H, Amar KO, Eshkar-Sebban L, Melamed M, Lev LB, Kedar E, Armengol J, Alemany J, Beyth S, Okon E, Kanduc D, Elgavish S, Wallach-Dayan SB, Cohen SJ, Naor D. MTADV 5-MER peptide suppresses chronic inflammations as well as autoimmune pathologies and unveils a new potential target-Serum Amyloid A. J Autoimmun 2021; 124:102713. [PMID: 34390919 DOI: 10.1016/j.jaut.2021.102713] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/21/2021] [Accepted: 07/24/2021] [Indexed: 11/17/2022]
Abstract
Despite the existence of potent anti-inflammatory biological drugs e.g., anti-TNF and anti IL-6 receptor antibodies, for treating chronic inflammatory and autoimmune diseases, these are costly and not specific. Cheaper oral available drugs remain an unmet need. Expression of the acute phase protein Serum Amyloid A (SAA) is dependent on release of pro-inflammatory cytokines IL-1, IL-6 and TNF-α during inflammation. Conversely, SAA induces pro-inflammatory cytokine secretion, including Th17, leading to a pathogenic vicious cycle and chronic inflammation. 5- MER peptide (5-MP) MTADV (methionine-threonine-alanine-aspartic acid-valine), also called Amilo-5MER, was originally derived from a sequence of a pro-inflammatory CD44 variant isolated from synovial fluid of a Rheumatoid Arthritis (RA) patient. This human peptide displays an efficient anti-inflammatory effects to ameliorate pathology and clinical symptoms in mouse models of RA, Inflammatory Bowel Disease (IBD) and Multiple Sclerosis (MS). Bioinformatics and qRT-PCR revealed that 5-MP, administrated to encephalomyelytic mice, up-regulates genes contributing to chronic inflammation resistance. Mass spectrometry of proteins that were pulled down from an RA synovial cell extract with biotinylated 5-MP, showed that it binds SAA. 5-MP disrupted SAA assembly, which is correlated with its pro-inflammatory activity. The peptide MTADV (but not scrambled TMVAD) significantly inhibited the release of pro-inflammatory cytokines IL-6 and IL-1β from SAA-activated human fibroblasts, THP-1 monocytes and peripheral blood mononuclear cells. 5-MP suppresses the pro-inflammatory IL-6 release from SAA-activated cells, but not from non-activated cells. 5-MP could not display therapeutic activity in rats, which are SAA deficient, but does inhibit inflammations in animal models of IBD and MS, both are SAA-dependent, as shown by others in SAA knockout mice. In conclusion, 5-MP suppresses chronic inflammation in animal models of RA, IBD and MS, which are SAA-dependent, but not in animal models, which are SAA-independent.
Collapse
Affiliation(s)
- Maayan Hemed-Shaked
- The Lautenberg Center of Immunology and Cancer Research, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Mary K Cowman
- Department of Biomedical Engineering, Tandon School of Engineering, New York University, New York, NY, USA
| | - Jin Ryoun Kim
- Othmer-Jacobs Department of Chemical and Biomolecular Engineering, Tandon School of Engineering, Brooklyn, USA
| | - Xiayun Huang
- Othmer-Jacobs Department of Chemical and Biomolecular Engineering, Tandon School of Engineering, Brooklyn, USA
| | - Edward Chau
- Othmer-Jacobs Department of Chemical and Biomolecular Engineering, Tandon School of Engineering, Brooklyn, USA
| | - Haim Ovadia
- Department of Neurology, Agnes Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Keren-Or Amar
- The Lautenberg Center of Immunology and Cancer Research, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Lora Eshkar-Sebban
- The Lautenberg Center of Immunology and Cancer Research, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Michal Melamed
- The Lautenberg Center of Immunology and Cancer Research, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Libat Bar Lev
- The Lautenberg Center of Immunology and Cancer Research, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Eli Kedar
- The Lautenberg Center of Immunology and Cancer Research, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | | | | | - Shaul Beyth
- Orthopedic Surgery Department, Hadassah University Hospital, Jerusalem, Israel
| | - Eli Okon
- Department of Pathology, Hadassah University Hospital, Jerusalem, Israel
| | - Darja Kanduc
- Biotechnologies and Biopharmaceutics, University of Bari, Bari, 70126, Italy
| | - Sharona Elgavish
- Bioinformatics Unit of the Hebrew University of Jerusalem and Hadassah Medical Center, Israel
| | - Shulamit B Wallach-Dayan
- Lung Cellular and Molecular Biology Laboratory, Institute of Pulmonary Medicine, Hadassah-Hebrew University Medical Center, Israel
| | - Shmuel Jaffe Cohen
- The Lautenberg Center of Immunology and Cancer Research, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - David Naor
- The Lautenberg Center of Immunology and Cancer Research, Faculty of Medicine, Hebrew University of Jerusalem, Israel.
| |
Collapse
|
10
|
Antipsychotics, versatility in action. Proc Natl Acad Sci U S A 2021; 118:2108946118. [PMID: 34244436 DOI: 10.1073/pnas.2108946118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
11
|
Lo CH, Sachs JN. The role of wild-type tau in Alzheimer's disease and related tauopathies. JOURNAL OF LIFE SCIENCES (WESTLAKE VILLAGE, CALIF.) 2020; 2:1-17. [PMID: 33665646 PMCID: PMC7929479 DOI: 10.36069/jols/20201201] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Tau oligomers have recently emerged as the principal toxic species in Alzheimer's disease (AD) and tauopathies. Tau oligomers are spontaneously self-assembled soluble tau proteins that are formed prior to fibrils, and they have been shown to play a central role in neuronal cell death and in the induction of neurodegeneration in animal models. As the therapeutic paradigm shifts to targeting toxic tau oligomers, this suggests the focus to study tau oligomerization in species that are less susceptible to fibrillization. While truncated and mutation containing tau as well as the isolated repeat domains are particularly prone to fibrillization, the wild-type (WT) tau proteins have been shown to be resistant to fibril formation in the absence of aggregation inducers. In this review, we will summarize and discuss the toxicity of WT tau both in vitro and in vivo, as well as its involvement in tau oligomerization and cell-to-cell propagation of pathology. Understanding the role of WT tau will enable more effective biomarker development and therapeutic discovery for treatment of AD and tauopathies.
Collapse
Affiliation(s)
- Chih Hung Lo
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
| | - Jonathan N. Sachs
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455
| |
Collapse
|
12
|
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
|
13
|
Huang YM, Hong XZ, Shen J, Geng LJ, Pan YH, Ling W, Zhao HL. Amyloids in Site-Specific Autoimmune Reactions and Inflammatory Responses. Front Immunol 2020; 10:2980. [PMID: 31993048 PMCID: PMC6964640 DOI: 10.3389/fimmu.2019.02980] [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: 05/07/2019] [Accepted: 12/04/2019] [Indexed: 12/15/2022] Open
Abstract
Amyloid deposition is a histological hallmark of common human disorders including Alzheimer's disease (AD) and type 2 diabetes. Although some reports highlight that amyloid fibrils might activate the innate immunity system via pattern recognition receptors, here, we provide multiple lines of evidence for the protection by site-specific amyloid protein analogs and fibrils against autoimmune attacks: (1) strategies targeting clearance of the AD-related brain amyloid plaque induce high risk of deadly autoimmune destructions in subjects with cognitive dysfunction; (2) administration of amyloidogenic peptides with either full length or core hexapeptide structure consistently ameliorates signs of experimental autoimmune encephalomyelitis; (3) experimental autoimmune encephalomyelitis is exacerbated following genetic deletion of amyloid precursor proteins; (4) absence of islet amyloid coexists with T-cell-mediated insulitis in autoimmune diabetes and autoimmune polyendocrine syndrome; (5) use of islet amyloid polypeptide agonists rather than antagonists improves diabetes care; and (6) common suppressive signaling pathways by regulatory T cells are activated in both local and systemic amyloidosis. These findings indicate dual modulation activity mediated by amyloid protein monomers, oligomers, and fibrils to maintain immune homeostasis. The protection from autoimmune destruction by amyloid proteins offers a novel therapeutic approach to regenerative medicine for common degenerative diseases.
