1
|
Van Alstyne M, Pratt J, Parker R. Diverse influences on tau aggregation and implications for disease progression. Genes Dev 2025; 39:555-581. [PMID: 40113250 PMCID: PMC12047666 DOI: 10.1101/gad.352551.124] [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] [Indexed: 03/22/2025]
Abstract
Tau is an intrinsically disordered protein that accumulates in fibrillar aggregates in neurodegenerative diseases. The misfolding of tau can be understood as an equilibrium between different states and their propensity to form higher-order fibers, which is affected by several factors. First, modulation of the biochemical state of tau due to ionic conditions, post-translational modifications, cofactors, and interacting molecules or assemblies can affect the formation and structure of tau fibrils. Second, cellular processes impact tau aggregation through modulating stability, clearance, disaggregation, and transport. Third, through interactions with glial cells, the neuronal microenvironment can affect intraneuronal conditions with impacts on tau fibrilization and toxicity. Importantly, tau fibrils propagate through the brain via a "prion-like" manner, contributing to disease progression. This review highlights the biochemical and cellular pathways that modulate tau aggregation and discusses implications for pathobiology and tau-directed therapeutic approaches.
Collapse
Affiliation(s)
- Meaghan Van Alstyne
- Department of Biochemistry, University of Colorado Boulder, Boulder, Colorado 80301, USA
- Howard Hughes Medical Institute, University of Colorado Boulder, Boulder, Colorado 80301, USA
| | - James Pratt
- Department of Biochemistry, University of Colorado Boulder, Boulder, Colorado 80301, USA
| | - Roy Parker
- Department of Biochemistry, University of Colorado Boulder, Boulder, Colorado 80301, USA;
- Howard Hughes Medical Institute, University of Colorado Boulder, Boulder, Colorado 80301, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, Colorado 80301, USA
| |
Collapse
|
2
|
Sordu P, Alaylıoğlu M, Samancı B, Bulu E, Güleç ZEK, Bilgiç B, Hanağası HA, Gürvit İH, Ulutin T, Dursun E, Gezen-Ak D. Cerebrospinal fluid HSP90AA1, HSPA4, and STUB1/CHIP levels in Alzheimer's disease, mild cognitive impairment, and frontotemporal dementia. J Alzheimers Dis 2025; 105:481-493. [PMID: 40116694 DOI: 10.1177/13872877251329540] [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] [Indexed: 03/23/2025]
Abstract
BackgroundThe data that we gathered from a protein-protein interaction (PPI) prediction tool, FpClass, and a limited number of studies indicated that the chaperones HSP90AA1, HSPA4, STUB1/CHIP might interact with amyloid-β (Aβ) and/or tau and could subsequently be co-released into the cerebrospinal fluid (CSF). Therefore, we investigated CSF levels of HSP90AA1, HSPA4, and STUB1/CHIP in Alzheimer's disease (AD), Non-AD mild cognitive impairment (Non-AD MCI), and frontotemporal dementia (FTD) cases.MethodsThe CSF levels of HSP90AA1, HSPA4, STUB/CHIP, and core AD biomarkers were determined by ELISA in AD (n = 90), Non-AD MCI (n = 27), FTD (n = 15), and subjective cognitive impairment (SCI) (n = 20) subjects.ResultsHSP90AA1 levels were significantly higher in AD cases compared to the SCI subjects. The CSF levels of STUB1/CHIP were significantly lower in AD, Non-AD MCI and FTD cases compared to the SCI subjects. STUB1/CHIP levels of FTD cases were significantly lower than all other groups. HSPA4 levels was correlated with core AD biomarkers (Aβ 1-42, p-Tau, t-Tau) regardless of disease. Non-APOE ε4 carrier FTD cases also had significantly lower STUB1/CHIP levels than other groups.ConclusionsThe STUB1/CHIP holds promise as a potential biomarker for distinguishing between SCI subjects, AD, and FTD. Furthermore, APOE might serve as an additional discriminatory factor that might be integrated with this chaperone for enhanced discrimination.
Collapse
Affiliation(s)
- Pelin Sordu
- Brain and Neurodegenerative Disorders Research Laboratories, Department of Neuroscience, Institute of Neurological Sciences, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Merve Alaylıoğlu
- Brain and Neurodegenerative Disorders Research Laboratories, Department of Neuroscience, Institute of Neurological Sciences, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Bedia Samancı
- Behavioral Neurology and Movement Disorders Unit, Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Ersel Bulu
- Brain and Neurodegenerative Disorders Research Laboratories, Department of Neuroscience, Institute of Neurological Sciences, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Zeynep Ece Kaya Güleç
- Brain and Neurodegenerative Disorders Research Laboratories, Department of Neuroscience, Institute of Neurological Sciences, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Başar Bilgiç
- Behavioral Neurology and Movement Disorders Unit, Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Haşmet Ayhan Hanağası
- Behavioral Neurology and Movement Disorders Unit, Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - İbrahim Hakan Gürvit
- Behavioral Neurology and Movement Disorders Unit, Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Turgut Ulutin
- Department of Medical Biology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Erdinç Dursun
- Brain and Neurodegenerative Disorders Research Laboratories, Department of Neuroscience, Institute of Neurological Sciences, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Duygu Gezen-Ak
- Brain and Neurodegenerative Disorders Research Laboratories, Department of Neuroscience, Institute of Neurological Sciences, Istanbul University-Cerrahpasa, Istanbul, Turkey
| |
Collapse
|
3
|
Hudák A, Letoha T. Endocytic Pathways Unveil the Role of Syndecans in the Seeding and Spreading of Pathological Protein Aggregates: Insights into Neurodegenerative Disorders. Int J Mol Sci 2025; 26:4037. [PMID: 40362276 PMCID: PMC12071627 DOI: 10.3390/ijms26094037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 04/07/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
Alzheimer's disease and other neurodegenerative disorders are characterized by the accumulation of misfolded proteins, such as amyloid-beta, tau, and α-synuclein, which disrupt neuronal function and contribute to cognitive decline. Heparan sulfate proteoglycans, particularly syndecans, play a pivotal role in the seeding, aggregation, and spreading of toxic protein aggregates through endocytic pathways. Among these, syndecan-3 is particularly critical in regulating the internalization of misfolded proteins, facilitating their propagation in a prion-like manner. This review examines the mechanisms by which syndecans, especially SDC3, contribute to the seeding and spreading of pathological protein aggregates in neurodegenerative diseases. Understanding these endocytic pathways provides valuable insights into the potential of syndecans as biomarkers and therapeutic targets for early intervention in Alzheimer's disease and other related neurodegenerative disorders.
Collapse
Affiliation(s)
- Anett Hudák
- Pharmacoidea Ltd., 6726 Szeged, Hungary;
- Doctoral School of Theoretical Medicine, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary
| | - Tamás Letoha
- Pharmacoidea Ltd., 6726 Szeged, Hungary;
- Doctoral School of Theoretical Medicine, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary
| |
Collapse
|
4
|
Rueter J, Rimbach G, Bilke S, Tholey A, Huebbe P. Readdressing the Localization of Apolipoprotein E (APOE) in Mitochondria-Associated Endoplasmic Reticulum (ER) Membranes (MAMs): An Investigation of the Hepatic Protein-Protein Interactions of APOE with the Mitochondrial Proteins Lon Protease (LONP1), Mitochondrial Import Receptor Subunit TOM40 (TOMM40) and Voltage-Dependent Anion-Selective Channel 1 (VDAC1). Int J Mol Sci 2024; 25:10597. [PMID: 39408926 PMCID: PMC11476584 DOI: 10.3390/ijms251910597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 09/27/2024] [Accepted: 09/29/2024] [Indexed: 10/20/2024] Open
Abstract
As a component of circulating lipoproteins, APOE binds to cell surface receptors mediating lipoprotein metabolism and cholesterol transport. A growing body of evidence, including the identification of a broad variety of cellular proteins interacting with APOE, suggests additional independent functions. Investigating cellular localization and protein-protein interactions in cultured human hepatocytes, we aimed to contribute to the elucidation of hitherto unnoted cellular functions of APOE. We observed a strong accumulation of APOE in MAMs, equally evident for the two major isoforms APOE3 and APOE4. Using mass spectrometry proteome analyses, novel and previously noted APOE interactors were identified, including the mitochondrial proteins TOMM40, LONP1 and VDAC1. All three interactors were present in MAM fractions, which we think initially facilitates interactions with APOE. LONP1 is a protease with chaperone activity, which migrated to MAMs in response to ER stress, displaying a reinforced interaction with APOE. We therefore hypothesize that APOE may help in the unfolded protein response (UPR) by acting as a co-chaperone in cooperation with LONP1 at the interface of mitochondria and ER membranes. The interaction of APOE with the integral proteins TOMM40 and VDAC1 may point to the formation of bridging complexes connecting mitochondria with other organelles.
Collapse
Affiliation(s)
- Johanna Rueter
- Institute of Human Nutrition and Food Science, University of Kiel, Hermann-Rodewald-Strasse 6, 24118 Kiel, Germany; (J.R.); (G.R.)
| | - Gerald Rimbach
- Institute of Human Nutrition and Food Science, University of Kiel, Hermann-Rodewald-Strasse 6, 24118 Kiel, Germany; (J.R.); (G.R.)
| | - Stephanie Bilke
- Institute of Experimental Medicine, University of Kiel, Niemannsweg 11, 24105 Kiel, Germany
| | - Andreas Tholey
- Institute of Experimental Medicine, University of Kiel, Niemannsweg 11, 24105 Kiel, Germany
| | - Patricia Huebbe
- Institute of Human Nutrition and Food Science, University of Kiel, Hermann-Rodewald-Strasse 6, 24118 Kiel, Germany; (J.R.); (G.R.)
| |
Collapse
|
5
|
Singh S, Mahajan M, Kumar D, Singh K, Chowdhary M, Amit. An inclusive study of recent advancements in Alzheimer's disease: A comprehensive review. Neuropeptides 2023; 102:102369. [PMID: 37611472 DOI: 10.1016/j.npep.2023.102369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 08/08/2023] [Accepted: 08/11/2023] [Indexed: 08/25/2023]
Abstract
Alzheimer's disease (AD) has remained elusive in revealing its pathophysiology and mechanism of development. In this review paper, we attempt to highlight several theories that abound about the exact pathway of AD development. The number of cases worldwide has prompted a constant flow of research to detect high-risk patients, slow the progression of the disease and discover improved methods of treatment that may prove effective. We shall focus on the two main classes of drugs that are currently in use; and emerging ones with novel mechanisms that are under development. As of late there has also been increased attention towards factors that were previously thought to be unrelated to AD, such as the gut microbiome, lifestyle habits, and diet. Studies have now shown that all these factors make an impact on AD progression, thus bringing to our attention more areas that could hold the key to combating this disease. This paper covers all the aforementioned factors concisely. We also briefly explore the relationship between mental health and AD, both before and after the diagnosis of the disease.
Collapse
Affiliation(s)
- Sukanya Singh
- Department of Zoology, Hansraj College, University of Delhi, New Delhi, Delhi 110007, India
| | - Mitali Mahajan
- Department of Zoology, Hansraj College, University of Delhi, New Delhi, Delhi 110007, India
| | - Dhawal Kumar
- Department of Zoology, Hansraj College, University of Delhi, New Delhi, Delhi 110007, India
| | - Kunika Singh
- Department of Zoology, Hansraj College, University of Delhi, New Delhi, Delhi 110007, India
| | - Mehvish Chowdhary
- Department of Zoology, Hansraj College, University of Delhi, New Delhi, Delhi 110007, India
| | - Amit
- Department of Zoology, Hansraj College, University of Delhi, New Delhi, Delhi 110007, India.
| |
Collapse
|
6
|
Žerovnik E. Human stefin B: from its structure, folding, and aggregation to its function in health and disease. Front Mol Neurosci 2022; 15:1009976. [PMID: 36340691 PMCID: PMC9634419 DOI: 10.3389/fnmol.2022.1009976] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 09/01/2022] [Indexed: 01/11/2024] Open
Abstract
Mutations in the gene for human stefin B (cystatin B) cause progressive myoclonic epilepsy type 1 (EPM1), a neurodegenerative disorder. The most common change is dodecamer repeats in the promoter region of the gene, though missense and frameshift mutations also appear. Human stefin B primarily acts as a cysteine cathepsin inhibitor, and it also exhibits alternative functions. It plays a protective role against oxidative stress, likely via reducing mitochondrial damage and thus generating fewer mitochondrial reactive oxygen species (ROS). Accordingly, lack of stefin B results in increased inflammation and NLRP3 inflammasome activation, producing more ROS. The protein is cytosolic but also has an important role in the nucleus, where it prevents cleavage of the N terminal part of histone 3 by inhibiting cathepsins L and B and thus regulates transcription and cell cycle. Furthermore, it has been shown that stefin B is oligomeric in cells and that it has a specific role in the physiology of the synapse and in vesicular transport. On the basis of my research team's data on the structure, folding, and aggregation of stefin B, we have proposed that it might regulate proteostasis, possessing a chaperone-like function. In this review, I synthesize these observations and derive some conclusions on possible sources of EPM1 pathology. The interaction partners of stefin B and other gene mutations leading to EPM1-like pathology are discussed and common pathways are pinpointed.
