1
|
Yang C, Liu G, Chen X, Le W. Cerebellum in Alzheimer's disease and other neurodegenerative diseases: an emerging research frontier. MedComm (Beijing) 2024; 5:e638. [PMID: 39006764 PMCID: PMC11245631 DOI: 10.1002/mco2.638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 06/04/2024] [Accepted: 06/12/2024] [Indexed: 07/16/2024] Open
Abstract
The cerebellum is crucial for both motor and nonmotor functions. Alzheimer's disease (AD), alongside other dementias such as vascular dementia (VaD), Lewy body dementia (DLB), and frontotemporal dementia (FTD), as well as other neurodegenerative diseases (NDs) like Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), Huntington's disease (HD), and spinocerebellar ataxias (SCA), are characterized by specific and non-specific neurodegenerations in central nervous system. Previously, the cerebellum's significance in these conditions was underestimated. However, advancing research has elevated its profile as a critical node in disease pathology. We comprehensively review the existing evidence to elucidate the relationship between cerebellum and the aforementioned diseases. Our findings reveal a growing body of research unequivocally establishing a link between the cerebellum and AD, other forms of dementia, and other NDs, supported by clinical evidence, pathological and biochemical profiles, structural and functional neuroimaging data, and electrophysiological findings. By contrasting cerebellar observations with those from the cerebral cortex and hippocampus, we highlight the cerebellum's distinct role in the disease processes. Furthermore, we also explore the emerging therapeutic potential of targeting cerebellum for the treatment of these diseases. This review underscores the importance of the cerebellum in these diseases, offering new insights into the disease mechanisms and novel therapeutic strategies.
Collapse
Affiliation(s)
- Cui Yang
- Institute of Neurology Sichuan Provincial People's Hospital School of Medicine University of Electronic Science and Technology of China Chengdu China
| | - Guangdong Liu
- Institute of Neurology Sichuan Provincial People's Hospital School of Medicine University of Electronic Science and Technology of China Chengdu China
| | - Xi Chen
- Institute of Neurology Sichuan Provincial People's Hospital School of Medicine University of Electronic Science and Technology of China Chengdu China
| | - Weidong Le
- Institute of Neurology Sichuan Provincial People's Hospital School of Medicine University of Electronic Science and Technology of China Chengdu China
| |
Collapse
|
2
|
Kebschull JM, Casoni F, Consalez GG, Goldowitz D, Hawkes R, Ruigrok TJH, Schilling K, Wingate R, Wu J, Yeung J, Uusisaari MY. Cerebellum Lecture: the Cerebellar Nuclei-Core of the Cerebellum. CEREBELLUM (LONDON, ENGLAND) 2024; 23:620-677. [PMID: 36781689 PMCID: PMC10951048 DOI: 10.1007/s12311-022-01506-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 12/10/2022] [Indexed: 02/15/2023]
Abstract
The cerebellum is a key player in many brain functions and a major topic of neuroscience research. However, the cerebellar nuclei (CN), the main output structures of the cerebellum, are often overlooked. This neglect is because research on the cerebellum typically focuses on the cortex and tends to treat the CN as relatively simple output nuclei conveying an inverted signal from the cerebellar cortex to the rest of the brain. In this review, by adopting a nucleocentric perspective we aim to rectify this impression. First, we describe CN anatomy and modularity and comprehensively integrate CN architecture with its highly organized but complex afferent and efferent connectivity. This is followed by a novel classification of the specific neuronal classes the CN comprise and speculate on the implications of CN structure and physiology for our understanding of adult cerebellar function. Based on this thorough review of the adult literature we provide a comprehensive overview of CN embryonic development and, by comparing cerebellar structures in various chordate clades, propose an interpretation of CN evolution. Despite their critical importance in cerebellar function, from a clinical perspective intriguingly few, if any, neurological disorders appear to primarily affect the CN. To highlight this curious anomaly, and encourage future nucleocentric interpretations, we build on our review to provide a brief overview of the various syndromes in which the CN are currently implicated. Finally, we summarize the specific perspectives that a nucleocentric view of the cerebellum brings, move major outstanding issues in CN biology to the limelight, and provide a roadmap to the key questions that need to be answered in order to create a comprehensive integrated model of CN structure, function, development, and evolution.