Collapse
Affiliation(s)
- Yan-Mei Huang
- Department of Immunology, Guangxi Area of Excellence, Guilin Medical University, Guilin, China.,Center for Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin, China
| | - Xue-Zhi Hong
- Department of Immunology, Guangxi Area of Excellence, Guilin Medical University, Guilin, China.,Department of Rheumatology and Immunology, The First Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Jian Shen
- Department of Immunology, Guangxi Area of Excellence, Guilin Medical University, Guilin, China.,Department of Pathology, The First Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Li-Jun Geng
- Department of Immunology, Guangxi Area of Excellence, Guilin Medical University, Guilin, China.,Center for Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin, China
| | - Yan-Hong Pan
- Department of Immunology, Guangxi Area of Excellence, Guilin Medical University, Guilin, China.,Center for Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin, China
| | - Wei Ling
- Department of Immunology, Guangxi Area of Excellence, Guilin Medical University, Guilin, China.,Department of Endocrinology, Xiangya Medical School, Central South University, Changsha, China
| | - Hai-Lu Zhao
- Department of Immunology, Guangxi Area of Excellence, Guilin Medical University, Guilin, China.,Center for Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin, China.,Institute of Basic Medical Sciences, Faculty of Basic Medicine, Guilin Medical University, Guilin, China
| |
Collapse
|
14
|
Trudler D, Levy‐Barazany H, Nash Y, Samuel L, Sharon R, Frenkel D. Alpha synuclein deficiency increases CD4
+
T‐cells pro‐inflammatory profile in a Nurr1‐dependent manner. J Neurochem 2019; 152:61-71. [DOI: 10.1111/jnc.14871] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 09/05/2019] [Accepted: 09/10/2019] [Indexed: 01/25/2023]
Affiliation(s)
- Dorit Trudler
- Department of Neurobiology George S. Wise Faculty of Life Sciences Tel Aviv University Tel Aviv Israel
- Sagol School of Neuroscience Tel Aviv University Tel Aviv Israel
| | - Hilit Levy‐Barazany
- Department of Neurobiology George S. Wise Faculty of Life Sciences Tel Aviv University Tel Aviv Israel
| | - Yuval Nash
- Department of Neurobiology George S. Wise Faculty of Life Sciences Tel Aviv University Tel Aviv Israel
- Sagol School of Neuroscience Tel Aviv University Tel Aviv Israel
| | - Liron Samuel
- Department of Neurobiology George S. Wise Faculty of Life Sciences Tel Aviv University Tel Aviv Israel
| | - Ronit Sharon
- Faculty of Medicine Biochemistry and Molecular Biology IMRIC The Hebrew University Jerusalem Jerusalem Israel
| | - Dan Frenkel
- Department of Neurobiology George S. Wise Faculty of Life Sciences Tel Aviv University Tel Aviv Israel
- Sagol School of Neuroscience Tel Aviv University Tel Aviv Israel
| |
Collapse
|
15
|
Rothbard JB, Kurnellas MP, Ousman SS, Brownell S, Rothbard JJ, Steinman L. Small Heat Shock Proteins, Amyloid Fibrils, and Nicotine Stimulate a Common Immune Suppressive Pathway with Implications for Future Therapies. Cold Spring Harb Perspect Med 2019; 9:cshperspect.a034223. [PMID: 30249602 DOI: 10.1101/cshperspect.a034223] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The α7 nicotinic acetylcholine receptor (α7nAChR) is central to the anti-inflammatory function of the vagus nerve in a physiological mechanism termed the inflammatory reflex. Studies on the inflammatory reflex have been instrumental for the current development of the field of bioelectronic medicine. An independent investigation of the biological role of αB-crystallin (HspB5), the most abundant gene transcript present in active multiple sclerosis lesions in human brains, also led to α7nAChR. Induction of experimental autoimmune encephalomyelitis (EAE) in HspB5-/- mice results in greater paralytic signs, increased levels of proinflammatory cytokines, and T-lymphocyte activation relative to wild-type animals. Administration of HspB5 was therapeutic in animal models of multiple sclerosis, retinal and cardiac ischemia, and stroke. Structure-activity studies established that residues 73-92 were as potent as the parent protein, but only when it formed amyloid fibrils. Amyloid fibrils and small heat shock proteins (sHsps) selectively bound α7nAChR on peritoneal macrophages (MΦs) and B lymphocytes, converting the MΦs to an immune suppressive phenotype and mobilizing the migration of both cell types from the peritoneum to secondary lymph organs. Here, we review multiple aspects of this work, which may be of interest for developing future therapeutic approaches for multiple sclerosis and other disorders.
Collapse
Affiliation(s)
- Jonathan B Rothbard
- Department of Neurology, Stanford University School of Medicine, Stanford, California 94305-5316
| | | | - Shalina S Ousman
- Department of Clinical Neurosciences, University of Calgary, Alberta T2N 1N4, Canada
| | - Sara Brownell
- School of Life Sciences, Arizona State University, Tempe, Arizona 85281
| | - Jesse J Rothbard
- Department of Neurology, Stanford University School of Medicine, Stanford, California 94305-5316
| | - Lawrence Steinman
- Department of Neurology, Stanford University School of Medicine, Stanford, California 94305-5316
| |
Collapse
|
16
|
Didonna A, Cantó E, Shams H, Isobe N, Zhao C, Caillier SJ, Condello C, Yamate-Morgan H, Tiwari-Woodruff SK, Mofrad MRK, Hauser SL, Oksenberg JR. Sex-specific Tau methylation patterns and synaptic transcriptional alterations are associated with neural vulnerability during chronic neuroinflammation. J Autoimmun 2019; 101:56-69. [PMID: 31010726 PMCID: PMC6561733 DOI: 10.1016/j.jaut.2019.04.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 04/03/2019] [Accepted: 04/04/2019] [Indexed: 12/19/2022]
Abstract
The molecular events underlying the transition from initial inflammatory flares to the progressive phase of multiple sclerosis (MS) remain poorly understood. Here, we report that the microtubule-associated protein (MAP) Tau exerts a gender-specific protective function on disease progression in the MS model experimental autoimmune encephalomyelitis (EAE). A detailed investigation of the autoimmune response in Tau-deficient mice excluded a strong immunoregulatory role for Tau, suggesting that its beneficial effects are presumably exerted within the central nervous system (CNS). Spinal cord transcriptomic data show increased synaptic dysfunctions and alterations in the NF-kB activation pathway upon EAE in Tau-deficient mice as compared to wildtype animals. We also performed the first comprehensive characterization of Tau post-translational modifications (PTMs) in the nervous system upon EAE. We report that the methylation levels of the conserved lysine residue K306 are significantly decreased in the chronic phase of the disease. By combining biochemical assays and molecular dynamics (MD) simulations, we demonstrate that methylation at K306 decreases the affinity of Tau for the microtubule network. Thus, the down-regulation of this PTM might represent a homeostatic response to enhance axonal stability against an autoimmune CNS insult. The results, altogether, position Tau as key mediator between the inflammatory processes and neurodegeneration that seems to unify many CNS diseases.
Collapse
Affiliation(s)
- Alessandro Didonna
- Department of Neurology and Weill Institute for Neurosciences, University of California at San Francisco, San Francisco, CA, 94158, USA.
| | - Ester Cantó
- Department of Neurology and Weill Institute for Neurosciences, University of California at San Francisco, San Francisco, CA, 94158, USA
| | - Hengameh Shams
- Department of Neurology and Weill Institute for Neurosciences, University of California at San Francisco, San Francisco, CA, 94158, USA
| | - Noriko Isobe
- Department of Neurology and Weill Institute for Neurosciences, University of California at San Francisco, San Francisco, CA, 94158, USA
| | - Chao Zhao
- Department of Neurology and Weill Institute for Neurosciences, University of California at San Francisco, San Francisco, CA, 94158, USA
| | - Stacy J Caillier
- Department of Neurology and Weill Institute for Neurosciences, University of California at San Francisco, San Francisco, CA, 94158, USA
| | - Carlo Condello
- Department of Neurology and Weill Institute for Neurosciences, University of California at San Francisco, San Francisco, CA, 94158, USA; Institute for Neurodegenerative Diseases, University of California, San Francisco, CA, 94158, USA
| | - Hana Yamate-Morgan
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, 92521, USA; Neuroscience Graduate Program, University of California Riverside, Riverside, CA, 92521, USA
| | - Seema K Tiwari-Woodruff
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, 92521, USA; Neuroscience Graduate Program, University of California Riverside, Riverside, CA, 92521, USA; Center for Glial-Neuronal Interactions, UCR School of Medicine, CA, 92506, USA
| | - Mohammad R K Mofrad
- Molecular Cell Biomechanics Laboratory, Departments of Bioengineering and Mechanical Engineering, University of California, Berkeley, CA, 94720, USA; Physical Biosciences Division, Lawrence Berkeley National Lab, Berkeley, CA, 94720, USA
| | - Stephen L Hauser
- Department of Neurology and Weill Institute for Neurosciences, University of California at San Francisco, San Francisco, CA, 94158, USA
| | - Jorge R Oksenberg
- Department of Neurology and Weill Institute for Neurosciences, University of California at San Francisco, San Francisco, CA, 94158, USA
| |
Collapse
|
17
|
Wang Y, Liu C, Wang H, Jiang Y, Wang P, Shang H. Systematic Review of Basic Research on Alzheimer's Disease with Shen Zhi Ling Oral Liquid. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2019; 2019:8216714. [PMID: 31118971 PMCID: PMC6500647 DOI: 10.1155/2019/8216714] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 02/28/2019] [Accepted: 03/18/2019] [Indexed: 11/17/2022]
Abstract
OBJECTIVE The present report systematically reviewed the basic research of Shen Zhi Ling oral liquid (Tiao Xin preparation) treatment on Alzheimer's disease (AD). METHODS CNKI, Wanfang, and VIP were searched, and the literature was selected according to inclusion and exclusion criteria. Data were extracted, and descriptive analysis was used. RESULTS Twenty-four articles were included, all of which were published as "Tiao Xin preparation." There were seven types of AD models involved. The mechanism of action of Shen Zhi Ling oral liquid in the treatment of AD primarily included suppression of Aβ deposition and tau hyperphosphorylation, regulation of multiple neurotransmitters, improvement in energy metabolism, and promotion of the expression of autophagy-related and learning-memory-associated proteins. CONCLUSIONS AD is a complex disease caused by multiple factors. Shen Zhi Ling oral liquid exhibited multiple and multitarget effects and great potential for treating AD. The continuous development of molecular biology and related disciplines will further elucidate the mechanism of Shen Zhi Ling oral liquid intervention in AD.