Collapse
Affiliation(s)
- Eva Žerovnik
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, Ljubljana, Slovenia
- Jožef Stefan International Postgraduate School, Ljubljana, Slovenia
| |
Collapse
|
7
|
Picone P, Sanfilippo T, Vasto S, Baldassano S, Guggino R, Nuzzo D, Bulone D, San Biagio PL, Muscolino E, Monastero R, Dispenza C, Giacomazza D. From Small Peptides to Large Proteins against Alzheimer’sDisease. Biomolecules 2022; 12:biom12101344. [PMID: 36291553 PMCID: PMC9599460 DOI: 10.3390/biom12101344] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 09/15/2022] [Accepted: 09/17/2022] [Indexed: 11/16/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common neurodegenerative disorder in the elderly. The two cardinal neuropathological hallmarks of AD are the senile plaques, which are extracellular deposits mainly constituted by beta-amyloids, and neurofibrillary tangles formed by abnormally phosphorylated Tau (p-Tau) located in the cytoplasm of neurons. Although the research has made relevant progress in the management of the disease, the treatment is still lacking. Only symptomatic medications exist for the disease, and, in the meantime, laboratories worldwide are investigating disease-modifying treatments for AD. In the present review, results centered on the use of peptides of different sizes involved in AD are presented.
Collapse
Affiliation(s)
- Pasquale Picone
- Istituto per la Ricerca e l’Innovazione Biomedica, Consiglio Nazionale delle Ricerche, Via U. La Malfa 153, 90146 Palermo, Italy
- Dipartmento of Scienze Biologiche, Chimiche, Farmaceutiche e Tecnologiche (STEBICEF), University of Palermo, 90128 Palermo, Italy
| | - Tiziana Sanfilippo
- Ambulatorio di Nutrizione Clinica ASP Palermo, Via G. Cusmano 24, 90141 Palermo, Italy
- Anestesia e Rianimazione, Presidio Ospedaliero “S. Cimino”, 90141 Termini Imerese, Italy
| | - Sonya Vasto
- Dipartmento of Scienze Biologiche, Chimiche, Farmaceutiche e Tecnologiche (STEBICEF), University of Palermo, 90128 Palermo, Italy
- Istituti Euro-Mediterranei di Scienza e Tecnologia (IEMEST), Via M. Miraglia 20, 90139 Palermo, Italy
| | - Sara Baldassano
- Dipartmento of Scienze Biologiche, Chimiche, Farmaceutiche e Tecnologiche (STEBICEF), University of Palermo, 90128 Palermo, Italy
| | - Rossella Guggino
- Ambulatorio di Nutrizione Clinica ASP Palermo, Via G. Cusmano 24, 90141 Palermo, Italy
- Anestesia e Rianimazione, Presidio Ospedaliero “S. Cimino”, 90141 Termini Imerese, Italy
| | - Domenico Nuzzo
- Istituto per la Ricerca e l’Innovazione Biomedica, Consiglio Nazionale delle Ricerche, Via U. La Malfa 153, 90146 Palermo, Italy
- Dipartmento of Scienze Biologiche, Chimiche, Farmaceutiche e Tecnologiche (STEBICEF), University of Palermo, 90128 Palermo, Italy
- Correspondence: (D.N.); (D.G.)
| | - Donatella Bulone
- Istituto di Biofisica, Consiglio Nazionale delle Ricerche, Via U. La Malfa 153, 90146 Palermo, Italy
| | - Pier Luigi San Biagio
- Istituto di Biofisica, Consiglio Nazionale delle Ricerche, Via U. La Malfa 153, 90146 Palermo, Italy
| | - Emanuela Muscolino
- Dipartimento di Ingegneria, Università degli Studi di Palermo, Viale delle Scienze, Bldg 6, 90128 Palermo, Italy
| | - Roberto Monastero
- Dipartimento di Biomedicina, Neuroscienze e Diagnostica Avanzata, Università degli Studi di Palermo, Via del Vespro 129, 90127 Palermo, Italy
| | - Clelia Dispenza
- Dipartimento di Ingegneria, Università degli Studi di Palermo, Viale delle Scienze, Bldg 6, 90128 Palermo, Italy
| | - Daniela Giacomazza
- Istituto di Biofisica, Consiglio Nazionale delle Ricerche, Via U. La Malfa 153, 90146 Palermo, Italy
- Correspondence: (D.N.); (D.G.)
| |
Collapse
|
8
|
Lopez-Toledo G, Silva-Lucero MDC, Herrera-Díaz J, García DE, Arias-Montaño JA, Cardenas-Aguayo MDC. Patient-Derived Fibroblasts With Presenilin-1 Mutations, That Model Aspects of Alzheimer’s Disease Pathology, Constitute a Potential Object for Early Diagnosis. Front Aging Neurosci 2022; 14:921573. [PMID: 35847683 PMCID: PMC9283986 DOI: 10.3389/fnagi.2022.921573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 06/13/2022] [Indexed: 11/13/2022] Open
Abstract
Alzheimer’s disease (AD), a neurodegenerative disorder that can occur in middle or old age, is characterized by memory loss, a continuous decline in thinking, behavioral and social skills that affect the ability of an individual to function independently. It is divided into sporadic and familial subtypes. Early-onset familial AD (FAD) is linked to mutations in genes coding for the amyloid-β protein precursor (AβPP), presenilin 1 (PS1), and presenilin 2 (PS2), which lead to alterations in AβPP processing, generation of the Amyloid-β peptide and hyperphosphorylation of tau protein. Identification of early biomarkers for AD diagnosis represents a challenge, and it has been suggested that molecular changes in neurodegenerative pathways identified in the brain of AD patients can be detected in peripheral non-neural cells derived from familial or sporadic AD patients. In the present study, we determined the protein expression, the proteomic and in silico characterization of skin fibroblasts from FAD patients with PS1 mutations (M146L or A246E) or from healthy individuals. Our results shown that fibroblasts from AD patients had increased expression of the autophagy markers LC3II, LAMP2 and Cathepsin D, a significant increase in total GSK3, phosphorylated ERK1/2 (Thr202/Tyr204) and phosphorylated tau (Thr231, Ser396, and Ser404), but no difference in the phosphorylation of Akt (Ser473) or the α (Ser21) and β (Ser9) GSK3 isoforms, highlighting the relevant role of abnormal protein post-translational modifications in age-related neurodegenerative diseases, such as AD. Both 2-DE gels and mass spectrometry showed significant differences in the expression of the signaling pathways associated with protein folding and the autophagic pathway mediated by chaperones with the expression of HSPA5, HSPE1, HSPD1, HSP90AA1, and HSPE1 and reticular stress in the FAD samples. Furthermore, expression of the heat shock proteins HSP90 and HSP70 was significantly higher in the cells from AD patients as confirmed by Western blot. Taken together our results indicate that fibroblasts from patients with FAD-PS1 present alterations in signaling pathways related to cellular stress, autophagy, lysosomes, and tau phosphorylation. Fibroblasts can therefore be useful in modeling pathways related to neurodegeneration, as well as for the identification of early AD biomarkers.
Collapse
Affiliation(s)
- Gustavo Lopez-Toledo
- Laboratory of Cellular Reprogramming, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav-IPN), Mexico City, Mexico
| | - Maria-del-Carmen Silva-Lucero
- Laboratory of Cellular Reprogramming, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Jorge Herrera-Díaz
- Unidad de Servicios de Apoyo a la Investigación y a la Industria, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - David-Erasmo García
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - José-Antonio Arias-Montaño
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav-IPN), Mexico City, Mexico
| | - Maria-del-Carmen Cardenas-Aguayo
- Laboratory of Cellular Reprogramming, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
- *Correspondence: Maria-del-Carmen Cardenas-Aguayo,
| |
Collapse
|
9
|
Gualtieri CT. Genomic Variation, Evolvability, and the Paradox of Mental Illness. Front Psychiatry 2021; 11:593233. [PMID: 33551865 PMCID: PMC7859268 DOI: 10.3389/fpsyt.2020.593233] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 11/27/2020] [Indexed: 12/30/2022] Open
Abstract
Twentieth-century genetics was hard put to explain the irregular behavior of neuropsychiatric disorders. Autism and schizophrenia defy a principle of natural selection; they are highly heritable but associated with low reproductive success. Nevertheless, they persist. The genetic origins of such conditions are confounded by the problem of variable expression, that is, when a given genetic aberration can lead to any one of several distinct disorders. Also, autism and schizophrenia occur on a spectrum of severity, from mild and subclinical cases to the overt and disabling. Such irregularities reflect the problem of missing heritability; although hundreds of genes may be associated with autism or schizophrenia, together they account for only a small proportion of cases. Techniques for higher resolution, genomewide analysis have begun to illuminate the irregular and unpredictable behavior of the human genome. Thus, the origins of neuropsychiatric disorders in particular and complex disease in general have been illuminated. The human genome is characterized by a high degree of structural and behavioral variability: DNA content variation, epistasis, stochasticity in gene expression, and epigenetic changes. These elements have grown more complex as evolution scaled the phylogenetic tree. They are especially pertinent to brain development and function. Genomic variability is a window on the origins of complex disease, neuropsychiatric disorders, and neurodevelopmental disorders in particular. Genomic variability, as it happens, is also the fuel of evolvability. The genomic events that presided over the evolution of the primate and hominid lineages are over-represented in patients with autism and schizophrenia, as well as intellectual disability and epilepsy. That the special qualities of the human genome that drove evolution might, in some way, contribute to neuropsychiatric disorders is a matter of no little interest.
Collapse
|
10
|
Owens LV, Benedetto A, Dawson N, Gaffney CJ, Parkin ET. Gene therapy-mediated enhancement of protective protein expression for the treatment of Alzheimer's disease. Brain Res 2021; 1753:147264. [PMID: 33422539 DOI: 10.1016/j.brainres.2020.147264] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 11/22/2020] [Accepted: 12/20/2020] [Indexed: 12/30/2022]
Abstract
Alzheimer's disease (AD) is the leading form of dementia but lacks curative treatments. Current understanding of AD aetiology attributes the development of the disease to the misfolding of two proteins; amyloid-β (Aβ) and hyperphosphorylated tau, with their pathological accumulation leading to concomitant oxidative stress, neuroinflammation, and neuronal death. These processes are regulated at multiple levels to maintain homeostasis and avert disease. However, many of the relevant regulatory proteins appear to be downregulated in the AD-afflicted brain. Enhancement/restoration of these 'protective' proteins, therefore, represents an attractive therapeutic avenue. Gene therapy is a desirable means of achieving this because it is not associated with the side-effects linked to systemic protein administration, and sustained protein expression virtually eliminates compliance issues. The current article represents a focused and succinct review of the better established 'protective' protein targets for gene therapy enhancement/restoration rather than being designed as an exhaustive review incorporating less validated protein subjects. In addition, we will discuss how the risks associated with uncontrolled or irreversible gene expression might be mitigated through combining neuronal-specific promoters, inducible expression systems and localised injections. Whilst many of the gene therapy targets reviewed herein are yet to enter clinical trials, preclinical testing has thus far demonstrated encouraging potential for the gene therapy-based treatment of AD.
Collapse
Affiliation(s)
- Lauren V Owens
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster LA1 4YG, UK
| | - Alexandre Benedetto
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster LA1 4YG, UK
| | - Neil Dawson
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster LA1 4YG, UK
| | - Christopher J Gaffney
- Lancaster Medical School, Faculty of Health and Medicine, Lancaster University, Lancaster LA1 4YG, UK
| | - Edward T Parkin
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster LA1 4YG, UK.
| |
Collapse
|
11
|
Su Q, Li T, He PF, Lu XC, Yu Q, Gao QC, Wang ZJ, Wu MN, Yang D, Qi JS. Trichostatin A ameliorates Alzheimer's disease-related pathology and cognitive deficits by increasing albumin expression and Aβ clearance in APP/PS1 mice. Alzheimers Res Ther 2021; 13:7. [PMID: 33397436 PMCID: PMC7784383 DOI: 10.1186/s13195-020-00746-8] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 12/08/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is an intractable neurodegenerative disorder in the elderly population, currently lacking a cure. Trichostatin A (TSA), a histone deacetylase inhibitor, showed some neuroprotective roles, but its pathology-improvement effects in AD are still uncertain, and the underlying mechanisms remain to be elucidated. The present study aims to examine the anti-AD effects of TSA, particularly investigating its underlying cellular and molecular mechanisms. METHODS Novel object recognition and Morris water maze tests were used to evaluate the memory-ameliorating effects of TSA in APP/PS1 transgenic mice. Immunofluorescence, Western blotting, Simoa assay, and transmission electron microscopy were utilized to examine the pathology-improvement effects of TSA. Microglial activity was assessed by Western blotting and transwell migration assay. Protein-protein interactions were analyzed by co-immunoprecipitation and LC-MS/MS. RESULTS TSA treatment not only reduced amyloid β (Aβ) plaques and soluble Aβ oligomers in the brain, but also effectively improved learning and memory behaviors of APP/PS1 mice. In vitro study suggested that the improvement of Aβ pathology by TSA was attributed to the enhancement of Aβ clearance, mainly by the phagocytosis of microglia, and the endocytosis and transport of microvascular endothelial cells. Notably, a meaningful discovery in the study was that TSA dramatically upregulated the expression level of albumin in cell culture, by which TSA inhibited Aβ aggregation and promoted the phagocytosis of Aβ oligomers. CONCLUSIONS These findings provide a new insight into the pathogenesis of AD and suggest TSA as a novel promising candidate for the AD treatment.