Collapse
Affiliation(s)
- Justus M Kebschull
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA.
| | - Filippo Casoni
- Division of Neuroscience, San Raffaele Scientific Institute, and San Raffaele University, Milan, Italy
| | - G Giacomo Consalez
- Division of Neuroscience, San Raffaele Scientific Institute, and San Raffaele University, Milan, Italy
| | - Daniel Goldowitz
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, Canada
| | - Richard Hawkes
- Department of Cell Biology & Anatomy and Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, T2N 4N1, Canada
| | - Tom J H Ruigrok
- Department of Neuroscience, Erasmus MC, Rotterdam, the Netherlands
| | - Karl Schilling
- Department of Anatomy, Anatomy & Cell Biology, Rheinische Friedrich-Wilhelms-Universität, 53115, Bonn, Federal Republic of Germany
| | - Richard Wingate
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Joshua Wu
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, Canada
| | - Joanna Yeung
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, Canada
| | - Marylka Yoe Uusisaari
- Neuronal Rhythms in Movement Unit, Okinawa Institute of Science and Technology, 1919-1 Tancha, Onna-Son, Kunigami-Gun, Okinawa, 904-0495, Japan.
| |
Collapse
|
3
|
Castillo-Mariqueo L, Giménez-Llort L. Clasping, ledge-score coordination and early gait impairments as primary behavioural markers of functional impairment in Alzheimer's disease. Behav Brain Res 2022; 435:114054. [PMID: 35961538 DOI: 10.1016/j.bbr.2022.114054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 07/23/2022] [Accepted: 08/08/2022] [Indexed: 11/30/2022]
Abstract
Motor performance facilitates the understanding of the functional state related to the progression of Alzheimer's disease (AD). At the translational level, this brief report refines the characterization of the motor dysfunction of the 3xTg-AD mouse model in different motor tasks, focusing on the abnormal clasping reflex and coordination impairments measured through the Phenotype Scoring System four items screening originally developed for models of ataxia. We studied male 3xTg-AD mice at 6, 12, and 16 months of age (mimicking the early, advanced, and late stages of the disease, respectively) and their age-matched non-transgenic counterparts with normal aging. According to the score, incidence, or severity of the four items and the total score, the 3xTg-AD mice showed deficiencies in all score elements. Clasping was increased independently of age, and its severity worsened with repeated testing. In contrast, the impairment of coordination worsened with the progress of the disease. The gait score was sensitive to genotype since early stages, and the worse ledge score was evident at 16 months. Kyphosis and ledge scores were sensitive to age. The impairments and functional limitations of male 3xTg-AD mice related to the stages of AD provide a scenario that allows understanding the heterogeneity of non-cognitive symptoms.
Collapse
Affiliation(s)
- Lidia Castillo-Mariqueo
- Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain; Department of Psychiatry and Forensic Medicine, School of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain.
| | - Lydia Giménez-Llort
- Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain; Department of Psychiatry and Forensic Medicine, School of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain.
| |
Collapse
|
4
|
Sun ZT, Ma C, Li GJ, Zheng XY, Hao YT, Yang Y, Wang X. Application of Antibody Fragments Against Aβ With Emphasis on Combined Application With Nanoparticles in Alzheimer's Disease. Front Pharmacol 2021; 12:654611. [PMID: 33967797 PMCID: PMC8100690 DOI: 10.3389/fphar.2021.654611] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 04/09/2021] [Indexed: 11/13/2022] Open
Abstract
Alzheimer’s disease (AD) is one of the most common neurodegenerative diseases and accumulating evidences suggest a key role of amyloid-β (Aβ) peptide in the pathogenesis of AD. According to the amyloid cascade hypothesis, the imbalance of producing and clearing Aβ is the beginning of neurodegeneration and dementia. Consequently, immunotherapy becomes popular through using antibodies against Aβ. However, many studies of monoclonal antibodies were stopped because adverse effects appeared or there were no evident benefits observed. Some antibody fragments have many advantages over monoclonal antibodies, such as small sizes, lack of the crystallizable fraction (Fc) and so on. There are three main antibody fragments, including single chain variable fragments (scFvs), Fab fragments and single-domain antibody fragments. Nanoparticles can facilitate the entry of drug molecules across the blood-brain barrier, making them become excellent carriers. Various kinds of nanoparticles have been applied in the treatment of AD. The combination of nanoparticles and antibody fragments against amyloid-β can be used in the diagnosis and treatment of Alzheimer’s disease. In this review, we summarize the progress of antibody fragments against amyloid-β in AD, focusing on the combined application with nanoparticles in the diagnosis and treatment of AD.