Collapse
Affiliation(s)
- Yahan Wang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Haiyuncang Lane, Dongcheng District, Beijing 100700, China
| | - Chunxiang Liu
- Evidence-Based Medicine Center, Tianjin University of Chinese Medicine, Beihua South Road, Jinghai District, Tianjin 301617, China
| | - Hui Wang
- Evidence-Based Medicine Center, Tianjin University of Chinese Medicine, Beihua South Road, Jinghai District, Tianjin 301617, China
| | - Yin Jiang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Haiyuncang Lane, Dongcheng District, Beijing 100700, China
| | - Pengwen Wang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Haiyuncang Lane, Dongcheng District, Beijing 100700, China
| | - Hongcai Shang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Haiyuncang Lane, Dongcheng District, Beijing 100700, China
- Center for Evidence-Based and Translational Medicine, Jiangxi University of Traditional Chinese Medicine, Wanli District, Nanchang, Jiangxi 330000, China
| |
Collapse
|
18
|
Zhang XX, Qiao YC, Li W, Zou X, Chen YL, Shen J, Liao QY, Zhang QJ, He L, Zhao HL. Human amylin induces CD4+Foxp3+ regulatory T cells in the protection from autoimmune diabetes. Immunol Res 2019; 66:179-186. [PMID: 28983871 DOI: 10.1007/s12026-017-8956-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Autoimmune diabetes is a disorder of immune homeostasis that leads to targeted insulin-secreting islet β cell destruction characterized by insulitis. Human amylin (hA) is an important neuroendocrine hormone co-secreted with insulin by pancreatic β cells. Here, we report hA immune-modulatory action through inducing regulatory T cells. We ex vivo-treated human peripheral blood mononuclear cells (hPBMCs) with hA for 24 h and counted CD4+Foxp3+ regulatory T cells (Treg) using flow cytometry. Diabetic status was monitored and splenic Treg were measured in non-obese diabetic (NOD) male mice. NOD mice were intraperitoneally injected once daily with hA (n = 25) or solvent for control (n = 25) for 7 months continuously. Spleen tissues were collected at the end of intervention and processed for flow cytometry and Western blot. We found a 2.9-fold (p < 0.05) increase of CD4+Foxp3+ Treg in hPBMCs treated with 10 nmol/L hA compared with negative control. Incidence of diabetes in hA-treated NOD mice decreased 44% (p = 0.045) in the 6th month and 57% (p = 0.0002) in the 7th month. Meanwhile, the hA treatment induced a 1.5-fold increase of CD4+Foxp3+ Treg from mouse splenocytes (p = 0.0013). Expression of transforming growth factor-β (TGF-β) and toll-like receptor-4 (TLR-4) were upregulated in hA-treated mice. Human amylin might protect against autoimmune diabetes via the induction of CD4+Foxp3+ Treg, which suggests a novel approach to improve autoimmune conditions.
Collapse
Affiliation(s)
- Xiao-Xi Zhang
- Centre of Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, and Department of Immunology, Guangxi Area of Excellence, Guilin Medical University, Huan Cheng North 2nd Road 109, Guilin, Guangxi, 541004, China
| | - Yong-Chao Qiao
- Department of Immunology, School of Basic Medical Sciences, Central South University, Changsha, Hunan, 410078, China
| | - Wan Li
- Centre of Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, and Department of Immunology, Guangxi Area of Excellence, Guilin Medical University, Huan Cheng North 2nd Road 109, Guilin, Guangxi, 541004, China
| | - Xia Zou
- Centre of Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, and Department of Immunology, Guangxi Area of Excellence, Guilin Medical University, Huan Cheng North 2nd Road 109, Guilin, Guangxi, 541004, China
| | - Yin-Ling Chen
- Centre of Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, and Department of Immunology, Guangxi Area of Excellence, Guilin Medical University, Huan Cheng North 2nd Road 109, Guilin, Guangxi, 541004, China
| | - Jian Shen
- Centre of Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, and Department of Immunology, Guangxi Area of Excellence, Guilin Medical University, Huan Cheng North 2nd Road 109, Guilin, Guangxi, 541004, China
| | - Qin-Yuan Liao
- Centre of Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, and Department of Immunology, Guangxi Area of Excellence, Guilin Medical University, Huan Cheng North 2nd Road 109, Guilin, Guangxi, 541004, China
| | - Qiu-Jin Zhang
- Centre of Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, and Department of Immunology, Guangxi Area of Excellence, Guilin Medical University, Huan Cheng North 2nd Road 109, Guilin, Guangxi, 541004, China
| | - Lan He
- Department of Microbiology, Prince of Wales Hospital, Shatin, Hong Kong
| | - Hai-Lu Zhao
- Centre of Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, and Department of Immunology, Guangxi Area of Excellence, Guilin Medical University, Huan Cheng North 2nd Road 109, Guilin, Guangxi, 541004, China. .,Department of Immunology, Guilin Medical University, Guilin, Guangxi, 541004, China.
| |
Collapse
|
19
|
Wang R, Chen ZH, Wang Y, Huang HB, Fan SJ, Chen LL. Recombination and identification of human alpha B-crystallin. Int J Ophthalmol 2018; 11:1916-1921. [PMID: 30588422 DOI: 10.18240/ijo.2018.12.06] [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: 05/16/2018] [Accepted: 10/22/2018] [Indexed: 11/23/2022] Open
Abstract
AIM To recombine the human alpha B-crystallin (αB-crystallin) using gene cloning technology and prokaryotic expression vector and confirm the biological activity of recombinant human αB-crystallin. METHODS Cloning the human αB-crystallin cDNA according to the nucleotide sequence of the human αB-crystallin, constructing the pET-28/CRYAB prokaryotic expression plasmid by restriction enzyme digestion method, and stably expressing transformed into the Escherichia coli (E. coli) DH5 alpha. The recombinant human αB-crystallin was purified by Q sepharose. By enzyme digestion analysis, Western blotting and sequencing, the recombinant human αB-crystallin was identified and the activity of its molecular protein was detected. RESULTS Compared with the gene bank (GeneBank), the cloned human sequence of human αB-crystallin cDNA has the same open reading frame. Identification and sequencing of the cloned human αB-crystallin cDNA in prokaryotic expression vector confirmed the full length sequence, and the vector was constructed successfully. The E. coli containing plasmid pET-28/CRYAB induced by isopropyl-β-D-thiogalactoside successfully expressed the human αB-crystallin. Insulin confirmed that the recombinant human αB-crystallin has a molecular chaperone activity. CONCLUSION The prokaryotic expression vector pET-28/CRYAB of recombinant human αB-crystallin is successfully constructed, and the recombinant human αB-crystallin with molecular chaperone activity is obtained, which lay a foundation for the research and application of the recombinant human αB-crystallin and its chaperone activity.
Collapse
Affiliation(s)
- Rui Wang
- Department of Ophthalmology, Hainan Branch of PLA General Hospital, Sanya 572000, Hainan Province, China
| | - Ze-Hua Chen
- Department of Ophthalmology, Hainan Branch of PLA General Hospital, Sanya 572000, Hainan Province, China
| | - Yi Wang
- Chongqing Aier General Hospital, Aier School of Ophthalmology, Central South University, Chongqing 400020, China
| | - Hou-Bin Huang
- Department of Ophthalmology, Hainan Branch of PLA General Hospital, Sanya 572000, Hainan Province, China
| | - Si-Jun Fan
- PLA Rocket Force General Hospital, Beijing 100088, China
| | - Lan-Lan Chen
- Department of Ophthalmology, Hainan Branch of PLA General Hospital, Sanya 572000, Hainan Province, China
| |
Collapse
|
20
|
Song JY, Larson NR, Thati S, Torres-Vazquez I, Martinez-Rivera N, Subelzu NJ, Leon MA, Rosa-Molinar E, Schöneich C, Forrest ML, Middaugh CR, Berkland CJ. Glatiramer acetate persists at the injection site and draining lymph nodes via electrostatically-induced aggregation. J Control Release 2018; 293:36-47. [PMID: 30414463 DOI: 10.1016/j.jconrel.2018.11.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 10/30/2018] [Accepted: 11/04/2018] [Indexed: 01/01/2023]
Abstract
Glatiramer acetate (GA) is widely prescribed for the treatment of relapsing-remitting multiple sclerosis, however, the mechanism of action is still not fully understood. We investigated the structural properties of GA and examined alterations to the drug upon injection into the subcutaneous space. First, a variety of biophysical characterization techniques were employed to characterize GA in solution. GA was found to exist as alpha helices in solution with a hydrodynamic radius of ~3 nm in size. To simulate GA behavior at the site of injection, GA was injected into a solution of 1.5 MDa hyaluronic acid (HA). Visible aggregates were observed immediately upon injection and subsequent testing indicated aggregation was driven by electrostatic interactions between the positively-charged GA and negatively-charged HA. In vivo testing confirmed GA formed spherical particles in the nano- to micrometer size range, suggesting this mechanism contributes to persistence at the injection site and in draining lymph nodes. The aggregates were found to associate with glycosaminoglycans, suggesting an electrostatic mechanism of induced aggregation like the simulated injection. These novel observations may help explain the complex immunomodulatory mechanisms of GA and adverse injection site reactions seen in patients.
Collapse
Affiliation(s)
- Jimmy Y Song
- Department of Pharmaceutical Chemistry, University of Kansas, USA
| | | | - Sharadvi Thati
- Department of Pharmaceutical Chemistry, University of Kansas, USA
| | - Irma Torres-Vazquez
- Microscopy and Analytical Imaging Laboratory, University of Kansas, USA; Department of Pharmacology and Toxicology, University of Kansas, USA
| | - Noraida Martinez-Rivera
- Microscopy and Analytical Imaging Laboratory, University of Kansas, USA; Department of Pharmacology and Toxicology, University of Kansas, USA
| | | | | | - Eduardo Rosa-Molinar
- Department of Bioengineering, University of Kansas, USA; Microscopy and Analytical Imaging Laboratory, University of Kansas, USA; Department of Pharmacology and Toxicology, University of Kansas, USA
| | | | - M Laird Forrest
- Department of Pharmaceutical Chemistry, University of Kansas, USA
| | | | - Cory J Berkland
- Department of Pharmaceutical Chemistry, University of Kansas, USA; Department of Chemistry, University of Kansas, USA; Department of Bioengineering, University of Kansas, USA; Department of Chemical and Petroleum Engineering, University of Kansas, USA.