Collapse
Affiliation(s)
- Qiang Su
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Tian Li
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Pei-Feng He
- Institute of Medical Data Sciences and School of Management, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.
| | - Xue-Chun Lu
- Department of Hematology, the Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Qi Yu
- Institute of Medical Data Sciences and School of Management, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Qi-Chao Gao
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Zhao-Jun Wang
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Mei-Na Wu
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Dan Yang
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Jin-Shun Qi
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.
| |
Collapse
|
12
|
Noorani AA, Yamashita H, Gao Y, Islam S, Sun Y, Nakamura T, Enomoto H, Zou K, Michikawa M. High temperature promotes amyloid β-protein production and γ-secretase complex formation via Hsp90. J Biol Chem 2020; 295:18010-18022. [PMID: 33067321 PMCID: PMC7939388 DOI: 10.1074/jbc.ra120.013845] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 09/30/2020] [Indexed: 12/19/2022] Open
Abstract
Alzheimer's disease (AD) is characterized by neuronal loss and accumulation of β-amyloid-protein (Aβ) in the brain parenchyma. Sleep impairment is associated with AD and affects about 25-40% of patients in the mild-to-moderate stages of the disease. Sleep deprivation leads to increased Aβ production; however, its mechanism remains largely unknown. We hypothesized that the increase in core body temperature induced by sleep deprivation may promote Aβ production. Here, we report temperature-dependent regulation of Aβ production. We found that an increase in temperature, from 37 °C to 39 °C, significantly increased Aβ production in amyloid precursor protein-overexpressing cells. We also found that high temperature (39 °C) significantly increased the expression levels of heat shock protein 90 (Hsp90) and the C-terminal fragment of presenilin 1 (PS1-CTF) and promoted γ-secretase complex formation. Interestingly, Hsp90 was associated with the components of the premature γ-secretase complex, anterior pharynx-defective-1 (APH-1), and nicastrin (NCT) but was not associated with PS1-CTF or presenilin enhancer-2. Hsp90 knockdown abolished the increased level of Aβ production and the increased formation of the γ-secretase complex at high temperature in culture. Furthermore, with in vivo experiments, we observed increases in the levels of Hsp90, PS1-CTF, NCT, and the γ-secretase complex in the cortex of mice housed at higher room temperature (30 °C) compared with those housed at standard room temperature (23 °C). Our results suggest that high temperature regulates Aβ production by modulating γ-secretase complex formation through the binding of Hsp90 to NCT/APH-1.
Collapse
Affiliation(s)
- Arshad Ali Noorani
- Department of Biochemistry, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Hitoshi Yamashita
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai, Japan
| | - Yuan Gao
- Department of Biochemistry, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Sadequl Islam
- Department of Biochemistry, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Yang Sun
- Department of Biochemistry, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Tomohisa Nakamura
- Department of Biochemistry, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Hiroyuki Enomoto
- Department of Biochemistry, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Kun Zou
- Department of Biochemistry, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan.
| | - Makoto Michikawa
- Department of Biochemistry, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan.
| |
Collapse
|
13
|
Breijyeh Z, Karaman R. Comprehensive Review on Alzheimer's Disease: Causes and Treatment. Molecules 2020; 25:E5789. [PMID: 33302541 PMCID: PMC7764106 DOI: 10.3390/molecules25245789] [Citation(s) in RCA: 1230] [Impact Index Per Article: 246.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/03/2020] [Accepted: 12/06/2020] [Indexed: 12/11/2022] Open
Abstract
Alzheimer's disease (AD) is a disorder that causes degeneration of the cells in the brain and it is the main cause of dementia, which is characterized by a decline in thinking and independence in personal daily activities. AD is considered a multifactorial disease: two main hypotheses were proposed as a cause for AD, cholinergic and amyloid hypotheses. Additionally, several risk factors such as increasing age, genetic factors, head injuries, vascular diseases, infections, and environmental factors play a role in the disease. Currently, there are only two classes of approved drugs to treat AD, including inhibitors to cholinesterase enzyme and antagonists to N-methyl d-aspartate (NMDA), which are effective only in treating the symptoms of AD, but do not cure or prevent the disease. Nowadays, the research is focusing on understanding AD pathology by targeting several mechanisms, such as abnormal tau protein metabolism, β-amyloid, inflammatory response, and cholinergic and free radical damage, aiming to develop successful treatments that are capable of stopping or modifying the course of AD. This review discusses currently available drugs and future theories for the development of new therapies for AD, such as disease-modifying therapeutics (DMT), chaperones, and natural compounds.
Collapse
Affiliation(s)
| | - Rafik Karaman
- Pharmaceutical Sciences Department, Faculty of Pharmacy, Al-Quds University, Jerusalem 20002, Palestine;
| |
Collapse
|
14
|
Blood-brain barrier integrity in the pathogenesis of Alzheimer's disease. Front Neuroendocrinol 2020; 59:100857. [PMID: 32781194 DOI: 10.1016/j.yfrne.2020.100857] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 07/27/2020] [Accepted: 07/29/2020] [Indexed: 02/06/2023]
Abstract
The blood-brain barrier (BBB) tightly controls the molecular exchange between the brain parenchyma and blood. Accumulated evidence from transgenic animal Alzheimer's disease (AD) models and human AD patients have demonstrated that BBB dysfunction is a major player in AD pathology. In this review, we discuss the role of the BBB in maintaining brain integrity and how this is mediated by crosstalk between BBB-associated cells within the neurovascular unit (NVU). We then discuss the role of the NVU, in particular its endothelial cell, pericyte, and glial cell constituents, in AD pathogenesis. The effect of substances released by the neuroendocrine system in modulating BBB function and AD pathogenesis is also discussed. We perform a systematic review of currently available AD treatments specifically targeting pericytes and BBB glial cells. In summary, this review provides a comprehensive overview of BBB dysfunction in AD and a new perspective on the development of therapeutics for AD.
Collapse
|
15
|
Lackie RE, Marques-Lopes J, Ostapchenko VG, Good S, Choy WY, van Oosten-Hawle P, Pasternak SH, Prado VF, Prado MAM. Increased levels of Stress-inducible phosphoprotein-1 accelerates amyloid-β deposition in a mouse model of Alzheimer's disease. Acta Neuropathol Commun 2020; 8:143. [PMID: 32825842 PMCID: PMC7441634 DOI: 10.1186/s40478-020-01013-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 08/01/2020] [Indexed: 02/08/2023] Open
Abstract
Molecular chaperones and co-chaperones, which are part of the protein quality control machinery, have been shown to regulate distinct aspects of Alzheimer's Disease (AD) pathology in multiple ways. Notably, the co-chaperone STI1, which presents increased levels in AD, can protect mammalian neurons from amyloid-β toxicity in vitro and reduced STI1 levels worsen Aβ toxicity in C. elegans. However, whether increased STI1 levels can protect neurons in vivo remains unknown. We determined that overexpression of STI1 and/or Hsp90 protected C. elegans expressing Aβ(3-42) against Aβ-mediated paralysis. Mammalian neurons were also protected by elevated levels of endogenous STI1 in vitro, and this effect was mainly due to extracellular STI1. Surprisingly, in the 5xFAD mouse model of AD, by overexpressing STI1, we find increased amyloid burden, which amplifies neurotoxicity and worsens spatial memory deficits in these mutants. Increased levels of STI1 disturbed the expression of Aβ-regulating enzymes (BACE1 and MMP-2), suggesting potential mechanisms by which amyloid burden is increased in mice. Notably, we observed that STI1 accumulates in dense-core AD plaques in both 5xFAD mice and human brain tissue. Our findings suggest that elevated levels of STI1 contribute to Aβ accumulation, and that STI1 is deposited in AD plaques in mice and humans. We conclude that despite the protective effects of STI1 in C. elegans and in mammalian cultured neurons, in vivo, the predominant effect of elevated STI1 is deleterious in AD.
Collapse
Affiliation(s)
- Rachel E Lackie
- Robarts Research Institute, The University of Western Ontario, 1151 Richmond St. N., London, Ontario, N6A 5B7, Canada
- Program in Neuroscience, The University of Western Ontario, 1151 Richmond St, London, N6A 3K7, Canada
| | - Jose Marques-Lopes
- Robarts Research Institute, The University of Western Ontario, 1151 Richmond St. N., London, Ontario, N6A 5B7, Canada
| | - Valeriy G Ostapchenko
- Robarts Research Institute, The University of Western Ontario, 1151 Richmond St. N., London, Ontario, N6A 5B7, Canada
| | - Sarah Good
- School of Molecular and Cell Biology and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Wing-Yiu Choy
- Department of Biochemistry, Schulich School of Medicine & Dentistry, The University of Western Ontario, Medical Sciences Building, 1151 Richmond St. N, London, N6A 5B7, Canada
| | - Patricija van Oosten-Hawle
- School of Molecular and Cell Biology and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Stephen H Pasternak
- Robarts Research Institute, The University of Western Ontario, 1151 Richmond St. N., London, Ontario, N6A 5B7, Canada
- St. Joseph's Health Care London-Parkwood Institute, St. Joseph's Hospital, 268 Grosvenor St Room A1-015, London, N6A 4V2, Canada
- Department of Clinical Neurological Sciences, Schulich School of Medicine & Dentistry, 1151 Richmond St, London, N6A 3K7, Canada
| | - Vania F Prado
- Robarts Research Institute, The University of Western Ontario, 1151 Richmond St. N., London, Ontario, N6A 5B7, Canada.
- Program in Neuroscience, The University of Western Ontario, 1151 Richmond St, London, N6A 3K7, Canada.
- Department of Anatomy & Cell Biology, The University of Western Ontario, 1151 Richmond St, London, N6A 3K7, Canada.
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario, 1151 Richmond St, London, N6A 3K7, Ontario, Canada.
| | - Marco A M Prado
- Robarts Research Institute, The University of Western Ontario, 1151 Richmond St. N., London, Ontario, N6A 5B7, Canada.
- Program in Neuroscience, The University of Western Ontario, 1151 Richmond St, London, N6A 3K7, Canada.
- Department of Anatomy & Cell Biology, The University of Western Ontario, 1151 Richmond St, London, N6A 3K7, Canada.
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario, 1151 Richmond St, London, N6A 3K7, Ontario, Canada.
| |
Collapse
|
16
|
Inflammation: major denominator of obesity, Type 2 diabetes and Alzheimer's disease-like pathology? Clin Sci (Lond) 2020; 134:547-570. [PMID: 32167154 DOI: 10.1042/cs20191313] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/27/2020] [Accepted: 02/28/2020] [Indexed: 02/08/2023]
Abstract
Adipose tissue is an active metabolic organ that contributes to processes such as energy storage and utilization and to the production of a number of metabolic agents, such as adipokines, which play a role in inflammation. In this review, we try to elucidate the connections between peripheral inflammation at obesity and Type 2 diabetes and the central inflammatory process. Multiple lines of evidence highlight the importance of peripheral inflammation and its link to neuroinflammation, which can lead to neurodegenerative diseases such as dementia, Alzheimer's disease (AD) and Parkinson's disease. In addition to the accumulation of misfolded amyloid beta (Aβ) peptide and the formation of the neurofibrillary tangles of hyperphosphorylated tau protein in the brain, activated microglia and reactive astrocytes are the main indicators of AD progression. They were found close to Aβ plaques in the brains of both AD patients and rodent models of Alzheimer's disease-like pathology. Cytokines are key players in pro- and anti-inflammatory processes and are also produced by microglia and astrocytes. The interplay of seemingly unrelated pathways between the periphery and the brain could, in fact, have a common denominator, with inflammation in general being a key factor affecting neuronal processes in the brain. An increased amount of white adipose tissue throughout the body seems to be an important player in pro-inflammatory processes. Nevertheless, other important factors should be studied to elucidate the pathological processes of and the relationship among obesity, Type 2 diabetes and neurodegenerative diseases.
Collapse
|
17
|
Turkez H, Cacciatore I, Arslan ME, Fornasari E, Marinelli L, Di Stefano A, Mardinoglu A. Histidyl-Proline Diketopiperazine Isomers as Multipotent Anti-Alzheimer Drug Candidates. Biomolecules 2020; 10:biom10050737. [PMID: 32397415 PMCID: PMC7277666 DOI: 10.3390/biom10050737] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 04/26/2020] [Accepted: 04/29/2020] [Indexed: 12/13/2022] Open
Abstract
Cyclic dipeptides administered by both parenteral and oral routes are suggested as promising candidates for the treatment of neurodegeneration-related pathologies. In this study, we tested Cyclo (His-Pro) isomers (cHP1-4) for their anti-Alzheimer potential using a differentiated human neuroblastoma cell line (SH-SY5Y) as an Alzheimer’s disease (AD) experimental model. The SH-SY5Y cell line was differentiated by the application of all-trans retinoic acid (RA) to obtain mature neuron-like cells. Amyloid-beta 1-42 (Aβ1-42) peptides, the main effector in AD, were administered to the differentiated cell cultures to constitute the in vitro disease model. Next, we performed cell viability analyses 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and lactate dehydrogenase (LDH) release assays) to investigate the neuroprotective concentrations of cyclodipeptides using the in vitro AD model. We evaluated acetylcholinesterase (AChE), α- and β-secretase activities (TACE and BACE1), antioxidant potency, and apoptotic/necrotic properties and performed global gene expression analysis to understand the main mechanism behind the neuroprotective features of cHP1-4. Moreover, we conducted sister chromatid exchange (SCE), micronucleus (MN), and 8-hydroxy-2′-deoxyguanosine (8-OHdG) analyses to evaluate the genotoxic damage potential after applications with cHP1-4 on cultured human lymphocytes. Our results revealed that cHP1-4 isomers provide a different degree of neuroprotection against Aβ1-42-induced cell death on the in vitro AD model. The applications with cHP1-4 isomers altered the activity of AChE but not the activity of TACE and BACE1. Our analysis indicated that the cHP1-4 increased the total antioxidant capacity without altering total oxidative status levels in the cellular AD model and that cHP1-4 modulated the alterations of gene expressions by Aβ1-42 exposure. We also observed that cHP1-4 exhibited noncytotoxic and non-genotoxic features in cultured human whole blood cells. In conclusion, cHP1-4 isomers, especially cHP4, have been explored as novel promising therapeutics against AD.