Collapse
Affiliation(s)
- Zhi-Ting Sun
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Chi Ma
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| | - Guang-Jian Li
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Xiang-Yu Zheng
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Yi-Tong Hao
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Yu Yang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Xu Wang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
5
|
Dennison JL, Ricciardi NR, Lohse I, Volmar CH, Wahlestedt C. Sexual Dimorphism in the 3xTg-AD Mouse Model and Its Impact on Pre-Clinical Research. J Alzheimers Dis 2021; 80:41-52. [PMID: 33459720 PMCID: PMC8075398 DOI: 10.3233/jad-201014] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Female sex is a leading risk factor for developing Alzheimer’s disease (AD). Sexual dimorphism in AD is gaining attention as clinical data show that women are not only more likely to develop AD but also to experience worse pathology and faster cognitive decline. Pre-clinical AD research in animal models often neglects to address sexual dimorphism in evaluation of behavioral or molecular characteristics and outcomes. This can compromise its translation to a clinical setting. The triple-transgenic AD mouse model (3xTg-AD) is a commonly used but unique AD model because it exhibits both amyloid and tau pathology, essential features of the human AD phenotype. Mounting evidence has revealed important sexually dimorphic characteristics of this animal model that have yet to be reviewed and thus, are often overlooked in studies using the 3xTg-AD model. In this review we conduct a thorough analysis of reports of sexual dimorphism in the 3xTg-AD model including findings of molecular, behavioral, and longevity-related sex differences in original research articles through August 2020. Importantly, we find results to be inconsistent, and that strain source and differing methodologies are major contributors to lack of consensus regarding traits of each sex. We first touch on the nature of sexual dimorphism in clinical AD, followed by a brief summary of sexual dimorphism in other major AD murine models before discussing the 3xTg-AD model in depth. We conclude by offering four suggestions to help unify pre-clinical mouse model AD research inspired by the NIH expectations for considering sex as a biological variable.
Collapse
Affiliation(s)
- Jessica L Dennison
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, USA.,Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Natalie R Ricciardi
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, USA.,Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Ines Lohse
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, USA.,Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Claude-Henry Volmar
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, USA.,Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Claes Wahlestedt
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, USA.,Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
6
|
Esquerda-Canals G, Roda AR, Martí-Clúa J, Montoliu-Gaya L, Rivera-Hernández G, Villegas S. Treatment with scFv-h3D6 Prevented Neuronal Loss and Improved Spatial Memory in Young 3xTg-AD Mice by Reducing the Intracellular Amyloid-β Burden. J Alzheimers Dis 2020; 70:1069-1091. [PMID: 31306135 DOI: 10.3233/jad-190484] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The intracellular deposition of amyloid-β (Aβ) peptides has been described in the brains of both Alzheimer's disease (AD) patients and animal models. A correlation between the intracellular amyloid burden and neurodegeneration has recently been reported in a triple-transgenic AD (3xTg-AD) murine model. In the present study, we assessed the effect of scFv-h3D6, an anti-Aβ single-chain variable fragment (scFv) derived from the antibody bapineuzumab, on amyloid pathology in 5-month-old 3xTg-AD female mice, focusing on intracellular Aβ clearance, neuronal survival, and functional abilities. We also examined neuroinflammation and the histology of peripheral organ samples to detect any adverse effects. A single intraperitoneal injection of scFv-h3D6 dramatically reduced intracellular Aβ burden in the deep layers of the cerebral cortex, pyramidal cells layer of the hippocampus, and basolateral amygdalar nucleus. The treatment prevented neuronal loss in the hippocampus and amygdala, while neither astrogliosis nor microgliosis was induced. Instead, an increase in the size of the white pulp after the treatment indicated that the spleen could be involved in the clearance mechanism. Although the treatment did not ameliorate behavioral and psychological symptoms of dementia-like symptoms, the results of cognitive testing pointed to a noticeable improvement in spatial memory. These findings indicated that the mechanism underlying the therapeutic effect of scFv-h3D6 was the clearance of intracellular Aβ, with subsequent prevention of neuronal loss and amelioration of cognitive disabilities. The treatment was safe in terms of neuroinflammation and kidney and liver function, whereas some effects on the spleen were observed.