| |
Collapse
|
21
|
Mahapatra S, Ying L, Ho PPK, Kurnellas M, Rothbard J, Steinman L, Cornfield DN. An amyloidogenic hexapeptide derived from amylin attenuates inflammation and acute lung injury in murine sepsis. PLoS One 2018; 13:e0199206. [PMID: 29990318 PMCID: PMC6039005 DOI: 10.1371/journal.pone.0199206] [Citation(s) in RCA: 2] [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: 03/04/2018] [Accepted: 06/04/2018] [Indexed: 12/03/2022] Open
Abstract
Although the accumulation of amyloidogenic proteins in neuroinflammatory conditions is generally considered pathologic, in a murine model of multiple sclerosis, amyloid-forming fibrils, comprised of hexapeptides, are anti-inflammatory. Whether these molecules modulate systemic inflammatory conditions remains unknown. We hypothesized that an amylin hexapeptide that forms fibrils can attenuate the systemic inflammatory response in a murine model of sepsis. To test this hypothesis, mice were pre-treated with either vehicle or amylin hexapeptide (20 μg) at 12 hours and 6 hours prior to intraperitoneal (i.p.) lipopolysaccharide (LPS, 20 mg/kg) administration. Illness severity and survival were monitored every 6 hours for 3 days. Levels of pro- (IL-6, TNF-α, IFN-γ) and anti-inflammatory (IL-10) cytokines were measured via ELISA at 1, 3, 6, 12, and 24 hours after LPS (i.p.). As a metric of lung injury, pulmonary artery endothelial cell (PAEC) barrier function was tested 24 hours after LPS administration by comparing lung wet-to-dry ratios, Evan’s blue dye (EBD) extravasation, lung histology and caspase-3 activity. Compared to controls, pretreatment with amylin hexapeptide significantly reduced mortality (p<0.05 at 72 h), illness severity (p<0.05), and pro-inflammatory cytokine levels, while IL-10 levels were elevated (p<0.05). Amylin pretreatment attenuated LPS-induced lung injury, as demonstrated by decreased lung water and caspase-3 activity (p<0.05, versus PBS). Hence, in a murine model of systemic inflammation, pretreatment with amylin hexapeptide reduced mortality, disease severity, and preserved lung barrier function. Amylin hexapeptide may represent a novel therapeutic tool to mitigate sepsis severity and lung injury.
Collapse
Affiliation(s)
- Sidharth Mahapatra
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- * E-mail:
| | - Lihua Ying
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California, United States of America
| | - Peggy Pui-Kay Ho
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, United States of America
| | | | - Jonathan Rothbard
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, United States of America
| | - Lawrence Steinman
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, United States of America
| | - David N. Cornfield
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California, United States of America
| |
Collapse
|
22
|
Identification of a common immune regulatory pathway induced by small heat shock proteins, amyloid fibrils, and nicotine. Proc Natl Acad Sci U S A 2018; 115:7081-7086. [PMID: 29915045 DOI: 10.1073/pnas.1804599115] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Although certain dogma portrays amyloid fibrils as drivers of neurodegenerative disease and neuroinflammation, we have found, paradoxically, that amyloid fibrils and small heat shock proteins (sHsps) are therapeutic in experimental autoimmune encephalomyelitis (EAE). They reduce clinical paralysis and induce immunosuppressive pathways, diminishing inflammation. A key question was the identification of the target for these molecules. When sHsps and amyloid fibrils were chemically cross-linked to immune cells, a limited number of proteins were precipitated, including the α7 nicotinic acetylcholine receptor (α7 NAChR). The α7 NAChR is noteworthy among the over 20 known receptors for amyloid fibrils, because it plays a central role in a well-defined immune-suppressive pathway. Competitive binding between amyloid fibrils and α-bungarotoxin to peritoneal macrophages (MΦs) confirmed the involvement of α7 NAChR. The mechanism of immune suppression was explored, and, similar to nicotine, amyloid fibrils inhibited LPS induction of a common set of inflammatory cytokines while inducing Stat3 signaling and autophagy. Consistent with this, previous studies have established that nicotine, sHsps, and amyloid fibrils all were effective therapeutics in EAE. Interestingly, B lymphocytes were needed for the therapeutic effect. These results suggest that agonists of α7 NAChR might have therapeutic benefit for a variety of inflammatory diseases.
Collapse
|
23
|
Mørkholt AS, Kastaniegaard K, Trabjerg MS, Gopalasingam G, Niganze W, Larsen A, Stensballe A, Nielsen S, Nieland JD. Identification of brain antigens recognized by autoantibodies in experimental autoimmune encephalomyelitis-induced animals treated with etomoxir or interferon-β. Sci Rep 2018; 8:7092. [PMID: 29728570 PMCID: PMC5935685 DOI: 10.1038/s41598-018-25391-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 04/19/2018] [Indexed: 11/08/2022] Open
Abstract
Multiple sclerosis (MS) is a neurodegenerative autoimmune disease, where chronic inflammation plays an essential role in its pathology. A feature of MS is the production of autoantibodies stimulated by an altered-peptide-ligand response and epitope spreading, resulting in loss of tolerance for self-proteins. The involvement of autoantibodies in MS pathogenesis has been suggested to initiate and drive progression of inflammation; however, the etiology of MS remains unknown. The effect of etomoxir and interferon-β (IFN-β) was examined in an experimental-autoimmune-encephalomyelitis (EAE) model of MS. Moreover, the impact of etomoxir and IFN-β on recognition of brain proteins in serum from EAE rats was examined with the purpose of identifying the autoantibody reactivities involved in MS. Animals treated with etomoxir on day 1 exhibited a statistically significantly lower disease score than animals treated with IFN-β (on day 1 or 5) or placebo. Etomoxir treatment on day 5 resulted in a significantly lower disease score than IFN-β treatment on day 1. After disease induction antibodies was induced to a broad pallet of antigens in the brain. Surprisingly, by blocking CPT1 and therewith lipid metabolism several alterations in the antibody response was observed suggesting that autoantibodies play a role in the EAE animal model.
Collapse
Affiliation(s)
| | | | | | - Gopana Gopalasingam
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Wanda Niganze
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Agnete Larsen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Allan Stensballe
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Søren Nielsen
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - John Dirk Nieland
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark.
| |
Collapse
|
24
|
Upadhyay A, Mishra A. Amyloids of multiple species: are they helpful in survival? Biol Rev Camb Philos Soc 2018; 93:1363-1386. [DOI: 10.1111/brv.12399] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 01/13/2018] [Accepted: 01/18/2018] [Indexed: 12/12/2022]
Affiliation(s)
- Arun Upadhyay
- Cellular and Molecular Neurobiology Unit; Indian Institute of Technology Jodhpur; Rajasthan 342011 India
| | - Amit Mishra
- Cellular and Molecular Neurobiology Unit; Indian Institute of Technology Jodhpur; Rajasthan 342011 India
| |
Collapse
|
25
|
Dubuisson N, Puentes F, Giovannoni G, Gnanapavan S. Science is 1% inspiration and 99% biomarkers. Mult Scler 2017; 23:1442-1452. [PMID: 28537780 DOI: 10.1177/1352458517709362] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Neurodegeneration plays a key role in multiple sclerosis (MS) contributing to long-term disability in patients. The prognosis is, however, unpredictable coloured by complex disease mechanisms which can only be clearly appreciated using biomarkers specific to pathobiology of the underlying process. Here, we describe six promising neurodegenerative biomarkers in MS (neurofilament proteins, neurofilament antibodies, tau, N-acetylaspartate, chitinase and chitinase-like proteins and osteopontin), critically evaluating the evidence using a modified Bradford Hill criteria.
Collapse
Affiliation(s)
- Nicolas Dubuisson
- Department of Neuroscience and Trauma, Blizard Institute, Queen Mary University of London, London, UK
| | - Fabiola Puentes
- Department of Neuroscience and Trauma, Blizard Institute, Queen Mary University of London, London, UK
| | - Gavin Giovannoni
- Department of Neuroscience and Trauma, Blizard Institute, Queen Mary University of London, London, UK
| | - Sharmilee Gnanapavan
- Department of Neuroscience and Trauma, Blizard Institute, Queen Mary University of London, London, UK
| |
Collapse
|
26
|
Amylin and its G-protein-coupled receptor: A probable pathological process and drug target for Alzheimer's disease. Neuroscience 2017; 356:44-51. [PMID: 28528968 DOI: 10.1016/j.neuroscience.2017.05.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 05/11/2017] [Accepted: 05/12/2017] [Indexed: 12/31/2022]
Abstract
G-protein-coupled receptors (GPCRs) are shown to be involved in Alzheimer's disease (AD) pathogenesis. However, because GPCRs include a large family of membrane receptors, it is unclear which specific GPCR or pathway with rational ligands can become effective therapeutic targets for AD. Amylin receptor (AmR) is a GPCR that mediates several activities, such as improving glucose metabolism, relaxing cerebrovascular structure, modulating inflammatory reactions and potentially enhancing neural regeneration. Recent studies show that peripheral treatments with amylin or its clinical analog, pramlintide, reduced several components of AD pathology, including amyloid plaques, tauopathy, neuroinflammation and other components in the brain, corresponding with improved learning and memory in AD mouse models. Because amylin shares a similar secondary structure with amyloid-β peptide (Aβ), I propose that the AmR/GPCR pathway is disturbed by a large amount of Aβ in the AD brain, leading to tau phosphorylation, neuroinflammation and neuronal death in the pathological cascade. Amylin-type peptides, readily crossing the blood-brain barrier (BBB), are the rational ligands to enhance this GPCR pathway and may exhibit utility as novel therapeutic agents for treating AD.