Collapse
Affiliation(s)
- Hasan Turkez
- Department of Medical Biology, Faculty of Medicine, Atatürk University, 25240 Erzurum, Turkey
- Correspondence: (H.T.); (A.M.)
| | - Ivana Cacciatore
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, via dei Vestini 31, 66100 Chieti Scalo (CH), Italy; (I.C.); (E.F.); (L.M.); (A.D.S.)
| | - Mehmet Enes Arslan
- Department of Molecular Biology and Genetics, Faculty of Science, Erzurum Technical University, 25050 Erzurum, Turkey;
| | - Erika Fornasari
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, via dei Vestini 31, 66100 Chieti Scalo (CH), Italy; (I.C.); (E.F.); (L.M.); (A.D.S.)
| | - Lisa Marinelli
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, via dei Vestini 31, 66100 Chieti Scalo (CH), Italy; (I.C.); (E.F.); (L.M.); (A.D.S.)
| | - Antonio Di Stefano
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, via dei Vestini 31, 66100 Chieti Scalo (CH), Italy; (I.C.); (E.F.); (L.M.); (A.D.S.)
| | - Adil Mardinoglu
- Science for Life Laboratory, KTH-Royal Institute of Technology, SE-17121 Stockholm, Sweden
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London SE1 9RT, UK
- Correspondence: (H.T.); (A.M.)
| |
Collapse
|
18
|
Sharma S, Saini R, Sharma P, Saini A, Nehru B. Maintenance of Amyloid-beta Homeostasis by Carbenoxolone Post Aβ-42 Oligomer Injection in Rat Brain. Neuroscience 2020; 431:86-102. [DOI: 10.1016/j.neuroscience.2020.02.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 02/03/2020] [Accepted: 02/04/2020] [Indexed: 10/25/2022]
|
19
|
Al Mamun A, Uddin MS, Kabir MT, Khanum S, Sarwar MS, Mathew B, Rauf A, Ahmed M, Ashraf GM. Exploring the Promise of Targeting Ubiquitin-Proteasome System to Combat Alzheimer’s Disease. Neurotox Res 2020; 38:8-17. [DOI: 10.1007/s12640-020-00185-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 02/15/2020] [Accepted: 02/18/2020] [Indexed: 12/13/2022]
|
20
|
Sahoo BR, Bekier ME, Liu Z, Kocman V, Stoddard AK, Anantharamaiah GM, Nowick J, Fierke CA, Wang Y, Ramamoorthy A. Structural Interaction of Apolipoprotein A-I Mimetic Peptide with Amyloid-β Generates Toxic Hetero-oligomers. J Mol Biol 2019; 432:1020-1034. [PMID: 31866295 DOI: 10.1016/j.jmb.2019.12.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 11/27/2019] [Accepted: 12/01/2019] [Indexed: 01/21/2023]
Abstract
Apolipoproteins are involved in pathological conditions of Alzheimer's disease (AD), and it has been reported that truncated apolipoprotein fragments and β-amyloid (Aβ) peptides coexist as neurotoxic heteromers within the plaques. Therefore, it is important to investigate these complexes at the molecular level to better understand their properties and roles in the pathology of AD. Here, we present a mechanistic insight into such heteromerization using a structurally homologue apolipoprotein fragment of apoA-I (4F) complexed with Aβ(M1-42) and characterize their toxicity. The 4F peptide slows down the aggregation kinetics of Aβ(M1-42) by constraining its structural plasticity. NMR and CD experiments identified 4F-Aβ(M1-42) heteromers comprised of unstructured Aβ(M1-42) and helical 4F. A uniform two-fold reduction in 15N/1H NMR signal intensities of Aβ(M1-42) with no observable chemical shift perturbation indicated the formation of a large complex, which was further confirmed by diffusion NMR experiments. Microsecond-scale atomistic molecular dynamics simulations showed that 4F interaction with Aβ(M1-42) is electrostatically driven and induces unfolding of Aβ(M1-42). Neurotoxicity profiling of Aβ(M1-42) complexed with 4F confirms a significant reduction in cell viability and neurite growth. Thus, the molecular architecture of heteromerization between 4F and Aβ(M1-42) discovered in this study provides evidence toward our understanding of the role of apolipoproteins or their truncated fragments in exacerbating AD pathology.
Collapse
Affiliation(s)
- Bikash Ranjan Sahoo
- Biophysics and Department of Chemistry, Biomedical Engineering, Macromolecular Science and Engineering, University of Michigan, Ann Arbor, MI, 48109-1055, USA
| | - Michael E Bekier
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109-1085, USA
| | - Zichen Liu
- Biophysics and Department of Chemistry, Biomedical Engineering, Macromolecular Science and Engineering, University of Michigan, Ann Arbor, MI, 48109-1055, USA
| | - Vojc Kocman
- Biophysics and Department of Chemistry, Biomedical Engineering, Macromolecular Science and Engineering, University of Michigan, Ann Arbor, MI, 48109-1055, USA
| | - Andrea K Stoddard
- Biophysics and Department of Chemistry, Biomedical Engineering, Macromolecular Science and Engineering, University of Michigan, Ann Arbor, MI, 48109-1055, USA
| | - G M Anantharamaiah
- Department of Medicine, University of Alabama at Birmingham Medical Center, Birmingham, AL, 35294, USA
| | - James Nowick
- Department of Chemistry, University of California-Irvine, Irvine, CA, 92697-2025, USA
| | - Carol A Fierke
- Department of Chemistry, University of Texas A&M, College Station, TX, 77843-3255, USA
| | - Yanzhuang Wang
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109-1085, USA
| | - Ayyalusamy Ramamoorthy
- Biophysics and Department of Chemistry, Biomedical Engineering, Macromolecular Science and Engineering, University of Michigan, Ann Arbor, MI, 48109-1055, USA.
| |
Collapse
|
21
|
Vilasi S, Carrotta R, Ricci C, Rappa GC, Librizzi F, Martorana V, Ortore MG, Mangione MR. Inhibition of Aβ 1-42 Fibrillation by Chaperonins: Human Hsp60 Is a Stronger Inhibitor than Its Bacterial Homologue GroEL. ACS Chem Neurosci 2019; 10:3565-3574. [PMID: 31298838 DOI: 10.1021/acschemneuro.9b00183] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Alzheimer's disease is a chronic neurodegenerative disease characterized by the accumulation of pathological aggregates of amyloid beta peptide. Many efforts have been focused on understanding peptide aggregation pathways and on identification of molecules able to inhibit aggregation in order to find an effective therapy. As a result, interest in neuroprotective proteins, such as molecular chaperones, has increased as their normal function is to assist in protein folding or to facilitate the disaggregation and/or clearance of abnormal aggregate proteins. Using biophysical techniques, we evaluated the effects of two chaperones, human Hsp60 and bacterial GroEL, on the fibrillogenesis of Aβ1-42. Both chaperonins interfere with Aβ1-42 aggregation, but the effect of Hsp60 is more significant and correlates with its more pronounced flexibility and stronger interaction with ANS, an indicator of hydrophobic regions. Dose-dependent ThT fluorescence kinetics and SAXS experiments reveal that Hsp60 does not change the nature of the molecular processes stochastically leading to the formation of seeds, but strongly delays them by recognition of hydrophobic sites of some peptide species crucial for triggering amyloid formation. Hsp60 reduces the initial chaotic heterogeneity of Aβ1-42 sample at high concentration regimes. The understanding of chaperone action in counteracting pathological aggregation could be a starting point for potential new therapeutic strategies against neurodegenerative diseases.
Collapse
Affiliation(s)
- Silvia Vilasi
- Institute of Biophysics, National Research Council, Palermo 90146, Italy
| | - Rita Carrotta
- Institute of Biophysics, National Research Council, Palermo 90146, Italy
| | - Caterina Ricci
- Department of Life and Environmental Sciences, Marche Polytechnic University, Ancona 60131, Italy
| | | | - Fabio Librizzi
- Institute of Biophysics, National Research Council, Palermo 90146, Italy
| | - Vincenzo Martorana
- Institute of Biophysics, National Research Council, Palermo 90146, Italy
| | - Maria Grazia Ortore
- Department of Life and Environmental Sciences, Marche Polytechnic University, Ancona 60131, Italy
| | | |
Collapse
|
22
|
Chaari A. Molecular chaperones biochemistry and role in neurodegenerative diseases. Int J Biol Macromol 2019; 131:396-411. [DOI: 10.1016/j.ijbiomac.2019.02.148] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 02/25/2019] [Accepted: 02/25/2019] [Indexed: 02/07/2023]
|
23
|
Nadeem A, Ho JCS, Tran TH, Paul S, Granqvist V, Despretz N, Svanborg C. Beta-sheet-specific interactions with heat shock proteins define a mechanism of delayed tumor cell death in response to HAMLET. J Mol Biol 2019; 431:2612-2627. [PMID: 31082436 DOI: 10.1016/j.jmb.2019.05.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 05/02/2019] [Accepted: 05/04/2019] [Indexed: 02/07/2023]
Abstract
As chaperones, heat shock proteins (HSPs) protect host cells against misfolded proteins that constitute a by-product of protein synthesis. Certain HSPs are also expressed on the surface of tumor cells, possibly to scavenge extracellular unfolded protein ligands and prevent them from becoming cytotoxic. HAMLET-a complex of partially unfolded alpha-lactalbumin and oleic acid-is relying on its N-terminal alpha-helical domain to perturb tumor cell membranes, and the cells die as a consequence of this interaction. Here we show that in parallel, cell surface HSPs bind the beta-sheet domain of alpha-lactalbumin and activate a temporarily protective loop, involving vesicular uptake and lysosomal accumulation. Later, HAMLET destroys lysosomal membrane integrity, and HAMLET release kills the remaining tumor cells. HSPs were identified as HAMLET targets in a proteomic screen and Hsp70-specific antibodies or shRNAs inhibited HAMLET uptake by tumor cells, which showed increased Hsp70 surface expression compared to differentiated cells. The results suggest that HAMLET engages tumor cells by two parallel recognition mechanisms, defined by alpha-helical- or beta-sheet domains of alpha-lactalbumin and resulting in an immediate death response, or a delay due to transient accumulation of the complex in the lysosomes. This dual response pattern was conserved among tumor cells but not seen in normal, differentiated cells. By two different mechanisms, HAMLET thus achieves a remarkably efficient elimination of tumor cells.
Collapse
Affiliation(s)
- Aftab Nadeem
- Department of Microbiology, Immunology and Glycobiology (MIG), Institute of Laboratory Medicine, Lund University, S-223 62 Lund, Sweden
| | - James C S Ho
- Department of Microbiology, Immunology and Glycobiology (MIG), Institute of Laboratory Medicine, Lund University, S-223 62 Lund, Sweden; Centre for Biomimetic Sensor Science, School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Drive, 637553, Singapore
| | - Tuan Hiep Tran
- Department of Microbiology, Immunology and Glycobiology (MIG), Institute of Laboratory Medicine, Lund University, S-223 62 Lund, Sweden
| | - Sanchari Paul
- Department of Microbiology, Immunology and Glycobiology (MIG), Institute of Laboratory Medicine, Lund University, S-223 62 Lund, Sweden
| | - Victoria Granqvist
- Department of Microbiology, Immunology and Glycobiology (MIG), Institute of Laboratory Medicine, Lund University, S-223 62 Lund, Sweden
| | - Nadege Despretz
- Department of Microbiology, Immunology and Glycobiology (MIG), Institute of Laboratory Medicine, Lund University, S-223 62 Lund, Sweden
| | - Catharina Svanborg
- Department of Microbiology, Immunology and Glycobiology (MIG), Institute of Laboratory Medicine, Lund University, S-223 62 Lund, Sweden.
| |
Collapse
|
24
|
Derf A, Mudududdla R, Bharate SB, Chaudhuri B. Inhibitors of Aβ42-induced endoplasmic reticular unfolded protein response (UPR ER), in yeast, also rescue yeast cells from Aβ42-mediated apoptosis. Eur J Pharm Sci 2019; 128:118-127. [PMID: 30502452 DOI: 10.1016/j.ejps.2018.11.029] [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: 09/27/2018] [Revised: 11/10/2018] [Accepted: 11/25/2018] [Indexed: 11/19/2022]
Abstract
Aggregated Aβ peptides which cause amyloid deposits, a characteristic of Alzheimer's disease (AD), activate a stress response in the endoplasmic reticulum (ER), known as the unfolded protein response, UPRER. Nascent UPRER induction helps in reducing ER stress by eliminating accumulated misfolded/aggregated secretory proteins. However, prolonged UPRER induction may trigger apoptosis. Here we show that, when expressed in yeast with an NH2-terminal secretory signal sequence (ss), the 42-amino acid human Aβ42 (h_Aβ42), but not the mouse/ratAβ42 (m_Aβ42) which reportedly does not misfold/aggregate, induces UPRER as monitored via an eGFP reporter. We also show that expression of ss-h_Aβ42, not ss-m_Aβ42, blocks yeast cell growth, with cells expressing ss-h_Aβ42 manifesting distinctive features of apoptosis such as loss of mitochondrial membrane potential, increase in ROS levels and DNA fragmentation. Screening for suppressors of ss-h_Aβ42-activated UPRER-eGFP induction, in a computationally-designed 29-compound methoxy-stilbene library, revealed three compounds that reduce >95% of UPRER-eGFP induction at 5 μM concentration, with EC50 values of 40-50 nM. Surprisingly, the compounds also rescue yeast cells from ss-h_Aβ42-mediated apoptosis, with EC50-s of 50-60 nM. These results provide direct evidence, probably for the first time, that there is a direct correlation between deactivation of UPRER and attenuation of apoptosis.