Collapse
Affiliation(s)
- Gisela Esquerda-Canals
- Protein Design and Immunotherapy Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Edifici C, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain.,Departament de Biologia Cellular, de Fisiologia i d'Immunologia, Unitat de Citologia i d'Histologia, Facultat de Biociències, Edifici C, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Alejandro R Roda
- Protein Design and Immunotherapy Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Edifici C, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Joaquim Martí-Clúa
- Protein Design and Immunotherapy Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Edifici C, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain.,Departament de Biologia Cellular, de Fisiologia i d'Immunologia, Unitat de Citologia i d'Histologia, Facultat de Biociències, Edifici C, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Laia Montoliu-Gaya
- Protein Design and Immunotherapy Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Edifici C, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain.,Current address: Department of Clinical Chemistry and Transfusion Medicine, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Geovanny Rivera-Hernández
- Protein Design and Immunotherapy Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Edifici C, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Sandra Villegas
- Protein Design and Immunotherapy Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Edifici C, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| |
Collapse
|
7
|
Cognitive Impairment in the 3xTg-AD Mouse Model of Alzheimer's Disease is Affected by Aβ-ImmunoTherapy and Cognitive Stimulation. Pharmaceutics 2020; 12:pharmaceutics12100944. [PMID: 33023109 PMCID: PMC7601886 DOI: 10.3390/pharmaceutics12100944] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/19/2020] [Accepted: 09/30/2020] [Indexed: 12/11/2022] Open
Abstract
Clinical symptoms of Alzheimer’s Disease (AD) include behavioral alterations and cognitive impairment. These functional phenotypes early occur in triple-transgenic (3xTg-AD) mice. Specifically, behavioral alterations are first detected when mice are at around 2.5 months old and cognitive impairment in between 3- and 5-month-old mice. In this work, the effect of chronic Aβ-immunotherapy on behavioral and cognitive abilities was tested by monthly administering the antibody fragment scFv-h3D6 to 3xTg-AD female mice from 5 to 9 months of age. An untreated group was used as a reference, as well as to attain some information on the effect of training during the longitudinal study. Behavioral and psychological symptoms of dementia (BPSD)-like symptoms were already evident in 5-month-old mice, in the form of neophobia and anxious-like behavior. The exploratory activity decreased over the longitudinal study, not only for 3xTgAD mice but also for the corresponding non-transgenic mice (NTg). Learning abilities of 3xTg-AD mice were not seriously compromised but an impairment in long-term spatial memory was evident at 5 months of age. Interestingly, scFv-h3D6-treatment affected the cognitive impairment displayed by 5-month-old 3xTg-AD mice. It is worth noting that training also reduced cognitive impairment of 3xTg-AD mice over the longitudinal study, suggesting that to properly quantify the isolated therapeutic potential of any drug on cognition using this model it is convenient to perform a prompt, age-matched study rather than a longitudinal study. In addition, a combination of both training and Aβ-immunotherapy could constitute a possible approach to treat Alzheimer’s disease.
Collapse
|
8
|
Roda AR, Montoliu-Gaya L, Serra-Mir G, Villegas S. Both Amyloid-β Peptide and Tau Protein Are Affected by an Anti-Amyloid-β Antibody Fragment in Elderly 3xTg-AD Mice. Int J Mol Sci 2020; 21:E6630. [PMID: 32927795 PMCID: PMC7554787 DOI: 10.3390/ijms21186630] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/02/2020] [Accepted: 09/04/2020] [Indexed: 12/15/2022] Open
Abstract
Alzheimer's disease (AD) is the most common dementia worldwide. According to the amyloid hypothesis, the early accumulation of the Aβ-peptide triggers tau phosphorylation, synaptic dysfunction, and eventually neuronal death leading to cognitive impairment, as well as behavioral and psychological symptoms of dementia. ScFv-h3D6 is a single-chain variable fragment that has already shown its ability to diminish the amyloid burden in 5-month-old 3xTg-AD mice. However, tau pathology is not evident at this early stage of the disease in this mouse model. In this study, the effects of scFv-h3D6 on Aβ and tau pathologies have been assessed in 22-month-old 3xTg-AD mice. Briefly, 3xTg-AD female mice were treated for 2 weeks with scFv-h3D6 and compared with 3xTg-AD and non-transgenic (NTg) mice treated with PBS. The treatment with scFv-h3D6 was unequivocally effective in reducing the area of Aβ staining. Furthermore, a tendency for a reduction in tau levels was also observed after treatment that points to the interplay between Aβ and tau pathologies. The pro-inflammatory state observed in the 3xTg-AD mice did not progress after scFv-h3D6 treatment. In addition, the treatment did not alter the levels of apolipoprotein E or apolipoprotein J. Thus, a 2-week treatment with scFv-h3D6 was able to reduce AD-like pathology in elderly 3xTg-AD female mice.