Collapse
|
27
|
Zhu H, Xue X, Wang E, Wallack M, Na H, Hooker JM, Kowall N, Tao Q, Stein TD, Wolozin B, Qiu WQ. Amylin receptor ligands reduce the pathological cascade of Alzheimer's disease. Neuropharmacology 2017; 119:170-181. [PMID: 28363773 DOI: 10.1016/j.neuropharm.2017.03.030] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 03/21/2017] [Accepted: 03/25/2017] [Indexed: 01/01/2023]
Abstract
Amylin is an important gut-brain axis hormone. Since amylin and amyloid-β peptide (Aβ) share similar β sheet secondary structure despite not having the same primary sequences, we hypothesized that the accumulation of Aβ in the brains of subjects with Alzheimer's disease (AD) might compete with amylin for binding to the amylin receptor (AmR). If true, adding exogenous amylin type peptides would compete with Aβ and reduce the AD pathological cascade, improving cognition. Here we report that a 10-week course of peripheral treatment with human amylin significantly reduced multiple different markers associated with AD pathology, including reducing levels of phospho-tau, insoluble tau, two inflammatory markers (Iba1 and CD68), as well as cerebral Aβ. Amylin treatment also led to improvements in learning and memory in two AD mouse models. Mechanistic studies showed that an amylin receptor antagonist successfully antagonized some protective effects of amylin in vivo, suggesting that the protective effects of amylin require interaction with its cognate receptor. Comparison of signaling cascades emanating from AmR suggest that amylin electively suppresses activation of the CDK5 pathway by Aβ. Treatment with amylin significantly reduced CDK5 signaling in a receptor dependent manner, dramatically decreasing the levels of p25, the active form of CDK5 with a corresponding reduction in tau phosphorylation. This is the first report documenting the ability of amylin treatment to reduce tauopathy and inflammation in animal models of AD. The data suggest that the clinical analog of amylin, pramlintide, might exhibit utility as a therapeutic agent for AD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Haihao Zhu
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA.
| | - Xiehua Xue
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA; Affiliated Rehabilitation Hospital of Fujian, TCM University, China
| | - Erming Wang
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Max Wallack
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA; Harvard Medical School, USA
| | - Hana Na
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Jacob M Hooker
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, USA; Harvard Medical School, USA
| | - Neil Kowall
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA; Alzheimer's Disease Center, Boston University School of Medicine, Boston, MA, USA
| | - Qiushan Tao
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Thor D Stein
- Department of Pathology, Boston University School of Medicine, Boston, MA, USA; Alzheimer's Disease Center, Boston University School of Medicine, Boston, MA, USA
| | - Benjamin Wolozin
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA; Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Wei Qiao Qiu
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA; Alzheimer's Disease Center, Boston University School of Medicine, Boston, MA, USA; Department of Psychiatry, Boston University School of Medicine, Boston, MA, USA.
| |
Collapse
|
28
|
Zhu H, Stern RA, Tao Q, Bourlas A, Essis MD, Chivukula M, Rosenzweig J, Steenkamp D, Xia W, Mercier GA, Tripodis Y, Farlow M, Kowall N, Qiu WQ. An amylin analog used as a challenge test for Alzheimer's disease. ALZHEIMERS & DEMENTIA-TRANSLATIONAL RESEARCH & CLINICAL INTERVENTIONS 2017; 3:33-43. [PMID: 28503657 PMCID: PMC5424531 DOI: 10.1016/j.trci.2016.12.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Preclinical studies demonstrate the potential of amylin in the diagnosis of Alzheimer's disease (AD). We aimed to lay the foundation for repurposing the amylin analog and a diabetes drug, pramlintide, for AD in humans. METHODS We administered a single subcutaneous injection of 60 μg of pramlintide to nondiabetic subjects under fasting conditions. RESULTS None of the participants developed hypoglycemia after the injection of pramlintide. The pramlintide challenge induced a significant surge of amyloid-β peptide and a decrease in total tau in the plasma of AD subjects but not in control participants. The pramlintide injection provoked an increase in interleukin 1 receptor antagonist and a decrease in retinol-binding protein 4, which separates AD subjects from control subjects. DISCUSSION Pramlintide use appeared to be safe in the absence of diabetes. The biomarker changes as a result of the pramlintide challenge, which distinguished AD from control subjects and mild cognitive impairment.
Collapse
Affiliation(s)
- Haihao Zhu
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Robert A Stern
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA.,Department of Neurosurgery, Boston University School of Medicine, Boston, MA, USA.,Alzheimer's Disease Center, Boston University School of Medicine, Boston, MA, USA
| | - Qiushan Tao
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Alexandra Bourlas
- Alzheimer's Disease Center, Boston University School of Medicine, Boston, MA, USA
| | - Maritza D Essis
- Alzheimer's Disease Center, Boston University School of Medicine, Boston, MA, USA
| | - Meenakshi Chivukula
- Alzheimer's Disease Center, Boston University School of Medicine, Boston, MA, USA
| | - James Rosenzweig
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Devin Steenkamp
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Weiming Xia
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Gustavo A Mercier
- Department of Radiology, Boston University School of Medicine, Boston, MA, USA
| | - Yorghos Tripodis
- Department of Psychiatry, Boston University School of Medicine, Boston, MA, USA
| | - Martin Farlow
- Alzheimer's Disease Center, Indiana University, Indianapolis, IN, USA
| | - Neil Kowall
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA.,Alzheimer's Disease Center, Boston University School of Medicine, Boston, MA, USA
| | - Wei Qiao Qiu
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA.,Alzheimer's Disease Center, Boston University School of Medicine, Boston, MA, USA.,Department of Psychiatry, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
29
|
Genetic background modulates outcome of therapeutic amyloid peptides in treatment of neuroinflammation. J Neuroimmunol 2016; 298:42-50. [PMID: 27609274 DOI: 10.1016/j.jneuroim.2016.06.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 05/27/2016] [Accepted: 06/22/2016] [Indexed: 12/17/2022]
Abstract
Amyloid hexapeptide molecules are effective in the treatment of the murine model of neuroinflammation, known as experimental autoimmune encephalomyelitis (EAE). Efficacy however differs between two inbred mouse strains, C57BL/6J (B6) and C57BL/10SnJ (B10). Amyloid hexapeptide treatments improved the clinical outcomes of B6, but not B10 mice, indicating that genetic background influences therapeutic efficacy. Moreover, although previous studies indicated that prion protein deficiency results in more severe EAE in B6 mice, we observed no such effect in B10 mice. In addition, we found that amyloid hexapeptide treatments of B10 and B6 mice elicited differential IL4 responses. Thus, the modulatory potential of prion protein and related treatments with other amyloid hexapeptides in EAE depends on mouse strain.
Collapse
|
30
|
Bhise V, Dhib-Jalbut S. Further understanding of the immunopathology of multiple sclerosis: impact on future treatments. Expert Rev Clin Immunol 2016; 12:1069-89. [PMID: 27191526 DOI: 10.1080/1744666x.2016.1191351] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION The understanding of the immunopathogenesis of multiple sclerosis (MS) has expanded with more research into T-cell subtypes, cytokine contributors, B-cell participation, mitochondrial dysfunction, and more. Treatment options have rapidly expanded with three relatively recent oral therapy alternatives entering the arena. AREAS COVERED In the following review, we discuss current mechanisms of immune dysregulation in MS, how they relate to current treatments, and the impact these findings will have on the future of therapy. Expert commentary: The efficacy of these medications and understanding their mechanisms of actions validates the immunopathogenic mechanisms thought to underlie MS. Further research has exposed new targets, while new promising therapies have shed light on new aspects into the pathophysiology of MS.
Collapse
Affiliation(s)
- Vikram Bhise
- a Rutgers Biomedical and Health Sciences - Departments of Pediatrics , Robert Wood Johnson Medical School , New Brunswick , NJ , USA
| | - Suhayl Dhib-Jalbut
- b Rutgers Biomedical and Health Sciences - Departments of Neurology , Robert Wood Johnson Medical School , New Brunswick , NJ , USA
| |
Collapse
|
31
|
Steinman L. A Journey in Science: The Privilege of Exploring the Brain and the Immune System. Mol Med 2016; 22:molmed.2015.00263. [PMID: 27652378 PMCID: PMC5004718 DOI: 10.2119/molmed.2015.00263] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 12/22/2015] [Indexed: 11/06/2022] Open
Abstract
Real innovations in medicine and science are historic and singular; the stories behind each occurrence are precious. At Molecular Medicine we have established the Anthony Cerami Award in Translational Medicine to document and preserve these histories. The monographs recount the seminal events as told in the voice of the original investigators who provided the crucial early insight. These essays capture the essence of discovery, chronicling the birth of ideas that created new fields of research; and launched trajectories that persisted and ultimately influenced how disease is prevented, diagnosed, and treated. In this volume, the Cerami Award Monograph is by Lawrence Steinman, MD, of Stanford University in California. A visionary in the field of neurology, this is the story of Dr. Steinman's scientific journey.
Collapse
Affiliation(s)
- Lawrence Steinman
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, California
| |
Collapse
|
32
|
Matías-Guiu JA, Oreja-Guevara C, Cabrera-Martín MN, Moreno-Ramos T, Carreras JL, Matías-Guiu J. Amyloid Proteins and Their Role in Multiple Sclerosis. Considerations in the Use of Amyloid-PET Imaging. Front Neurol 2016; 7:53. [PMID: 27065425 PMCID: PMC4814935 DOI: 10.3389/fneur.2016.00053] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 03/22/2016] [Indexed: 02/06/2023] Open
Abstract
Thioflavin T derivatives are used in positron-emission tomography (PET) studies to detect amyloid protein deposits in patients with Alzheimer disease. These tracers bind extensively to white matter, which suggests that they may be useful in studies of multiple sclerosis (MS), and that proteins resulting from proteolytic processing of the amyloid precursor protein (APP) may contribute to MS. This article reviews data from both clinical and preclinical studies addressing the role of these proteins, whether they are detected in CSF studies or using PET imaging. APP is widely expressed in demyelinated axons and may have a protective effect in MS and in experimental allergic encephalomyelitis in animals. Several mechanisms associated with this increased expression may affect the degree of remyelination in MS. Amyloid-PET imaging may help determine the degree of demyelination and provide information on the molecular changes linked to APP proteolytic processing experienced by patients with MS.