Collapse
Affiliation(s)
- Asma Derf
- Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, UK
| | - Ramesh Mudududdla
- Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India
| | - Sandip B Bharate
- Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India; Academy of Scientific & Innovative Research, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India.
| | - Bhabatosh Chaudhuri
- Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, UK.
| |
Collapse
|
25
|
Jian WX, Zhang Z, Chu SF, Peng Y, Chen NH. Potential roles of brain barrier dysfunctions in the early stage of Alzheimer’s disease. Brain Res Bull 2018; 142:360-367. [DOI: 10.1016/j.brainresbull.2018.08.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 08/03/2018] [Accepted: 08/17/2018] [Indexed: 02/07/2023]
|
26
|
Jha MK, Kim JH, Song GJ, Lee WH, Lee IK, Lee HW, An SSA, Kim S, Suk K. Functional dissection of astrocyte-secreted proteins: Implications in brain health and diseases. Prog Neurobiol 2017; 162:37-69. [PMID: 29247683 DOI: 10.1016/j.pneurobio.2017.12.003] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 10/23/2017] [Accepted: 12/08/2017] [Indexed: 02/07/2023]
Abstract
Astrocytes, which are homeostatic cells of the central nervous system (CNS), display remarkable heterogeneity in their morphology and function. Besides their physical and metabolic support to neurons, astrocytes modulate the blood-brain barrier, regulate CNS synaptogenesis, guide axon pathfinding, maintain brain homeostasis, affect neuronal development and plasticity, and contribute to diverse neuropathologies via secreted proteins. The identification of astrocytic proteome and secretome profiles has provided new insights into the maintenance of neuronal health and survival, the pathogenesis of brain injury, and neurodegeneration. Recent advances in proteomics research have provided an excellent catalog of astrocyte-secreted proteins. This review categorizes astrocyte-secreted proteins and discusses evidence that astrocytes play a crucial role in neuronal activity and brain function. An in-depth understanding of astrocyte-secreted proteins and their pathways is pivotal for the development of novel strategies for restoring brain homeostasis, limiting brain injury/inflammation, counteracting neurodegeneration, and obtaining functional recovery.
Collapse
Affiliation(s)
- Mithilesh Kumar Jha
- Department of Pharmacology, Brain Science and Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu, Republic of Korea; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jong-Heon Kim
- Department of Pharmacology, Brain Science and Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Gyun Jee Song
- Department of Pharmacology, Brain Science and Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Won-Ha Lee
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
| | - In-Kyu Lee
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Ho-Won Lee
- Department of Neurology, Brain Science and Engineering Institute, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Seong Soo A An
- Department of BioNano Technology, Gachon University, Gyeonggi-do, Republic of Korea
| | - SangYun Kim
- Department of Neurology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Gyeonggi-do, Republic of Korea
| | - Kyoungho Suk
- Department of Pharmacology, Brain Science and Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu, Republic of Korea.
| |
Collapse
|
27
|
Zenaro E, Piacentino G, Constantin G. The blood-brain barrier in Alzheimer's disease. Neurobiol Dis 2017; 107:41-56. [PMID: 27425887 PMCID: PMC5600438 DOI: 10.1016/j.nbd.2016.07.007] [Citation(s) in RCA: 492] [Impact Index Per Article: 61.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 07/06/2016] [Accepted: 07/13/2016] [Indexed: 12/17/2022] Open
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disorder characterized by the pathological accumulation of amyloid beta (Aβ) peptides and neurofibrillary tangles containing hyperphosphorylated neuronal tau protein. AD pathology is also characterized by chronic brain inflammation, which promotes disease pathogenesis. In this context, the blood-brain barrier (BBB), a highly specialized endothelial cell membrane that lines cerebral microvessels, represents the interface between neural cells and circulating cells of the immune system. The BBB thus plays a key role in the generation and maintenance of chronic inflammation during AD. The BBB operates within the neurovascular unit (NVU), which includes clusters of glial cells, neurons and pericytes. The NVU becomes dysfunctional during AD, and each of its components may undergo functional changes that contribute to neuronal injury and cognitive deficit. In transgenic animals with AD-like pathology, recent studies have shown that circulating leukocytes migrate through the activated brain endothelium when certain adhesion molecules are expressed, penetrating into the brain parenchyma, interacting with the NVU components and potentially affecting their structural integrity and functionality. Therefore, migrating immune system cells in cerebral vessels act in concert with the modified BBB and may be integrated into the dysfunctional NVU. Notably, blocking the adhesion mechanisms controlling leukocyte-endothelial interactions inhibits both Aβ deposition and tau hyperphosphorylation, and reduces memory loss in AD models. The characterization of molecular mechanisms controlling vascular inflammation and leukocyte trafficking could therefore help to determine the basis of BBB dysfunction during AD and may lead to the development of new therapeutic approaches.
Collapse
Affiliation(s)
- Elena Zenaro
- Department of Medicine, Section of General Pathology, University of Verona, Strada le Grazie 8, 37134 Verona, Italy
| | - Gennj Piacentino
- Department of Medicine, Section of General Pathology, University of Verona, Strada le Grazie 8, 37134 Verona, Italy
| | - Gabriela Constantin
- Department of Medicine, Section of General Pathology, University of Verona, Strada le Grazie 8, 37134 Verona, Italy.
| |
Collapse
|
28
|
Roberts BR, Lind M, Wagen AZ, Rembach A, Frugier T, Li QX, Ryan TM, McLean CA, Doecke JD, Rowe CC, Villemagne VL, Masters CL. Biochemically-defined pools of amyloid-β in sporadic Alzheimer's disease: correlation with amyloid PET. Brain 2017; 140:1486-1498. [PMID: 28383676 DOI: 10.1093/brain/awx057] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 01/20/2017] [Indexed: 02/06/2023] Open
Abstract
We fractionated frontal cortical grey matter from human Alzheimer's disease and control subjects into four biochemically defined pools that represent four distinct compartments: soluble/cytosolic, peripheral membrane/vesicular cargo, integral lipid/membranous pools and aggregated/insoluble debris. Most of the readily extractable amyloid-β remains associated with a lipid/membranous compartment. There is an exchange of amyloid-β between the biochemical pools that was lost for the amyloid-β42 species in Alzheimer's disease, consistent with the peptide being irreversibly trapped in extracellular deposits. The quantitative amyloid-β data, combined with magnetic resonance imaging volumetric analysis of the amount of cortical grey matter in brain, allowed us to estimate the total mass of amyloid-β in Alzheimer's disease (6.5 mg) and control (1.7 mg) brains. The threshold positron emission tomography standard uptake value ratio of 1.4 equates to 5.0 μg amyloid-β/g of grey matter and the mean Alzheimer's disease dementia standard uptake value ratio level of 2.3 equates to 11.20 μg amyloid-β/g of grey matter. It takes 19 years to accumulate amyloid from the threshold positron emission tomography standard uptake value ratio to the mean value observed for Alzheimer's disease dementia. This accumulation time window combined with the difference of 4.8 mg of amyloid-β between Alzheimer's disease and control brain allows for a first approximation of amyloid-β accumulation of 28 ng/h. This equates to an estimated 2-5% of the total amyloid-β production being deposited as insoluble plaques. Understanding these rates of amyloid-β accumulation allows for a more quantitative approach in targeting the failure of amyloid-β clearance in sporadic Alzheimer's disease.
Collapse
Affiliation(s)
- Blaine R Roberts
- The Florey Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Monica Lind
- The Florey Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Aaron Z Wagen
- The Florey Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Alan Rembach
- The Florey Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Tony Frugier
- Department of Pharmacology and Therapeutics, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Qiao-Xin Li
- The Florey Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Timothy M Ryan
- The Florey Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Catriona A McLean
- The Florey Institute, The University of Melbourne, Parkville, Victoria 3010, Australia.,Department of Anatomical Pathology, Alfred Hospital, Prahran, Victoria 3004, Australia
| | - James D Doecke
- CSIRO Health and Biosecurity/Australian e-Health Research Centre and Cooperative Research Centre of Mental Health, Brisbane, QLD, Australia
| | - Christopher C Rowe
- The Florey Institute, The University of Melbourne, Parkville, Victoria 3010, Australia.,Department of Nuclear Medicine and Centre for PET, Austin Health, Heidelberg, Victoria, Australia
| | - Victor L Villemagne
- The Florey Institute, The University of Melbourne, Parkville, Victoria 3010, Australia.,Department of Nuclear Medicine and Centre for PET, Austin Health, Heidelberg, Victoria, Australia
| | - Colin L Masters
- The Florey Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
29
|
Co-chaperoning by amyloid-forming proteins: cystatins vs. crystallins. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2017; 46:789-793. [DOI: 10.1007/s00249-017-1214-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 04/10/2017] [Accepted: 04/24/2017] [Indexed: 02/04/2023]
|
30
|
Biza KV, Nastou KC, Tsiolaki PL, Mastrokalou CV, Hamodrakas SJ, Iconomidou VA. The amyloid interactome: Exploring protein aggregation. PLoS One 2017; 12:e0173163. [PMID: 28249044 PMCID: PMC5383009 DOI: 10.1371/journal.pone.0173163] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 02/15/2017] [Indexed: 11/22/2022] Open
Abstract
Protein-protein interactions are the quintessence of physiological activities, but also participate in pathological conditions. Amyloid formation, an abnormal protein-protein interaction process, is a widespread phenomenon in divergent proteins and peptides, resulting in a variety of aggregation disorders. The complexity of the mechanisms underlying amyloid formation/amyloidogenicity is a matter of great scientific interest, since their revelation will provide important insight on principles governing protein misfolding, self-assembly and aggregation. The implication of more than one protein in the progression of different aggregation disorders, together with the cited synergistic occurrence between amyloidogenic proteins, highlights the necessity for a more universal approach, during the study of these proteins. In an attempt to address this pivotal need we constructed and analyzed the human amyloid interactome, a protein-protein interaction network of amyloidogenic proteins and their experimentally verified interactors. This network assembled known interconnections between well-characterized amyloidogenic proteins and proteins related to amyloid fibril formation. The consecutive extended computational analysis revealed significant topological characteristics and unraveled the functional roles of all constituent elements. This study introduces a detailed protein map of amyloidogenicity that will aid immensely towards separate intervention strategies, specifically targeting sub-networks of significant nodes, in an attempt to design possible novel therapeutics for aggregation disorders.
Collapse
Affiliation(s)
- Konstantina V. Biza
- Section of Cell Biology and Biophysics, Department of Biology, School of Sciences, National and Kapodistrian University of Athens, Panepistimiopolis, Athens, Greece
| | - Katerina C. Nastou
- Section of Cell Biology and Biophysics, Department of Biology, School of Sciences, National and Kapodistrian University of Athens, Panepistimiopolis, Athens, Greece
| | - Paraskevi L. Tsiolaki
- Section of Cell Biology and Biophysics, Department of Biology, School of Sciences, National and Kapodistrian University of Athens, Panepistimiopolis, Athens, Greece
| | - Chara V. Mastrokalou
- Section of Cell Biology and Biophysics, Department of Biology, School of Sciences, National and Kapodistrian University of Athens, Panepistimiopolis, Athens, Greece
| | - Stavros J. Hamodrakas
- Section of Cell Biology and Biophysics, Department of Biology, School of Sciences, National and Kapodistrian University of Athens, Panepistimiopolis, Athens, Greece
| | - Vassiliki A. Iconomidou
- Section of Cell Biology and Biophysics, Department of Biology, School of Sciences, National and Kapodistrian University of Athens, Panepistimiopolis, Athens, Greece
- * E-mail:
| |
Collapse
|
31
|
Mathews PM, Levy E. Cystatin C in aging and in Alzheimer's disease. Ageing Res Rev 2016; 32:38-50. [PMID: 27333827 DOI: 10.1016/j.arr.2016.06.003] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 06/08/2016] [Accepted: 06/08/2016] [Indexed: 12/13/2022]
Abstract
Under normal conditions, the function of catalytically active proteases is regulated, in part, by their endogenous inhibitors, and any change in the synthesis and/or function of a protease or its endogenous inhibitors may result in inappropriate protease activity. Altered proteolysis as a result of an imbalance between active proteases and their endogenous inhibitors can occur during normal aging, and such changes have also been associated with multiple neuronal diseases, including Amyotrophic Lateral Sclerosis (ALS), rare heritable neurodegenerative disorders, ischemia, some forms of epilepsy, and Alzheimer's disease (AD). One of the most extensively studied endogenous inhibitor is the cysteine-protease inhibitor cystatin C (CysC). Changes in the expression and secretion of CysC in the brain have been described in various neurological disorders and in animal models of neurodegeneration, underscoring a role for CysC in these conditions. In the brain, multiple in vitro and in vivo findings have demonstrated that CysC plays protective roles via pathways that depend upon the inhibition of endosomal-lysosomal pathway cysteine proteases, such as cathepsin B (Cat B), via the induction of cellular autophagy, via the induction of cell proliferation, or via the inhibition of amyloid-β (Aβ) aggregation. We review the data demonstrating the protective roles of CysC under conditions of neuronal challenge and the protective pathways induced by CysC under various conditions. Beyond highlighting the essential role that balanced proteolytic activity plays in supporting normal brain aging, these findings suggest that CysC is a therapeutic candidate that can potentially prevent brain damage and neurodegeneration.