Collapse
Affiliation(s)
- Alejandro R. Roda
- Protein Design and Immunotherapy Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain; (A.R.R.); (L.M.-G.); (G.S.-M.)
| | - Laia Montoliu-Gaya
- Protein Design and Immunotherapy Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain; (A.R.R.); (L.M.-G.); (G.S.-M.)
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, 431 41 Mölndal, Sweden
| | - Gabriel Serra-Mir
- Protein Design and Immunotherapy Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain; (A.R.R.); (L.M.-G.); (G.S.-M.)
| | - Sandra Villegas
- Protein Design and Immunotherapy Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain; (A.R.R.); (L.M.-G.); (G.S.-M.)
| |
Collapse
|
9
|
Güell-Bosch J, Lope-Piedrafita S, Esquerda-Canals G, Montoliu-Gaya L, Villegas S. Progression of Alzheimer's disease and effect of scFv-h3D6 immunotherapy in the 3xTg-AD mouse model: An in vivo longitudinal study using Magnetic Resonance Imaging and Spectroscopy. NMR IN BIOMEDICINE 2020; 33:e4263. [PMID: 32067292 DOI: 10.1002/nbm.4263] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 12/19/2019] [Accepted: 01/11/2020] [Indexed: 06/10/2023]
Abstract
Alzheimer's disease (AD) is an incurable disease that affects most of the 47 million people estimated as living with dementia worldwide. The main histopathological hallmarks of AD are extracellular β-amyloid (Aβ) plaques and intracellular neurofibrillary tangles (NFTs) composed of hyperphosphorylated tau protein. In recent years, Aβ-immunotherapy has been revealed as a potential tool in AD treatment. One strategy consists of using single-chain variable fragments (scFvs), which avoids the fragment crystallizable (Fc) effects that are supposed to trigger a microglial response, leading to microhemorrhages and vasogenic edemas, as evidenced in clinical trials with bapineuzumab. The scFv-h3D6 generated by our research group derives from this monoclonal antibody, which targets the N-terminal of the Aβ peptide and recognizes monomers, oligomers and fibrils. In this study, 3xTg-AD mice were intraperitoneally and monthly treated with 100 μg of scFv-h3D6 (a dose of ~3.3 mg/kg) or PBS, from 5 to 12 months of age (-mo), the age at which the mice were sacrificed and samples collected for histological and biochemical analyses. During treatments, four monitoring sessions using magnetic resonance imaging and spectroscopy (MRI/MRS) were performed at 5, 7, 9, and 12 months of age. MRI/MRS techniques are widely used in both human and mouse research, allowing to draw an in vivo picture of concrete aspects of the pathology in a non-invasive manner and allowing to monitor its development across time. Compared with the genetic background, 3xTg-AD mice presented a smaller volume in almost all cerebral regions and ages examined, an increase in both the intra and extracellular Aβ1-42 at 12-mo, and an inflammation process at this age, in both the hippocampus (IL-6 and mIns) and cortex (IL-6). In addition, treatment with scFv-h3D6 partially recovered the values in brain volume, and Aβ, IL-6, and mIns concentrations, among others, encouraging further studies with this antibody fragment.
Collapse
Affiliation(s)
- J Güell-Bosch
- Protein Design and Immunotherapy Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| | - S Lope-Piedrafita
- Servei de Ressonància Magnètica Nuclear, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
- Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - G Esquerda-Canals
- Protein Design and Immunotherapy Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| | - L Montoliu-Gaya
- Protein Design and Immunotherapy Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| | - S Villegas
- Protein Design and Immunotherapy Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| |
Collapse
|
10
|
Esquerda-Canals G, Martí-Clúa J, Villegas S. Pharmacokinetic parameters and mechanism of action of an efficient anti-Aβ single chain antibody fragment. PLoS One 2019; 14:e0217793. [PMID: 31150495 PMCID: PMC6544282 DOI: 10.1371/journal.pone.0217793] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 05/17/2019] [Indexed: 12/16/2022] Open
Abstract
The success of the targeting of amyloid-β (Aβ) oligomers through immunotherapy in Alzheimer’s disease (AD) mouse models has not been translated into the clinics. The use of single-chain variable fragments (scFvs) has been proposed to prevent the potential severe effects of full-length mAbs by precluding crystallizable fraction-mediated microglia activation. The efficacy of scFv-h3D6, a bapineuzumab-derived anti-Aβ scFv, has been extensively proven. In this work, we compared scFv-h3D6-EL, an elongated variant of the scFv-h3D6, with its original version to assess whether its characteristic higher thermodynamic stability improved its pharmacokinetic parameters. Although scFv-h3D6-EL had a longer half-life than its original version, its absorption from the peritoneal cavity into the systemic compartment was lower than that of the original version. Moreover, we attempted to determine the mechanism underlying the protective effect of scFv-h3D6. We found that scFv-h3D6 showed compartmental distribution and more interestingly crossed the blood–brain barrier. In the brain, scFv-h3D6 was engulfed by glial cells or internalized by Aβ peptide-containing neurons in the early phase post-injection, and was colocalized with the Aβ peptide almost exclusively in glial cells in the late phase post-injection. Aβ peptide levels in the brain decreased simultaneously with an increase in scFv-h3D6 levels. This observation in addition to the increased tumor necrosis factor-α levels in the late phase post-injection suggested that the engulfment of Aβ peptide/scFv-h3D6 complex extruded from large neurons by phagocytic cells was the mechanism underlying Aβ peptide withdrawal. The mechanism of action of scFv-h3D6 demonstrates the effectivity of Aβ-immunotherapy and lays the background for other studies focused on the finding of a treatment for AD.