Collapse
Affiliation(s)
- Jordi A Matías-Guiu
- Department of Neurology, Hospital Clínico San Carlos, San Carlos Institute for Health Research (IdISSC), Complutense University of Madrid , Madrid , Spain
| | - Celia Oreja-Guevara
- Department of Neurology, Hospital Clínico San Carlos, San Carlos Institute for Health Research (IdISSC), Complutense University of Madrid , Madrid , Spain
| | - María Nieves Cabrera-Martín
- Department of Nuclear Medicine, Hospital Clínico San Carlos, San Carlos Institute for Health Research (IdISSC), Complutense University of Madrid , Madrid , Spain
| | - Teresa Moreno-Ramos
- Department of Neurology, Hospital Clínico San Carlos, San Carlos Institute for Health Research (IdISSC), Complutense University of Madrid , Madrid , Spain
| | - José Luis Carreras
- Department of Nuclear Medicine, Hospital Clínico San Carlos, San Carlos Institute for Health Research (IdISSC), Complutense University of Madrid , Madrid , Spain
| | - Jorge Matías-Guiu
- Department of Neurology, Hospital Clínico San Carlos, San Carlos Institute for Health Research (IdISSC), Complutense University of Madrid , Madrid , Spain
| |
Collapse
|
33
|
αB-crystallin is essential for the TGF-β2-mediated epithelial to mesenchymal transition of lens epithelial cells. Biochem J 2016; 473:1455-69. [PMID: 26987815 DOI: 10.1042/bcj20160128] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 03/16/2016] [Indexed: 01/01/2023]
Abstract
Transforming growth factor (TGF)-β2-mediated pathways play a major role in the epithelial to mesenchymal transition (EMT) of lens epithelial cells (LECs) during secondary cataract formation, which is also known as posterior capsule opacification (PCO). Although αB-crystallin is a major protein in LEC, its role in the EMT remains unknown. In a human LEC line (FHL124), TGF-β2 treatment resulted in changes in the EMT-associated proteins at the mRNA and protein levels. This was associated with nuclear localization of αB-crystallin, phosphorylated Smad2 (pSmad2) (S245/250/255), pSmad3 (S423/425), Smad4 and Snail and the binding of αB-crystallin to these transcription factors, all of which were reduced by the down-regulation of αB-crystallin. Expression of the functionally defective R120G mutant of αB-crystallin reduced TGF-β2-induced EMT in LECs of αB-crystallin knockout (KO) mice. Treatment of bovine lens epithelial explants and mouse LEC with TGF-β2 resulted in changes in the EMT-associated proteins at the mRNA and protein levels. This was accompanied by increase in phosphorylation of p44/42 mitogen-activated protein kinases (MAPK) (T202/Y204), p38 MAPK (T180/Y182), protein kinase B (Akt) (S473) and Smad2 when compared with untreated cells. These changes were significantly reduced in αB-crystallin depleted or knocked out LEC. The removal of the fibre cell mass from the lens of wild-type (WT) mice resulted in the up-regulation of EMT-associated genes in the capsule-adherent epithelial cells, which was reduced in the αB-crystallin KO mice. Together, our data show that αB-crystallin plays a central role in the TGF-β2-induced EMT of LEC. αB-Crystallin could be targeted to prevent PCO and pathological fibrosis in other tissues.
Collapse
|
34
|
Profile of Lawrence Steinman. Proc Natl Acad Sci U S A 2016; 113:1468-70. [PMID: 26811468 DOI: 10.1073/pnas.1600083113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
35
|
Abstract
PURPOSE OF REVIEW Recent studies indicate a role for immune dysregulation in the pathogenesis of multiple sclerosis, an inflammatory demyelinating and degenerative disease of the central nervous system. This review addresses the current mechanisms of immune dysregulation in the development of multiple sclerosis, including the impact of environmental risk factors on immunity in both multiple sclerosis and its animal models. RECENT FINDINGS CD4 T-helper (Th) cells have long been implicated as the main drivers of pathogenesis of multiple sclerosis. However, current studies indicate that multiple sclerosis is largely a heterogeneous disease process, which involves both innate and adaptive immune-mediated inflammatory mechanisms that ultimately contribute to demyelination and neurodegeneration. Therefore, B cells, CD8 T cells, and microglia/macrophages can also play an important role in the immunopathogenesis of multiple sclerosis apart from proinflammatory CD4 Th1/Th17 cell subsets. Furthermore, increasing evidence indicates that environmental risk factors, such as Vitamin D deficiency, Epstein-Barr virus, smoking, Western diet, and the commensal microbiota, influence the development of multiple sclerosis through interactions with genetic variants of multiple sclerosis, thus leading to the dysregulation of immune responses. SUMMARY A better understanding of immune-mediated mechanisms in the pathogenesis of multiple sclerosis and the contribution of environmental risk factors toward the development of multiple sclerosis will help further improve therapeutic approaches to prevent disease progression.
Collapse
|
36
|
Kurnellas MP, Ghosn EEB, Schartner JM, Baker J, Rothbard JJ, Negrin RS, Herzenberg LA, Fathman CG, Steinman L, Rothbard JB. Amyloid fibrils activate B-1a lymphocytes to ameliorate inflammatory brain disease. Proc Natl Acad Sci U S A 2015; 112:15016-23. [PMID: 26621719 PMCID: PMC4679000 DOI: 10.1073/pnas.1521206112] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Amyloid fibrils composed of peptides as short as six amino acids are therapeutic in experimental autoimmune encephalomyelitis (EAE), reducing paralysis and inflammation, while inducing several pathways of immune suppression. Intraperitoneal injection of fibrils selectively activates B-1a lymphocytes and two populations of resident macrophages (MΦs), increasing IL-10 production, and triggering their exodus from the peritoneum. The importance of IL-10-producing B-1a cells in this effective therapy was established in loss-of-function experiments where neither B-cell-deficient (μMT) nor IL10(-/-) mice with EAE responded to the fibrils. In gain-of-function experiments, B-1a cells, adoptively transferred to μMT mice with EAE, restored their therapeutic efficacy when Amylin 28-33 was administered. Stimulation of adoptively transferred bioluminescent MΦs and B-1a cells by amyloid fibrils resulted in rapid (within 60 min of injection) trafficking of both cell types to draining lymph nodes. Analysis of gene expression indicated that the fibrils activated the CD40/B-cell receptor pathway in B-1a cells and induced a set of immune-suppressive cell-surface proteins, including BTLA, IRF4, and Siglec G. Collectively, these data indicate that the fibrils activate B-1a cells and F4/80(+) MΦs, resulting in their migration to the lymph nodes, where IL-10 and cell-surface receptors associated with immune-suppression limit antigen presentation and T-cell activation. These mechanisms culminate in reduction of paralytic signs of EAE.
Collapse
Affiliation(s)
- Michael Phillip Kurnellas
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305
| | - Eliver Eid Bou Ghosn
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305
| | - Jill M Schartner
- Division of Immunology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Jeanette Baker
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Jesse J Rothbard
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305
| | - Robert S Negrin
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Leonore A Herzenberg
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305
| | - C Garrison Fathman
- Division of Immunology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Lawrence Steinman
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305;
| | - Jonathan B Rothbard
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305; Division of Immunology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
37
|
Role of amyloid-β CSF levels in cognitive deficit in MS. Clin Chim Acta 2015; 449:23-30. [DOI: 10.1016/j.cca.2015.01.035] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 01/21/2015] [Indexed: 11/18/2022]
|
38
|
Neural Plasticity in Multiple Sclerosis: The Functional and Molecular Background. Neural Plast 2015; 2015:307175. [PMID: 26229689 PMCID: PMC4503575 DOI: 10.1155/2015/307175] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 06/09/2015] [Accepted: 06/21/2015] [Indexed: 01/19/2023] Open
Abstract
Multiple sclerosis is an autoimmune neurodegenerative disorder resulting in motor dysfunction and cognitive decline. The inflammatory and neurodegenerative changes seen in the brains of MS patients lead to progressive disability and increasing brain atrophy. The most common type of MS is characterized by episodes of clinical exacerbations and remissions. This suggests the presence of compensating mechanisms for accumulating damage. Apart from the widely known repair mechanisms like remyelination, another important phenomenon is neuronal plasticity. Initially, neuroplasticity was connected with the developmental stages of life; however, there is now growing evidence confirming that structural and functional reorganization occurs throughout our lifetime. Several functional studies, utilizing such techniques as fMRI, TBS, or MRS, have provided valuable data about the presence of neuronal plasticity in MS patients. CNS ability to compensate for neuronal damage is most evident in RR-MS; however it has been shown that brain plasticity is also preserved in patients with substantial brain damage. Regardless of the numerous studies, the molecular background of neuronal plasticity in MS is still not well understood. Several factors, like IL-1β, BDNF, PDGF, or CB1Rs, have been implicated in functional recovery from the acute phase of MS and are thus considered as potential therapeutic targets.
Collapse
|
39
|
Abstract
The pathogenesis of Alzheimer's disease (AD) is a critical unsolved question; and although recent studies have demonstrated a strong association between altered brain immune responses and disease progression, the mechanistic cause of neuronal dysfunction and death is unknown. We have previously described the unique CVN-AD mouse model of AD, in which immune-mediated nitric oxide is lowered to mimic human levels, resulting in a mouse model that demonstrates the cardinal features of AD, including amyloid deposition, hyperphosphorylated and aggregated tau, behavioral changes, and age-dependent hippocampal neuronal loss. Using this mouse model, we studied longitudinal changes in brain immunity in relation to neuronal loss and, contrary to the predominant view that AD pathology is driven by proinflammatory factors, we find that the pathology in CVN-AD mice is driven by local immune suppression. Areas of hippocampal neuronal death are associated with the presence of immunosuppressive CD11c(+) microglia and extracellular arginase, resulting in arginine catabolism and reduced levels of total brain arginine. Pharmacologic disruption of the arginine utilization pathway by an inhibitor of arginase and ornithine decarboxylase protected the mice from AD-like pathology and significantly decreased CD11c expression. Our findings strongly implicate local immune-mediated amino acid catabolism as a novel and potentially critical mechanism mediating the age-dependent and regional loss of neurons in humans with AD.