Collapse
Affiliation(s)
- Paul M Mathews
- Departments of Psychiatry, New York University School of Medicine, USA; Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY 10962, USA
| | - Efrat Levy
- Departments of Psychiatry, New York University School of Medicine, USA; Biochemistry and Molecular Pharmacology, New York University School of Medicine, USA; Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY 10962, USA.
| |
Collapse
|
32
|
Žerovnik E. Putative alternative functions of human stefin B (cystatin B): binding to amyloid-beta, membranes, and copper. J Mol Recognit 2016; 30. [PMID: 27577977 DOI: 10.1002/jmr.2562] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 07/25/2016] [Accepted: 07/26/2016] [Indexed: 12/17/2022]
Abstract
We describe studies performed thus far on stefin B from the family of cystatins as a model protein for folding and amyloid fibril formation studies. We also briefly mention our studies on aggregation of some of the missense EPM1 mutants of stefin B in cells, which mimic additional pathological traits (gain in toxic function) in selected patients with EPM1 disease. We collected data on the reported interactors of stefin B and discuss several hypotheses of possible cytosolic alternative functions.
Collapse
Affiliation(s)
- Eva Žerovnik
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, Ljubljana, Slovenia.,CipKeBip-Centre of Excellence for Integrated Approaches in Chemistry and Biology of Proteins, Ljubljana, Slovenia
| |
Collapse
|
33
|
Mangione MR, Vilasi S, Marino C, Librizzi F, Canale C, Spigolon D, Bucchieri F, Fucarino A, Passantino R, Cappello F, Bulone D, San Biagio PL. Hsp60, amateur chaperone in amyloid-beta fibrillogenesis. Biochim Biophys Acta Gen Subj 2016; 1860:2474-2483. [PMID: 27474204 DOI: 10.1016/j.bbagen.2016.07.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 07/14/2016] [Accepted: 07/24/2016] [Indexed: 10/21/2022]
Abstract
BACKGROUND Molecular chaperones are a very special class of proteins that play essential roles in many cellular processes like folding, targeting and transport of proteins. Moreover, recent evidence indicates that chaperones can act as potentially strong suppressor agents in Alzheimer's disease (AD). Indeed, in vitro experiments demonstrate that several chaperones are able to significantly slow down or suppress aggregation of Aβ peptide and in vivo studies reveal that treatment with specific chaperones or their overexpression can ameliorate some distinct pathological signs characterizing AD. METHODS Here we investigate using a biophysical approach (fluorescence, circular dichroism (CD), transmission electron (TEM) and atomic force (AFM) microscopy, size exclusion chromatography (SEC)) the effect of the human chaperonin Hsp60 on Aβ fibrillogenesis. RESULTS We found that Hsp60 powerfully inhibits Aβ amyloid aggregation, by closing molecular pathways leading to peptide fibrillogenesis. CONCLUSIONS We observe that Hsp60 inhibits Aβ aggregation through a more complex mechanism than a simple folding chaperone action. The action is specifically directed toward the early oligomeric species behaving as aggregation seeds for on-pathway amyloid fibrillogenesis. GENERAL SIGNIFICANCE Understanding the specificity of the molecular interactions of Hsp60 with amyloid Aβ peptide allowed us to emphasize the important aspects to be taken into consideration when considering the recent promising therapeutic strategies for neurodegeneration.
Collapse
Affiliation(s)
| | - Silvia Vilasi
- Institute of Biophysics, National Research Council, Palermo, Italy.
| | - Claudia Marino
- Institute of Biophysics, National Research Council, Palermo, Italy; Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA; Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Italy
| | - Fabio Librizzi
- Institute of Biophysics, National Research Council, Palermo, Italy
| | - Claudio Canale
- Nanophysics Department, Istituto Italiano di Tecnologia, Italy
| | - Dario Spigolon
- Institute of Biophysics, National Research Council, Palermo, Italy; Department of Physics and Chemistry, University of Palermo, Palermo, Italy
| | - Fabio Bucchieri
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Italy; Institute of Biomedicine and Molecular Immunology, National Research Council, Palermo, Italy; Euro-Mediterranean Institute of Science and Technology, Palermo, Italy
| | - Alberto Fucarino
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Italy; Euro-Mediterranean Institute of Science and Technology, Palermo, Italy
| | - Rosa Passantino
- Institute of Biophysics, National Research Council, Palermo, Italy
| | - Francesco Cappello
- Institute of Biophysics, National Research Council, Palermo, Italy; Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Italy; Euro-Mediterranean Institute of Science and Technology, Palermo, Italy
| | - Donatella Bulone
- Institute of Biophysics, National Research Council, Palermo, Italy
| | | |
Collapse
|
34
|
Gadhave K, Bolshette N, Ahire A, Pardeshi R, Thakur K, Trandafir C, Istrate A, Ahmed S, Lahkar M, Muresanu DF, Balea M. The ubiquitin proteasomal system: a potential target for the management of Alzheimer's disease. J Cell Mol Med 2016; 20:1392-407. [PMID: 27028664 PMCID: PMC4929298 DOI: 10.1111/jcmm.12817] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 01/17/2016] [Indexed: 01/06/2023] Open
Abstract
The cellular quality control system degrades abnormal or misfolded proteins and consists of three different mechanisms: the ubiquitin proteasomal system (UPS), autophagy and molecular chaperones. Any disturbance in this system causes proteins to accumulate, resulting in neurodegenerative diseases such as amyotrophic lateral sclerosis, Alzheimer's disease (AD), Parkinson's disease, Huntington's disease and prion or polyglutamine diseases. Alzheimer's disease is currently one of the most common age-related neurodegenerative diseases. However, its exact cause and pathogenesis are unknown. Currently approved medications for AD provide symptomatic relief; however, they fail to influence disease progression. Moreover, the components of the cellular quality control system represent an important focus for the development of targeted and potent therapies for managing AD. This review aims to evaluate whether existing evidence supports the hypothesis that UPS impairment causes the early pathogenesis of neurodegenerative disorders. The first part presents basic information about the UPS and its molecular components. The next part explains how the UPS is involved in neurodegenerative disorders. Finally, we emphasize how the UPS influences the management of AD. This review may help in the design of future UPS-related therapies for AD.
Collapse
Affiliation(s)
- Kundlik Gadhave
- Laboratory of Neurobiology, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Gauhati Medical College, Guwahati, Assam, India
| | - Nityanand Bolshette
- Institutional Level Biotech hub (IBT hub), Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Gauhati Medical College, Guwahati, Assam, India
| | - Ashutosh Ahire
- Laboratory of Neurobiology, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Gauhati Medical College, Guwahati, Assam, India
| | - Rohit Pardeshi
- Institutional Level Biotech hub (IBT hub), Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Gauhati Medical College, Guwahati, Assam, India
| | - Krishan Thakur
- Institutional Level Biotech hub (IBT hub), Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Gauhati Medical College, Guwahati, Assam, India
| | - Cristiana Trandafir
- Faculty of Medicine, University of Medicine and Pharmacy "Iuliu Hatieganu", Cluj-Napoca, Romania
| | - Alexandru Istrate
- Faculty of Medicine, University of Medicine and Pharmacy "Iuliu Hatieganu", Cluj-Napoca, Romania
| | - Sahabuddin Ahmed
- Laboratory of Neurobiology, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Gauhati Medical College, Guwahati, Assam, India
| | - Mangala Lahkar
- Laboratory of Neurobiology, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Gauhati Medical College, Guwahati, Assam, India
- Institutional Level Biotech hub (IBT hub), Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Gauhati Medical College, Guwahati, Assam, India
| | - Dafin F Muresanu
- Faculty of Medicine, University of Medicine and Pharmacy "Iuliu Hatieganu", Cluj-Napoca, Romania
- Department of Clinical Neurosciences, University of Medicine and Pharmacy "Iuliu Hatieganu", Cluj-Napoca, Romania
- "RoNeuro" Institute for Neurological Research and Diagnostic, Cluj-Napoca, Romania
| | - Maria Balea
- Department of Clinical Neurosciences, University of Medicine and Pharmacy "Iuliu Hatieganu", Cluj-Napoca, Romania
| |
Collapse
|
35
|
Rosen RF, Tomidokoro Y, Farberg AS, Dooyema J, Ciliax B, Preuss TM, Neubert TA, Ghiso JA, LeVine H, Walker LC. Comparative pathobiology of β-amyloid and the unique susceptibility of humans to Alzheimer's disease. Neurobiol Aging 2016; 44:185-196. [PMID: 27318146 DOI: 10.1016/j.neurobiolaging.2016.04.019] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 04/22/2016] [Accepted: 04/23/2016] [Indexed: 10/21/2022]
Abstract
The misfolding and accumulation of the protein fragment β-amyloid (Aβ) is an early and essential event in the pathogenesis of Alzheimer's disease (AD). Despite close biological similarities among primates, humans appear to be uniquely susceptible to the profound neurodegeneration and dementia that characterize AD, even though nonhuman primates deposit copious Aβ in senile plaques and cerebral amyloid-β angiopathy as they grow old. Because the amino acid sequence of Aβ is identical in all primates studied to date, we asked whether differences in the properties of aggregated Aβ might underlie the vulnerability of humans and the resistance of other primates to AD. In a comparison of aged squirrel monkeys (Saimiri sciureus) and humans with AD, immunochemical and mass spectrometric analyses indicate that the populations of Aβ fragments are largely similar in the 2 species. In addition, Aβ-rich brain extracts from the brains of aged squirrel monkeys and AD patients similarly seed the deposition of Aβ in a transgenic mouse model. However, the epitope exposure of aggregated Aβ differs in sodium dodecyl sulfate-stable oligomeric Aβ from the 2 species. In addition, the high-affinity binding of (3)H Pittsburgh Compound B to Aβ is significantly diminished in tissue extracts from squirrel monkeys compared with AD patients. These findings support the hypothesis that differences in the pathobiology of aggregated Aβ among primates are linked to post-translational attributes of the misfolded protein, such as molecular conformation and/or the involvement of species-specific cofactors.
Collapse
Affiliation(s)
- Rebecca F Rosen
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA.
| | | | - Aaron S Farberg
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Jeromy Dooyema
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Brian Ciliax
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Todd M Preuss
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA; Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Thomas A Neubert
- Department of Biochemistry and Molecular Pharmacology, Kimmel Center for Biology and Medicine at the Skirball Institute, NYU School of Medicine, New York, NY, USA
| | - Jorge A Ghiso
- Department of Pathology, NYU School of Medicine, New York, NY, USA; Department of Psychiatry, NYU School of Medicine, New York, NY, USA
| | - Harry LeVine
- Department of Molecular & Cellular Biochemistry, Center on Aging, Center for Structural Biology, University of Kentucky, Lexington, KY, USA
| | - Lary C Walker
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA; Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
36
|
Wilhelmus MMM, de Jager M, Smit AB, van der Loo RJ, Drukarch B. Catalytically active tissue transglutaminase colocalises with Aβ pathology in Alzheimer's disease mouse models. Sci Rep 2016; 6:20569. [PMID: 26837469 PMCID: PMC4738336 DOI: 10.1038/srep20569] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 01/04/2016] [Indexed: 02/06/2023] Open
Abstract
Alzheimer's disease (AD) is characterised by amyloid-beta (Aβ) protein deposition in the brain. Posttranslational modifications in Aβ play an important role in Aβ deposition. Tissue transglutaminase (tTG) is an enzyme involved in posttranslational cross-linking of proteins. tTG levels and activity are increased in AD brains, and tTG is associated with Aβ deposits and lesion-associated astrocytes in AD cases. Furthermore, Aβ is a substrate of tTG-catalysed cross-linking. To study the role of tTG in Aβ pathology, we compared tTG distribution and activity in both the APPSWE/PS1ΔE9 and APP23 mice models with human AD. Using immunohistochemistry, we found association of both tTG and in situ active tTG with Aβ plaques and vascular Aβ, in early and late stages of Aβ deposition. In addition, tTG staining colocalised with Aβ-associated reactive astrocytes. Thus, alike human AD cases, tTG was associated with Aβ depositions in these AD models. Although, distribution pattern and spatial overlay of both tTG and its activity with Aβ pathology was substantially different from human AD cases, our findings provide evidence for an early role of tTG in Aβ pathology. Yet, species differences should be taken into account when using these models to study the role of tTG in Aβ pathology.