Collapse
Affiliation(s)
- Gisela Esquerda-Canals
- Protein Design and Immunotherapy Group, Departament de Bioquímica i Biologia Molecular, Unitat de Biociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Unitat de Citologia i d’Histologia, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Joaquim Martí-Clúa
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Unitat de Citologia i d’Histologia, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Sandra Villegas
- Protein Design and Immunotherapy Group, Departament de Bioquímica i Biologia Molecular, Unitat de Biociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
- * E-mail:
| |
Collapse
|
11
|
Pozzi S, Thammisetty SS, Codron P, Rahimian R, Plourde KV, Soucy G, Bareil C, Phaneuf D, Kriz J, Gravel C, Julien JP. Virus-mediated delivery of antibody targeting TAR DNA-binding protein-43 mitigates associated neuropathology. J Clin Invest 2019; 129:1581-1595. [PMID: 30667370 DOI: 10.1172/jci123931] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 01/15/2019] [Indexed: 12/12/2022] Open
Abstract
The cytoplasmic aggregation of TAR DNA-binding protein-43 (TDP-43) is a hallmark of degenerating neurons in amyotrophic lateral sclerosis (ALS) and subsets of frontotemporal dementia (FTD). In order to reduce TDP-43 pathology, we generated single-chain (scFv) antibodies against the RNA recognition motif 1 (RRM1) of TDP-43, which is involved in abnormal protein self-aggregation and interaction with p65 NF-κB. Virus-mediated delivery into the nervous system of a scFv antibody, named VH7Vk9, reduced microgliosis in a mouse model of acute neuroinflammation and mitigated cognitive impairment, motor defects, TDP-43 proteinopathy, and neuroinflammation in transgenic mice expressing ALS-linked TDP-43 mutations. These results suggest that antibodies targeting the RRM1 domain of TDP-43 might provide new therapeutic avenues for the treatment of ALS and FTD.
Collapse
Affiliation(s)
- Silvia Pozzi
- CERVO Brain Research Centre, Québec, Québec, Canada
| | | | - Philippe Codron
- CERVO Brain Research Centre, Québec, Québec, Canada.,MITOVASC Institute, Centre National de la Recherche Scientifique (CNRS) 6015, INSERM U1083, University of Angers, Angers, France
| | | | | | | | | | | | - Jasna Kriz
- CERVO Brain Research Centre, Québec, Québec, Canada.,Department of Psychiatry and Neuroscience, Université Laval, Québec City, Québec, Canada
| | - Claude Gravel
- CERVO Brain Research Centre, Québec, Québec, Canada.,Department of Psychiatry and Neuroscience, Université Laval, Québec City, Québec, Canada
| | - Jean-Pierre Julien
- CERVO Brain Research Centre, Québec, Québec, Canada.,Department of Psychiatry and Neuroscience, Université Laval, Québec City, Québec, Canada
| |
Collapse
|
12
|
Hoxha E, Lippiello P, Zurlo F, Balbo I, Santamaria R, Tempia F, Miniaci MC. The Emerging Role of Altered Cerebellar Synaptic Processing in Alzheimer's Disease. Front Aging Neurosci 2018; 10:396. [PMID: 30542279 PMCID: PMC6278174 DOI: 10.3389/fnagi.2018.00396] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 11/15/2018] [Indexed: 11/13/2022] Open
Abstract
The role of the cerebellum in Alzheimer's disease (AD) has been neglected for a long time. Recent studies carried out using transgenic mouse models have demonstrated that amyloid-β (Aβ) is deposited in the cerebellum and affects synaptic transmission and plasticity, sometimes before plaque formation. A wide variability of motor phenotype has been observed in the different murine models of AD, without a consistent correlation with the extent of cerebellar histopathological changes or with cognitive deficits. The loss of noradrenergic drive may contribute to the impairment of cerebellar synaptic function and motor learning observed in these mice. Furthermore, cerebellar neurons, particularly granule cells, have been used as in vitro model of Aβ-induced neuronal damage. An unexpected conclusion is that the cerebellum, for a long time thought to be somehow protected from AD pathology, is actually considered as a region vulnerable to Aβ toxic damage, even at the early stage of the disease, with consequences on motor performance.