Collapse
|
40
|
Crystallins and neuroinflammation: The glial side of the story. Biochim Biophys Acta Gen Subj 2015; 1860:278-86. [PMID: 26049079 DOI: 10.1016/j.bbagen.2015.05.023] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 05/18/2015] [Accepted: 05/27/2015] [Indexed: 12/31/2022]
Abstract
BACKGROUND There is an abundance of evidence to support the association of damaging neuroinflammation and neurodegeneration across a multitude of diseases. One of the links between these pathological phenomena is the role of chaperone proteins as both neuroprotective and immune-regulatory agents. SCOPE OF REVIEW Chaperone proteins are highly expressed at sites of neuroinflammation both in glial cells and in the injured neurons that initiate the immune response. For this reason, the use of chaperones as treatment for various diseases associated with neuroinflammation is a highly active area of investigation. This review explores the various ways that the small heat shock protein chaperones, α-crystallins, can affect glial cell function with a specific focus on their implication in the inflammatory response associated with neurodegenerative disorders, and their potential as therapeutic treatment. MAJOR CONCLUSIONS Although the mechanisms are still under investigation, a clear link has now been established between alpha-crystallins and neuroinflammation, especially through their roles in microglial and macroglial cells. Interestingly, similar to inflammation in itself, crystallins can have a beneficial or detrimental impact on the CNS based on the context and duration of the condition. GENERAL SIGNIFICANCE Overall this review points out the novel roles that chaperones such as alpha-crystallins can play outside of the classical protein folding pathways, and their potential in the development of new therapies for the treatment of neuroinflammatory/neurodegenerative diseases. This article is part of a Special Issue entitled Crystallin Biochemistry in Health and Disease.
Collapse
|
41
|
Abstract
The brain under immunological attack does not surrender quietly. Investigation of brain lesions in multiple sclerosis (MS) reveals a coordinated molecular response involving various proteins and small molecules ranging from heat shock proteins to small lipids, neurotransmitters, and even gases, which provide protection and foster repair. Reduction of inflammation serves as a necessary prerequisite for effective recovery and regeneration. Remarkably, many lesion-resident molecules activate pathways leading to both suppression of inflammation and promotion of repair mechanisms. These guardian molecules and their corresponding physiologic pathways could potentially be exploited to silence inflammation and repair the injured and degenerating brain and spinal cord in both relapsing-remitting and progressive forms of MS and may be beneficial in other neurologic and psychiatric conditions.
Collapse
|
42
|
Nagaraj RH, Nahomi RB, Mueller NH, Raghavan CT, Ammar DA, Petrash JM. Therapeutic potential of α-crystallin. Biochim Biophys Acta Gen Subj 2015; 1860:252-7. [PMID: 25840354 DOI: 10.1016/j.bbagen.2015.03.012] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 03/26/2015] [Indexed: 01/18/2023]
Abstract
BACKGROUND The findings that α-crystallins are multi-functional proteins with diverse biological functions have generated considerable interest in understanding their role in health and disease. Recent studies have shown that chaperone peptides of α-crystallin could be delivered into cultured cells and in experimental animals with beneficial effects against protein aggregation, oxidation, inflammation and apoptosis. SCOPE OF REVIEW In this review, we will summarize the latest developments on the therapeutic potential of α-crystallins and their functional peptides. MAJOR CONCLUSIONS α-Crystallins and their functional peptides have shown significant favorable effects against several diseases. Their targeted delivery to tissues would be of great therapeutic benefit. However, α-crystallins can also function as disease-causing proteins. These seemingly contradictory functions must be carefully considered prior to their therapeutic use. GENERAL SIGNIFICANCE αA and αB-Crystallin are members of the small heat shock protein family. These proteins exhibit molecular chaperone and anti-apoptotic activities. The core crystallin domain within these proteins is largely responsible for these prosperities. Recent studies have identified peptides within the crystallin domain of both α- and αB-crystallins with remarkable chaperone and anti-apoptotic activities. Administration of α-crystallin or their functional peptides has shown substantial inhibition of pathologies in several diseases. However, α-crystallins have been shown to promote disease-causing pathways. These two sides of the proteins are discussed in this review. This article is part of a Special Issue entitled Crystallin Biochemistry in Health and Disease.
Collapse
Affiliation(s)
- Ram H Nagaraj
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, CO 80045, USA.
| | - Rooban B Nahomi
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Niklaus H Mueller
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Cibin T Raghavan
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - David A Ammar
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - J Mark Petrash
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| |
Collapse
|
43
|
Positive or negative involvement of heat shock proteins in multiple sclerosis pathogenesis: an overview. J Neuropathol Exp Neurol 2015; 73:1092-106. [PMID: 25383635 DOI: 10.1097/nen.0000000000000136] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Multiple sclerosis (MS) is the most diffuse chronic inflammatory disease of the central nervous system. Both immune-mediated and neurodegenerative processes apparently play roles in the pathogenesis of this disease. Heat shock proteins (HSPs) are a family of highly evolutionarily conserved proteins; their expression in the nervous system is induced in a variety of pathologic states, including cerebral ischemia, neurodegenerative diseases, epilepsy, and trauma. To date, investigators have observed protective effects of HSPs in a variety of brain disease models (e.g. of Alzheimer disease and Parkinson disease). In contrast, unequivocal data have been obtained for their roles in MS that depend on the HSP family and particularly on their localization (i.e. intracellular or extracellular). This article reviews our current understanding of the involvement of the principal HSP families in MS.
Collapse
|
44
|
Development of therapies for autoimmune disease at Stanford: a tale of multiple shots and one goal. Immunol Res 2015; 58:307-14. [PMID: 24771483 DOI: 10.1007/s12026-014-8509-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The title of this contribution on Immunology at Stanford is purposely ambiguous. One goal is the development of safe and effective therapy for autoimmune diseases. Another definition of goal is to score, and this would ultimately mean the development of an approved drug. Indeed, the efforts in my four decades at Stanford, have included the discovery and subsequent development of a monoclonal antibody to block homing to the inflamed brain, leading to natalizumab, an approved therapeutic for two autoimmune diseases: relapsing-remitting MS and for inflammatory bowel disease. Multiple attempts to develop new therapies for autoimmune disease are described here: The trimolecular complex and the immune synapse serve as one major set of targets, with attempts to inhibit particular major histocompatibility molecules, the variable regions of the T cell receptor, and CD4. Other approaches focusing on antigen-specific tolerance include ongoing attempts with tolerizing DNA vaccines in type 1 diabetes. Finally, the repurposing of popular drugs approved for other indications, including statins and inhibitors of angiotensin converting enzyme is under development and showing promise in the clinic, particularly for secondary progressive multiple sclerosis. The milieu within Stanford Immunology has helped to nurture these efforts to translate discoveries in immunology and to take them from bench to bedside.
Collapse
|
45
|
Chandra A. Role of amyloid from a multiple sclerosis perspective: a literature review. Neuroimmunomodulation 2015; 22:343-6. [PMID: 25766575 DOI: 10.1159/000375309] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 01/16/2015] [Indexed: 11/19/2022] Open
Abstract
The traditional concept of multiple sclerosis (MS), that it is primarily a white matter inflammatory disease, has changed a great deal. Thanks to the recent development witnessed in MS research, a whole new idea has emerged that MS is a neurodegenerative disease, and neurodegeneration occurs rather earlier in the pathological process. This has also led to the foundation of the hypothesis that two fundamentally different diseases, Alzheimer's disease (AD) and MS, may share a common mechanism of neurodegeneration. Conventionally, amyloid is thought to be a consequence of protein misfolding and aggregation and is most notorious for its association with debilitating and chronic human diseases. Amyloid is implicated to be related with the deterioration and progression of AD. The finding of amyloid precursor protein expression in axons around the plaque in MS, as well as the correlation of amyloid-β (Aβ) with different stages of MS, has clearly indicated that amyloid plays some kind of key role in MS disease pathogenesis. Excitingly, a paradoxical phenomenon of Aβ has also been observed in several studies recently. It has been shown that amyloid might actually be helping in ameliorating the inflammatory effect in diseases like AD and MS. Amyloid imaging allows earlier diagnosis of MS by taking advantage of the relation of amyloid with MS. This will have a big impact on patient diagnosis and management. In this review I have included the findings of research studies dating from several years back to the most recent ones. Through this review I have tried to show the critical role of amyloid in MS and the importance of investigating through PET imaging.
Collapse
Affiliation(s)
- Avinash Chandra
- Buffalo Neuroimaging Analysis Center (BNAC), Department of Neurology, Buffalo General Hospital, Buffalo, N.Y., USA
| |
Collapse
|
46
|
Banerjee PR, Pande A, Shekhtman A, Pande J. Molecular mechanism of the chaperone function of mini-α-crystallin, a 19-residue peptide of human α-crystallin. Biochemistry 2014; 54:505-15. [PMID: 25478825 PMCID: PMC4303307 DOI: 10.1021/bi5014479] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
![]()
α-Crystallin
is the archetypical chaperone of the small heat-shock
protein family, all members of which contain the so-called “α-crystallin
domain” (ACD). This domain and the N- and C-terminal extensions
are considered the main functional units in its chaperone function.
Previous studies have shown that a 19-residue fragment of the ACD
of human αA-crystallin called mini-αA-crystallin (MAC)
shows chaperone properties similar to those of the parent protein.
Subsequent studies have confirmed the function of this peptide, but
no studies have addressed the mechanistic basis for the chaperone
function of MAC. Using human γD-crystallin (HGD), a key substrate
protein for parent α-crystallin in the ocular lens, we show
here that MAC not only protects HGD from aggregation during thermal
and chemical unfolding but also binds weakly and reversibly to HGD
(Kd ≈ 200–700 μM)
even when HGD is in the native state. However, at temperatures favoring
the unfolding of HGD, MAC forms a stable complex with HGD similar
to parent α-crystallin. Using nuclear magnetic resonance spectroscopy,
we identify the residues in HGD that are involved in these two modes
of binding and show that MAC protects HGD from aggregation by binding
to Phe 56 and Val 132 at the domain interface of the target protein,
and residues Val 164 to Leu 167 in the core of the C-terminal domain.