Collapse
Affiliation(s)
- Micha M M Wilhelmus
- Department of Anatomy and Neurosciences, Neuroscience Campus Amsterdam, VU medical center, Amsterdam, The Netherlands
| | - Mieke de Jager
- Department of Anatomy and Neurosciences, Neuroscience Campus Amsterdam, VU medical center, Amsterdam, The Netherlands
| | - August B Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, VU University, Amsterdam, the Netherlands
| | - Rolinka J van der Loo
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, VU University, Amsterdam, the Netherlands
| | - Benjamin Drukarch
- Department of Anatomy and Neurosciences, Neuroscience Campus Amsterdam, VU medical center, Amsterdam, The Netherlands
| |
Collapse
|
37
|
Moore KM, Girens RE, Larson SK, Jones MR, Restivo JL, Holtzman DM, Cirrito JR, Yuede CM, Zimmerman SD, Timson BF. A spectrum of exercise training reduces soluble Aβ in a dose-dependent manner in a mouse model of Alzheimer's disease. Neurobiol Dis 2015; 85:218-224. [PMID: 26563933 DOI: 10.1016/j.nbd.2015.11.004] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 09/17/2015] [Accepted: 11/07/2015] [Indexed: 01/17/2023] Open
Abstract
Physical activity has long been hypothesized to influence the risk and pathology of Alzheimer's disease. However, the amount of physical activity necessary for these benefits is unclear. We examined the effects of three months of low and high intensity exercise training on soluble Aβ40 and Aβ42 levels in extracellular enriched fractions from the cortex and hippocampus of young Tg2576 mice. Low (LOW) and high (HI) intensity exercise training animals ran at speeds of 15m/min on a level treadmill and 32 m/min at a 10% grade, respectively for 60 min per day, five days per week, from three to six months of age. Sedentary mice (SED) were placed on a level, non-moving, treadmill for the same duration. Soleus muscle citrate synthase activity increased by 39% in the LOW group relative to SED, and by 71% in the HI group relative to LOW, indicating an exercise training effect in these mice. Soluble Aβ40 concentrations decreased significantly in an exercise training dose-dependent manner in the cortex. In the hippocampus, concentrations were decreased significantly in the HI group relative to LOW and SED. Soluble Aβ42 levels also decreased significantly in an exercise training dose-dependent manner in both the cortex and hippocampus. Five proteins involved in Aβ clearance (neprilysin, IDE, MMP9, LRP1 and HSP70) were elevated by exercise training with its intensity playing a role in each case. Our data demonstrate that exercise training reduces extracellular soluble Aβ in the brains of Tg2576 mice in a dose-dependent manner through an up-regulation of Aβ clearance.
Collapse
Affiliation(s)
- Kaitlin M Moore
- Department of Biomedical Sciences, Missouri State University, Springfield, MO 65897, USA
| | - Renee E Girens
- Department of Biomedical Sciences, Missouri State University, Springfield, MO 65897, USA
| | - Sara K Larson
- Department of Biomedical Sciences, Missouri State University, Springfield, MO 65897, USA
| | - Maria R Jones
- Department of Biomedical Sciences, Missouri State University, Springfield, MO 65897, USA
| | - Jessica L Restivo
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - David M Holtzman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA; Hope Center, Washington University School of Medicine, St. Louis, MO 63110, USA; Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - John R Cirrito
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA; Hope Center, Washington University School of Medicine, St. Louis, MO 63110, USA; Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Carla M Yuede
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA; Hope Center, Washington University School of Medicine, St. Louis, MO 63110, USA; Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Scott D Zimmerman
- Department of Biomedical Sciences, Missouri State University, Springfield, MO 65897, USA
| | - Benjamin F Timson
- Department of Biomedical Sciences, Missouri State University, Springfield, MO 65897, USA.
| |
Collapse
|
38
|
Amemori T, Jendelova P, Ruzicka J, Urdzikova LM, Sykova E. Alzheimer's Disease: Mechanism and Approach to Cell Therapy. Int J Mol Sci 2015; 16:26417-51. [PMID: 26556341 PMCID: PMC4661820 DOI: 10.3390/ijms161125961] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 10/26/2015] [Accepted: 10/26/2015] [Indexed: 12/19/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common form of dementia. The risk of AD increases with age. Although two of the main pathological features of AD, amyloid plaques and neurofibrillary tangles, were already recognized by Alois Alzheimer at the beginning of the 20th century, the pathogenesis of the disease remains unsettled. Therapeutic approaches targeting plaques or tangles have not yet resulted in satisfactory improvements in AD treatment. This may, in part, be due to early-onset and late-onset AD pathogenesis being underpinned by different mechanisms. Most animal models of AD are generated from gene mutations involved in early onset familial AD, accounting for only 1% of all cases, which may consequently complicate our understanding of AD mechanisms. In this article, the authors discuss the pathogenesis of AD according to the two main neuropathologies, including senescence-related mechanisms and possible treatments using stem cells, namely mesenchymal and neural stem cells.
Collapse
Affiliation(s)
- Takashi Amemori
- Department of Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Videnska 1083, 142 20 Prague 4, Czech Republic.
| | - Pavla Jendelova
- Department of Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Videnska 1083, 142 20 Prague 4, Czech Republic.
- Department of Neuroscience, 2nd Faculty of Medicine, Charles University, V Uvalu 84, 150 06 Prague 5, Czech Republic.
| | - Jiri Ruzicka
- Department of Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Videnska 1083, 142 20 Prague 4, Czech Republic.
| | - Lucia Machova Urdzikova
- Department of Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Videnska 1083, 142 20 Prague 4, Czech Republic.
| | - Eva Sykova
- Department of Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Videnska 1083, 142 20 Prague 4, Czech Republic.
- Department of Neuroscience, 2nd Faculty of Medicine, Charles University, V Uvalu 84, 150 06 Prague 5, Czech Republic.
| |
Collapse
|
39
|
Bruggink KA, Kuiperij HB, Gloerich J, Otte-Höller I, Rozemuller AJM, Claassen JAHR, Küsters B, Verbeek MM. Dickkopf-related protein 3 is a potential Aβ-associated protein in Alzheimer's Disease. J Neurochem 2015; 134:1152-62. [PMID: 26119087 DOI: 10.1111/jnc.13216] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 06/15/2015] [Accepted: 06/16/2015] [Indexed: 11/27/2022]
Abstract
Amyloid-β (Aβ) is the most prominent protein in Alzheimer's disease (AD) senile plaques. In addition, Aβ interacts with a variety of Aβ-associated proteins (AAPs), some of which can form complexes with Aβ and influence its clearance, aggregation or toxicity. Identification of novel AAPs may shed new light on the pathophysiology of AD and the metabolic fate of Aβ. In this study, we aimed to identify new AAPs by searching for proteins that may form soluble complexes with Aβ in CSF, using a proteomics approach. We identified the secreted Wnt pathway protein Dickkopf-related protein 3 (Dkk-3) as a potential Aβ-associated protein. Using immunohistochemistry on human AD brain tissue, we observed that (i) Dkk-3 co-localizes with Aβ in the brain, both in diffuse and classic plaques. (ii) Dkk-3 is expressed in neurons and in blood vessel walls in the brain and (iii) is secreted by leptomeningeal smooth muscle cells in vitro. Finally, measurements using ELISA revealed that (iv) Dkk-3 protein is abundantly present in both cerebrospinal fluid and serum, but its levels are similar in non-demented controls and patients with AD, Lewy body dementia, and frontotemporal dementia. Our study demonstrates that Dkk-3 is a hitherto unidentified Aβ-associated protein which, given its relatively high cerebral concentrations and co-localization with Aβ, is potentially involved in AD pathology. In this study, we propose that Dickkopf-related protein-3 (Dkk-3) might be a novel Amyloid-β (Aβ) associated protein. We demonstrate that Dkk-3 is expressed in the brain, especially in vessel walls, and co-localizes with Aβ in senile plaques. Furthermore, Dkk-3 levels in cerebrospinal fluid strongly correlate with Aβ40 levels, but were not suitable to discriminate non-demented controls and patients with dementia.
Collapse
Affiliation(s)
- Kim A Bruggink
- Department of Neurology, Department of Laboratory Medicine, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Radboud Alzheimer Centre, Nijmegen, The Netherlands
| | - H Bea Kuiperij
- Department of Neurology, Department of Laboratory Medicine, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Radboud Alzheimer Centre, Nijmegen, The Netherlands
| | - Jolein Gloerich
- Department of Laboratory Medicine, Radboud Proteomics Centre, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Irene Otte-Höller
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Annemieke J M Rozemuller
- Department of Pathology and Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Jurgen A H R Claassen
- Department of Geriatric Medicine, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Radboud Alzheimer Centre, Nijmegen, The Netherlands
| | - Benno Küsters
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Pathology, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Marcel M Verbeek
- Department of Neurology, Department of Laboratory Medicine, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Radboud Alzheimer Centre, Nijmegen, The Netherlands
| |
Collapse
|
40
|
Marsh NM, Wareham A, White BG, Miskiewicz EI, Landry J, MacPhee DJ. HSPB8 and the Cochaperone BAG3 Are Highly Expressed During the Synthetic Phase of Rat Myometrium Programming During Pregnancy. Biol Reprod 2015; 92:131. [PMID: 25904010 DOI: 10.1095/biolreprod.114.125401] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 04/09/2015] [Indexed: 12/17/2022] Open
Abstract
The small heat shock protein (HSP) B family of proteins are a group of molecular chaperones that enable tissues to adapt to changes in their local environments during differentiation, stress, or disease conditions. The objective of this research was to characterize the expression of HSPB8 and its cochaperone Bcl2-associated athanogene 3 (BAG3) in nonpregnant (NP) and pregnant rat myometrium during myometrial programming. Rat myometrium was collected from NP and pregnant rats as well as 1 day postpartum (PP) and samples prepared for immunoblot and immunofluorescence analysis. Immunoblot analysis determined that HSPB8 protein expression was significantly elevated at Day (D) 15, D17, and D19 compared to expression at NP and D6, while BAG3 expression was significantly elevated at D15 compared to NP, and D17 compared to NP, D6, D23, and PP time points (P < 0.05). In situ, HSPB8 and BAG3 were predominantly localized to myometrial cells throughout pregnancy, with intense cytoplasmic HSPB8 and BAG3 detection on D15 and D17 in both longitudinal and circular muscle layers. Immunoblot analysis of HSPB8 and BAG3 protein expression in myometrium from unilateral pregnancies also revealed that expression of both proteins was significantly increased at D15 in gravid compared to nongravid horns. Thus, HSPB8 and BAG3 are highly expressed during the synthetic phase of myometrial differentiation marked by initiation of uterine distension and myometrial hypertrophy. HSPB8 and BAG3 could be regulators of the protein quality control required for this process.
Collapse
Affiliation(s)
- Noelle M Marsh
- Division of Biomedical Sciences, Health Sciences Centre, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland and Labrador, Canada
| | - Angela Wareham
- Division of Biomedical Sciences, Health Sciences Centre, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland and Labrador, Canada
| | - Bryan G White
- Okanagan College-Salmon Arm Campus, Salmon Arm, British Columbia, Canada
| | - Ewa I Miskiewicz
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Jacques Landry
- Centre de Recherche en Cancerologie de l'Universite Laval, L'Hotel-Dieu de Quebec, Quebec, Quebec, Canada
| | - Daniel J MacPhee
- One Reproductive Health Research Group, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
41
|
Smith HL, Li W, Cheetham ME. Molecular chaperones and neuronal proteostasis. Semin Cell Dev Biol 2015; 40:142-52. [PMID: 25770416 PMCID: PMC4471145 DOI: 10.1016/j.semcdb.2015.03.003] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 02/27/2015] [Accepted: 03/04/2015] [Indexed: 12/13/2022]
Abstract
Protein homeostasis (proteostasis) is essential for maintaining the functionality of the proteome. The disruption of proteostasis, due to genetic mutations or an age-related decline, leads to aberrantly folded proteins that typically lose their function. The accumulation of misfolded and aggregated protein is also cytotoxic and has been implicated in the pathogenesis of neurodegenerative diseases. Neurons have developed an intrinsic protein quality control network, of which molecular chaperones are an essential component. Molecular chaperones function to promote efficient folding and target misfolded proteins for refolding or degradation. Increasing molecular chaperone expression can suppress protein aggregation and toxicity in numerous models of neurodegenerative disease; therefore, molecular chaperones are considered exciting therapeutic targets. Furthermore, mutations in several chaperones cause inherited neurodegenerative diseases. In this review, we focus on the importance of molecular chaperones in neurodegenerative diseases, and discuss the advances in understanding their protective mechanisms.