Collapse
Affiliation(s)
- Eriola Hoxha
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Turin, Italy.,Department of Neuroscience, University of Torino, Turin, Italy
| | - Pellegrino Lippiello
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Fabio Zurlo
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Ilaria Balbo
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Turin, Italy.,Department of Neuroscience, University of Torino, Turin, Italy
| | - Rita Santamaria
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Filippo Tempia
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Turin, Italy.,Department of Neuroscience, University of Torino, Turin, Italy.,National Institute of Neuroscience (INN), Turin, Italy
| | - Maria Concetta Miniaci
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| |
Collapse
|
13
|
Montoliu-Gaya L, Güell-Bosch J, Esquerda-Canals G, Roda AR, Serra-Mir G, Lope-Piedrafita S, Sánchez-Quesada JL, Villegas S. Differential effects of apoE and apoJ mimetic peptides on the action of an anti-Aβ scFv in 3xTg-AD mice. Biochem Pharmacol 2018; 155:380-392. [PMID: 30026023 DOI: 10.1016/j.bcp.2018.07.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 07/13/2018] [Indexed: 12/31/2022]
Abstract
Anti-Aβ immunotherapy has emerged as a promising approach to treat Alzheimer's disease (AD). The single-chain variable fragment scFv-h3D6 is an anti-Aβ antibody fragment that lacks the Fc region, which is associated with the induction of microglial reactivity by the full-length monoclonal antibody bapineuzumab. ScFv-h3D6 was previously shown to restore the levels of apolipoprotein E (apoE) and apolipoprotein J (apoJ) in a triple-transgenic-AD (3xTg-AD) mouse model. Since apoE and apoJ play an important role in the development of AD, we aimed to study the in vivo effect of the combined therapy of scFv-h3D6 with apoE and apoJ mimetic peptides (MPs). Four-and-a-half-month-old 3xTg-AD mice were treated for six weeks with scFv-h3D6, apoE-MP, apoJ-MP, or a combination of scFv-h3D6 with each of the MPs, or a vehicle, and then the results were compared to non-transgenic mice. Magnetic Resonance Imaging showed a general tendency of the different treatments to protect against the reduction in brain volume. Aβ burden decreased after treatment with scFv-h3D6, apoE-MP, or apoJ-MP, but the effect was not as evident with the combined therapies. In terms of glial reactivity, apoE-MP showed a potent anti-inflammatory effect that was eased by the presence of scFv-h3D6, whereas the combination of apoJ-MP and scFv-h3D6 was not detrimental. ScFv-h3D6 alone did not induce microglial reactivity, as full-length antibodies do; rather, it reduced it. Endogenous apoE and apoJ levels were decreased by scFv-h3D6, but the MPs lead to a simultaneous increase of both apolipoproteins. While apoE-MP and apoJ-MP demonstrated different effects in the combined therapies with scFv-h3D6, they did not improve the overall protective effect of scFv-h3D6 in reducing the Aβ burden, apolipoproteins levels or microglial reactivity.