Furthermore, we suggest that the low-affinity, reversible binding
of MAC on the surface of HGD in the native state is involved in facilitating
its binding to both the domain interface and core regions during the
early stages of the unfolding of HGD. This work highlights some structural
features of MAC and MAC-like peptides that affect their chaperone
activity and can potentially be manipulated for translational studies.
Collapse
Affiliation(s)
- Priya R Banerjee
- Department of Chemistry, Life Sciences, University at Albany, State University of New York , Albany, New York 12222, United States
| | | | | | | |
Collapse
|
47
|
Steinman L, Shoenfeld Y. From defining antigens to new therapies in multiple sclerosis: honoring the contributions of Ruth Arnon and Michael Sela. J Autoimmun 2014; 54:1-7. [PMID: 25308417 DOI: 10.1016/j.jaut.2014.08.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2014] [Accepted: 08/12/2014] [Indexed: 12/21/2022]
Abstract
Ruth Arnon and Michael Sela profoundly influenced the development of a model system to test new therapies in multiple sclerosis (MS). Their application of the animal model, known as experimental autoimmune encephalomyelitis (EAE), for the discovery of Copaxone, opened a new path for testing of drug candidates in MS. By measuring clinical, pathologic, and immunologic outcomes, the biological implications of new drugs could be elucidated. Using EAE they established the efficacy of Copaxone as a therapy for preventing and reducing paralysis and inflammation in the central nervous system without massive immune suppression. This had a huge impact on the field of drug discovery for MS. Much like the use of parabiosis to discover soluble factors associated with obesity, or the replica plating system to probe antibiotic resistance in bacteria, the pioneering research on Copaxone using the EAE model, paved the way for the discovery of other therapeutics in MS, including Natalizumab and Fingolimod. Future applications of this approach may well elucidate novel therapies for the neurodegenerative phase of multiple sclerosis associated with disease progression.
Collapse
Affiliation(s)
- Lawrence Steinman
- Beckman Center for Molecular Medicine, Stanford University, Stanford, CA 94305, USA.
| | | |
Collapse
|
48
|
Maarouf CL, Kokjohn TA, Walker DG, Whiteside CM, Kalback WM, Whetzel A, Sue LI, Serrano G, Jacobson SA, Sabbagh MN, Reiman EM, Beach TG, Roher AE. Biochemical assessment of precuneus and posterior cingulate gyrus in the context of brain aging and Alzheimer's disease. PLoS One 2014; 9:e105784. [PMID: 25166759 PMCID: PMC4148328 DOI: 10.1371/journal.pone.0105784] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Accepted: 07/24/2014] [Indexed: 12/12/2022] Open
Abstract
Defining the biochemical alterations that occur in the brain during “normal” aging is an important part of understanding the pathophysiology of neurodegenerative diseases and of distinguishing pathological conditions from aging-associated changes. Three groups were selected based on age and on having no evidence of neurological or significant neurodegenerative disease: 1) young adult individuals, average age 26 years (n = 9); 2) middle-aged subjects, average age 59 years (n = 5); 3) oldest-old individuals, average age 93 years (n = 6). Using ELISA and Western blotting methods, we quantified and compared the levels of several key molecules associated with neurodegenerative disease in the precuneus and posterior cingulate gyrus, two brain regions known to exhibit early imaging alterations during the course of Alzheimer’s disease. Our experiments revealed that the bioindicators of emerging brain pathology remained steady or decreased with advancing age. One exception was S100B, which significantly increased with age. Along the process of aging, neurofibrillary tangle deposition increased, even in the absence of amyloid deposition, suggesting the presence of amyloid plaques is not obligatory for their development and that limited tangle density is a part of normal aging. Our study complements a previous assessment of neuropathology in oldest-old subjects, and within the limitations of the small number of individuals involved in the present investigation, it adds valuable information to the molecular and structural heterogeneity observed along the course of aging and dementia. This work underscores the need to examine through direct observation how the processes of amyloid deposition unfold or change prior to the earliest phases of dementia emergence.
Collapse
Affiliation(s)
- Chera L. Maarouf
- The Longtine Center for Neurodegenerative Biochemistry, Banner Sun Health Research Institute, Sun City, Arizona, United States of America
| | - Tyler A. Kokjohn
- Department of Microbiology, Midwestern University, Glendale, Arizona, United States of America
| | - Douglas G. Walker
- Laboratory of Neuroinflammation, Banner Sun Health Research Institute, Sun City, Arizona, United States of America
| | - Charisse M. Whiteside
- The Longtine Center for Neurodegenerative Biochemistry, Banner Sun Health Research Institute, Sun City, Arizona, United States of America
| | - Walter M. Kalback
- The Longtine Center for Neurodegenerative Biochemistry, Banner Sun Health Research Institute, Sun City, Arizona, United States of America
| | - Alexis Whetzel
- Laboratory of Neuroinflammation, Banner Sun Health Research Institute, Sun City, Arizona, United States of America
| | - Lucia I. Sue
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, Arizona, United States of America
| | - Geidy Serrano
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, Arizona, United States of America
| | - Sandra A. Jacobson
- Cleo Roberts Center for Clinical Research, Banner Sun Health Research Institute, Sun City, Arizona, United States of America
| | - Marwan N. Sabbagh
- Cleo Roberts Center for Clinical Research, Banner Sun Health Research Institute, Sun City, Arizona, United States of America
| | - Eric M. Reiman
- Banner Alzheimer’s Institute, Phoenix, Arizona, United States of America
| | - Thomas G. Beach
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, Arizona, United States of America
| | - Alex E. Roher
- The Longtine Center for Neurodegenerative Biochemistry, Banner Sun Health Research Institute, Sun City, Arizona, United States of America
- * E-mail:
| |
Collapse
|
49
|
Kurnellas MP, Schartner JM, Fathman CG, Jagger A, Steinman L, Rothbard JB. Mechanisms of action of therapeutic amyloidogenic hexapeptides in amelioration of inflammatory brain disease. ACTA ACUST UNITED AC 2014; 211:1847-56. [PMID: 25073790 PMCID: PMC4144739 DOI: 10.1084/jem.20140107] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Amyloid fibrils composed of peptides as short as six amino acids are effective therapeutics for experimental autoimmune encephalomyelitis (EAE). Immunosuppression arises from at least two pathways: (1) expression of type 1 IFN by pDCs, which were induced by neutrophil extracellular traps arising from the endocytosis of the fibrils; and (2) the reduced expression of IFN-γ, TNF, and IL-6. The two independent pathways stimulated by the fibrils can act in concert to be immunosuppressive in Th1 indications, or in opposition, resulting in inflammation when Th17 T lymphocytes are predominant. The generation of type 1 IFN can be minimized by using polar, nonionizable, amyloidogenic peptides, which are effective in both Th1 and Th17 polarized EAE.
Collapse
Affiliation(s)
- Michael P Kurnellas
- Department of Neurology and Neurological Sciences and Department of Medicine, Division of Immunology, Stanford University School of Medicine, Stanford, CA 94305
| | - Jill M Schartner
- Department of Neurology and Neurological Sciences and Department of Medicine, Division of Immunology, Stanford University School of Medicine, Stanford, CA 94305
| | - C Garrison Fathman
- Department of Neurology and Neurological Sciences and Department of Medicine, Division of Immunology, Stanford University School of Medicine, Stanford, CA 94305
| | - Ann Jagger
- Department of Neurology and Neurological Sciences and Department of Medicine, Division of Immunology, Stanford University School of Medicine, Stanford, CA 94305
| | - Lawrence Steinman
- Department of Neurology and Neurological Sciences and Department of Medicine, Division of Immunology, Stanford University School of Medicine, Stanford, CA 94305
| | - Jonathan B Rothbard
- Department of Neurology and Neurological Sciences and Department of Medicine, Division of Immunology, Stanford University School of Medicine, Stanford, CA 94305 Department of Neurology and Neurological Sciences and Department of Medicine, Division of Immunology, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
50
|
Steinman L, Rothbard JB, Kurnellas MP. Janus faces of amyloid proteins in neuroinflammation. J Clin Immunol 2014; 34 Suppl 1:S61-3. [PMID: 24711007 DOI: 10.1007/s10875-014-0034-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Accepted: 03/28/2014] [Indexed: 12/20/2022]
Abstract
Amyloid forming molecules are generally considered harmful. In Alzheimer's Disease two amyloid molecules Aβ A4 and tau vie for consideration as the main pathogenic culprit. But molecules obey the laws of chemistry and defy the way we categorize them as humans with our well-known proclivities to bias in our reasoning. We have been exploring the brains of multiple sclerosis patients to identify molecules that are associated with protection from inflammation and degeneration. In 2001 we noted that aB crystallin (cryab) was the most abundant transcript found in MS lesions, but not in healthy brains. Cryab can reverse paralysis and attenuate inflammation in several models of inflammation including experimental autoimmune encephalomyelitis (EAE), and various models of ischemia. Cryab is an amyloid forming molecule. We have identified a core structure common to many amyloids including amyloid protein Aβ A4, tau, amylin, prion protein, serum amyloid protein P, and cryab. The core hexapeptide structure is highly immune suppressive and can reverse paralysis in EAE when administered systemically. Administration of this amyloid forming hexapeptide quickly lowers inflammatory cytokines in plasma like IL-6 and IL-2. The hexapeptide bind a set of proinflammatory mediators in plasma, including acute phase reactants and complement components. The beneficial properties of amyloid forming hexapeptides provide a potential new therapeutic direction. These experiments indicate that amyloid forming molecules have Janus faces, providing unexpected benefit for neuroinflammatory conditions.
Collapse
Affiliation(s)
- Lawrence Steinman
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA,
| | | | | |
Collapse
|