Collapse
Affiliation(s)
- Heather L Smith
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | - Wenwen Li
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | | |
Collapse
|
42
|
Filipcik P, Cente M, Zilka N, Smolek T, Hanes J, Kucerak J, Opattova A, Kovacech B, Novak M. Intraneuronal accumulation of misfolded tau protein induces overexpression of Hsp27 in activated astrocytes. Biochim Biophys Acta Mol Basis Dis 2015; 1852:1219-29. [PMID: 25772164 DOI: 10.1016/j.bbadis.2015.03.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 02/13/2015] [Accepted: 03/06/2015] [Indexed: 01/20/2023]
Abstract
Accumulation of misfolded forms of microtubule associated, neuronal protein tau causes neurofibrillary degeneration typical of Alzheimer's disease and other tauopathies. This process is accompanied by elevated cellular stress and concomitant deregulation of heat-shock proteins. We used a transgenic rat model of tauopathy to study involvement of heat shock protein 27 (Hsp27) in the process of neurofibrillary degeneration, its cell type specific expression and correlation with the amount of insoluble tau protein aggregates. The expression of Hsp27-mRNA is more than doubled and levels of Hsp27 protein tripled in aged transgenic animals with tau pathology. The data revealed a strong positive and highly significant correlation between Hsp27-mRNA and amount of sarkosyl insoluble tau. Interestingly, intracellular accumulation of insoluble misfolded tau protein in neurons was associated with overexpression of Hsp27 almost exclusively in reactive astrocytes, not in neurons. The topological dissociation of neuronally expressed pathological tau and the induction of astrocytic Hsp27, GFAP, and Vimentin along with up-regulation of microglia specific markers such as CD18, CD68 and C3 point to cooperation of astrocytes, microglia and neurons in response to intra-neuronal accumulation of insoluble tau. Our data suggest that over expression of Hsp27 represents a part of microglia-mediated astrocytic response mechanism in the process of neurofibrillary degeneration, which is not necessarily associated with neuroprotection and which in contrary may accelerate neurodegeneration in late stage of the disease. This phenomenon should be considered during development of disease modifying strategies for treatment of tauopathies and AD via regulation of activity of Hsp27.
Collapse
Affiliation(s)
- Peter Filipcik
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, 84510 Bratislava, Slovakia; Axon Neuroscience SE, Bratislava, Slovakia
| | - Martin Cente
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, 84510 Bratislava, Slovakia; Axon Neuroscience SE, Bratislava, Slovakia
| | - Norbert Zilka
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, 84510 Bratislava, Slovakia; Axon Neuroscience SE, Bratislava, Slovakia
| | - Tomas Smolek
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, 84510 Bratislava, Slovakia; Axon Neuroscience SE, Bratislava, Slovakia
| | | | - Juraj Kucerak
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, 84510 Bratislava, Slovakia; Axon Neuroscience SE, Bratislava, Slovakia
| | - Alena Opattova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, 84510 Bratislava, Slovakia
| | - Branislav Kovacech
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, 84510 Bratislava, Slovakia; Axon Neuroscience SE, Bratislava, Slovakia
| | - Michal Novak
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, 84510 Bratislava, Slovakia; Axon Neuroscience SE, Bratislava, Slovakia.
| |
Collapse
|
43
|
|
44
|
Enhanced bio-ethanol production via simultaneous saccharification and fermentation through a cell free enzyme system prepared by disintegration of waste of beer fermentation broth. KOREAN J CHEM ENG 2014. [DOI: 10.1007/s11814-014-0242-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
45
|
Ortega L, Calvillo M, Luna F, Pérez-Severiano F, Rubio-Osornio M, Guevara J, Limón ID. 17-AAG improves cognitive process and increases heat shock protein response in a model lesion with Aβ25-35. Neuropeptides 2014; 48:221-32. [PMID: 24819277 DOI: 10.1016/j.npep.2014.04.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Revised: 03/14/2014] [Accepted: 04/15/2014] [Indexed: 11/22/2022]
Abstract
Molecular chaperones, or heat shock proteins (HSP), have been implicated in numerous neurodegenerative disorders characterized by the accumulation of protein aggregates, such as Alzheimer disease. The agglomeration of insoluble structures of Aβ is thought to be responsible for neuronal death, which in turn leads to the loss of cognitive functions. Recent findings have shown that the induction of HSP decreases the level of abnormal protein aggregates, as well as demonstrating that 17-(allylamino)-17-demethoxygeldanamycin (17-AAG), an analogue of geldanamycin (GA), increases Aβ clearance through the induction of molecular chaperones in cell culture. In light of this discovery that HSP overexpression can be neuroprotective, the search for a way to pharmacologically induce the overexpression of HSP and other associated chaperones may lead to a promising approach for the treatment of neurodegenerative diseases. The aim of our study was to evaluate both the effect of 17-AAG on the cognitive process and the HSP response in rats injected with Aβ25-35 into the CA1 of the hippocampus. The results show that the injection of Aβ caused a significant increase in the expression of the HSP involved in the regulation of cellular proteostasis. While the HSP did not reverse excitotoxic damage, given that experimental subjects showed learning and memory deficits, the administration of 17-AAG prior to the injection of Aβ25-35 did show an improvement in the behavioral assessment that correlated with the upregulation of HSP70 in subjects injured with Aβ. Overall, our data shows that the pharmacological induction of HSP using 17-AAG may be an alternative treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Laura Ortega
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla 72570, Mexico; Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, México D.F. 04510, Mexico.
| | - Minerva Calvillo
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Instituto Nacional de Neurología y Neurocirugía, México D.F. 14269, Mexico.
| | - Félix Luna
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla 72570, Mexico.
| | - Francisca Pérez-Severiano
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía, México D.F. 14269, Mexico.
| | - Moisés Rubio-Osornio
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Instituto Nacional de Neurología y Neurocirugía, México D.F. 14269, Mexico.
| | - Jorge Guevara
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, México D.F. 04510, Mexico.
| | - Ilhuicamina Daniel Limón
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla 72570, Mexico.
| |
Collapse
|
46
|
Kakkar V, Meister-Broekema M, Minoia M, Carra S, Kampinga HH. Barcoding heat shock proteins to human diseases: looking beyond the heat shock response. Dis Model Mech 2014; 7:421-34. [PMID: 24719117 PMCID: PMC3974453 DOI: 10.1242/dmm.014563] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
There are numerous human diseases that are associated with protein misfolding and the formation of toxic protein aggregates. Activating the heat shock response (HSR)--and thus generally restoring the disturbed protein homeostasis associated with such diseases--has often been suggested as a therapeutic strategy. However, most data on activating the HSR or its downstream targets in mouse models of diseases associated with aggregate formation have been rather disappointing. The human chaperonome consists of many more heat shock proteins (HSPs) that are not regulated by the HSR, however, and researchers are now focusing on these as potential therapeutic targets. In this Review, we summarize the existing literature on a set of aggregation diseases and propose that each of them can be characterized or 'barcoded' by a different set of HSPs that can rescue specific types of aggregation. Some of these 'non-canonical' HSPs have demonstrated effectiveness in vivo, in mouse models of protein-aggregation disease. Interestingly, several of these HSPs also cause diseases when mutated--so-called chaperonopathies--which are also discussed in this Review.
Collapse
Affiliation(s)
- Vaishali Kakkar
- University Medical Center Groningen, University of Groningen, Department of Cell Biology, A. Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | - Melanie Meister-Broekema
- University Medical Center Groningen, University of Groningen, Department of Cell Biology, A. Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | - Melania Minoia
- University Medical Center Groningen, University of Groningen, Department of Cell Biology, A. Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | - Serena Carra
- Università degli Studi di Modena e Reggio Emilia, Dipartimento di Scienze Biomediche, Metaboliche e Neuroscienze, via G. Campi 287, 41125 Modena, Italy
| | - Harm H. Kampinga
- University Medical Center Groningen, University of Groningen, Department of Cell Biology, A. Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| |
Collapse
|
47
|
Minsky BB, Zheng B, Dubin PL. Inhibition of antithrombin and bovine serum albumin native state aggregation by heparin. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2014; 30:278-287. [PMID: 24313340 DOI: 10.1021/la4039232] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Protein native state aggregation, a major problem in pharmaceutical and biological processes, has been addressed pharmacologically by the addition of protein-binding excipients. Heparin (Hp), a highly sulfated polysaccharide, interacts with numerous proteins with moderate to high affinity, but reports about its effect on protein aggregation are contradictory. We studied the pH dependence of the aggregation of antithrombin (AT) and bovine serum albumin (BSA) in the presence and absence of heparin. High-precision turbidimetry showed strong aggregation for both AT and BSA in I = 10 mM NaCl, conditions at which electrostatically driven Hp binding and aggregation both occur, with more obvious aggregation of heparin-free AT appearing as larger aggregate size. Aggregation of AT was dramatically inhibited at Hp: protein 6:1 (mole ratio); however, the effect at 0.5:1 Hp:protein was greater for BSA. Frontal analysis capillary electrophoresis showed a much larger equilibrium association constant Kobs between Hp and AT, in accord with the onset of Hp binding at a higher pH; both effects are explained by the higher charge density of the positive domain for AT as revealed by modeling with DelPhi. The corresponding modeling images showed that these domains persist at high salt only for AT, consistent with the 160-fold drop in Kobs at 100 mM salt for BSA-Hp binding. The smaller inhibition effect for AT arises from the tendency of its uncomplexed monomer to form larger aggregates more rapidly, but the stronger binding of Hp to AT does not facilitate Hp-induced aggregate dissolution which occurs more readily for BSA. This can be attributed to the higher density of AT aggregates evidenced by higher fractal dimensions. Differences between inhibition and reversal by Hp arise because the former may depend on the stage at which Hp enters the aggregation process and the latter on aggregate size and morphology.
Collapse
Affiliation(s)
- Burcu Baykal Minsky
- Department of Chemistry, University of Massachusetts , Amherst, Massachusetts, 01003, United States
| | | | | |
Collapse
|
48
|
Hardas SS, Sultana R, Warrier G, Dan M, Wu P, Grulke EA, Tseng MT, Unrine JM, Graham UM, Yokel RA, Butterfield DA. Rat hippocampal responses up to 90 days after a single nanoceria dose extends a hierarchical oxidative stress model for nanoparticle toxicity. Nanotoxicology 2013; 8 Suppl 1:155-66. [DOI: 10.3109/17435390.2013.868059] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
| | | | | | - Mo Dan
- Department of Pharmaceutical Sciences and
| | - Peng Wu
- Chemical and Materials Engineering Department, University of Kentucky, Lexington, KY, USA,
| | - Eric A. Grulke
- Chemical and Materials Engineering Department, University of Kentucky, Lexington, KY, USA,
| | - Michael T. Tseng
- Department of Anatomical Sciences & Neurobiology, University of Louisville, Louisville, KY, USA,
| | - Jason M. Unrine
- Department of Plant and Soil Sciences, University of Kentucky, Lexington, KY, USA,
| | - Uschi M. Graham
- Center for Applied Energy Research, University of Kentucky, Lexington, KY, USA,
| | - Robert A. Yokel
- Department of Pharmaceutical Sciences and
- Graduate Center for Toxicology, University of Kentucky Academic Medical Center, Lexington, KY, USA, and
| | - D. Allan Butterfield
- Department of Chemistry,
- Center of Membrane Sciences, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
49
|
Rispoli A, Cipollini E, Catania S, Di Giaimo R, Pulice G, van Houte S, Sparla F, Dal Piaz F, Roncarati D, Trost P, Melli M. Insights in progressive myoclonus epilepsy: HSP70 promotes cystatin B polymerization. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2013; 1834:2591-9. [DOI: 10.1016/j.bbapap.2013.09.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Revised: 09/16/2013] [Accepted: 09/17/2013] [Indexed: 12/15/2022]
|
50
|
Calvillo M, Diaz A, Limon DI, Mayoral MA, Chánez-Cárdenas ME, Zenteno E, Montaño LF, Guevara J, Espinosa B. Amyloid-β(25-35) induces a permanent phosphorylation of HSF-1, but a transitory and inflammation-independent overexpression of Hsp-70 in C6 astrocytoma cells. Neuropeptides 2013; 47:339-46. [PMID: 23850171 DOI: 10.1016/j.npep.2013.06.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Revised: 05/17/2013] [Accepted: 06/12/2013] [Indexed: 11/28/2022]
Abstract
Two hallmarks of Alzheimer diseases are the continuous inflammatory process, and the brain deposit of Amyloid b (Aβ), a cytotoxic protein. The intracellular accumulation of Aβ(25-35) fractions, in the absence of Heat Shock proteins (Hsṕs), could be responsible for its cytotoxic activity. As, pro-inflammatory mediators and nitric oxide control the expression of Hsṕs, our aim was to investigate the effect of Aβ(25-35) on the concentration of IL-1β, TNF-α and nitrite levels, and their relation to pHSF-1, Hsp-60, -70 and -90 expressions, in the rat C6 astrocyte cells. Interleukin-specific ELISA kits, immunohistochemistry with monoclonal anti-Hsp and anti pHSF-1 antibodies, and histochemistry techniques, were used. Our results showed that Aβ25-35 treatment of C6 cells increased, significantly and consistently the concentration of IL-1β, TNF-α and nitrite 3 days after initiating treatment. The immunoreactivity of C6 cells to Hsp-70 reached its peak after 3 days of treatment followed by an abrupt decrease, as opposed to Hsp-60 and -90 expressions that showed an initial and progressive increase after 3 days of Aβ(25-35) treatment. pHSF-1 was identified throughout the experimental period. Nevertheless, progressive and sustained cell death was observed during all the treatment times and it was not caspase-3 dependent. Our results suggest that Hsp-70 temporary expression serves as a trigger to inhibit casapase-3 pathway and allow the expression of Hsp-60 and -90 in C6 astrocytoma cells stimulated with Aβ(25-35).
Collapse
Affiliation(s)
- Minerva Calvillo
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Instituto Nacional de Neurología y Neurocirugía "MVS", Mexico D.F. 14059, Mexico
| | | | | | | | | | | | | | | | | |
Collapse
|