Collapse
Affiliation(s)
- Laia Montoliu-Gaya
- Protein Folding and Stability Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain.
| | - Jofre Güell-Bosch
- Protein Folding and Stability Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain.
| | - Gisela Esquerda-Canals
- Protein Folding and Stability Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain.
| | - Alejandro R Roda
- Protein Folding and Stability Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain.
| | - Gabriel Serra-Mir
- Protein Folding and Stability Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain.
| | - Silvia Lope-Piedrafita
- Servei de Ressonància Magnètica Nuclear, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain; Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain.
| | - Jose Luis Sánchez-Quesada
- Cardiovascular Biochemistry Group, Research Institute of the Hospital de Sant Pau (IIB Sant Pau), Barcelona, Spain; CIBER of Diabetes and Metabolic Diseases (CIBERDEM), Spain.
| | - Sandra Villegas
- Protein Folding and Stability Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain.
| |
Collapse
|
14
|
Montoliu-Gaya L, Mulder SD, Herrebout MA, Baayen JC, Villegas S, Veerhuis R. Aβ-oligomer uptake and the resulting inflammatory response in adult human astrocytes are precluded by an anti-Aβ single chain variable fragment in combination with an apoE mimetic peptide. Mol Cell Neurosci 2018; 89:49-59. [DOI: 10.1016/j.mcn.2018.03.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 03/30/2018] [Accepted: 03/31/2018] [Indexed: 10/17/2022] Open
|
15
|
Manoutcharian K, Perez-Garmendia R, Gevorkian G. Recombinant Antibody Fragments for Neurodegenerative Diseases. Curr Neuropharmacol 2018; 15:779-788. [PMID: 27697033 PMCID: PMC5771054 DOI: 10.2174/1570159x01666160930121647] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 09/04/2016] [Accepted: 09/28/2016] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Recombinant antibody fragments are promising alternatives to full-length immunoglobulins and offer important advantages compared with conventional monoclonal antibodies: extreme specificity, higher affinity, superior stability and solubility, reduced immunogenicity as well as easy and inexpensive large-scale production. OBJECTIVE In this article we will review and discuss recombinant antibodies that are being evaluated for neurodegenerative diseases in pre-clinical models and in clinical studies and will summarize new strategies that are being developed to optimize their stability, specificity and potency for advancing their use. METHODS Articles describing recombinant antibody fragments used for neurological diseases were selected (PubMed) and evaluated for their significance. RESULTS Different antibody formats such as single-chain fragment variable (scFv), single-domain antibody fragments (VHHs or sdAbs), bispecific antibodies (bsAbs), intrabodies and nanobodies, are currently being studied in pre-clinical models of cancer as well as infectious and autoimmune diseases and many of them are being tested as therapeutics in clinical trials. Immunotherapy approaches have shown therapeutic efficacy in several animal models of Alzheimer´s disease (AD), Parkinson disease (PD), dementia with Lewy bodies (DLB), frontotemporal dementia (FTD), Huntington disease (HD), transmissible spongiform encephalopathies (TSEs) and multiple sclerosis (MS). It has been demonstrated that recombinant antibody fragments may neutralize toxic extra- and intracellular misfolded proteins involved in the pathogenesis of AD, PD, DLB, FTD, HD or TSEs and may target toxic immune cells participating in the pathogenesis of MS. CONCLUSION Recombinant antibody fragments represent a promising tool for the development of antibody-based immunotherapeutics for neurodegenerative diseases.
Collapse
Affiliation(s)
- Karen Manoutcharian
- Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de Mexico (UNAM), Mexico DF. Mexico
| | - Roxanna Perez-Garmendia
- Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de Mexico (UNAM), Mexico DF. Mexico
| | - Goar Gevorkian
- Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de Mexico (UNAM), Apartado Postal 70228, Cuidad Universitaria, Mexico DF, CP 04510, Mexico. 0
| |
Collapse
|
16
|
Montoliu-Gaya L, Mulder SD, Veerhuis R, Villegas S. Effects of an Aβ-antibody fragment on Aβ aggregation and astrocytic uptake are modulated by apolipoprotein E and J mimetic peptides. PLoS One 2017; 12:e0188191. [PMID: 29155887 PMCID: PMC5695774 DOI: 10.1371/journal.pone.0188191] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Accepted: 11/02/2017] [Indexed: 01/23/2023] Open
Abstract
Aβ-Immunotherapy has long been studied in the treatment of Alzheimer’s disease (AD), but not how other molecules involved in the disease can affect antibody performance. We previously designed an antibody fragment, scFv-h3D6, and showed that it precludes Aβ-induced cytotoxicity by withdrawing Aβ oligomers from the amyloid pathway towards a non-toxic, worm-like pathway. ScFv-h3D6 was effective at the behavioral, cellular, and molecular levels in the 3xTg-AD mouse model. Because scFv-h3D6 treatment restored apolipoprotein E (apoE) and J (apoJ) concentrations to non-pathological values, and Aβ internalization by glial cells was found to be decreased in the presence of these apolipoproteins, we now aimed to test the influence of scFv-h3D6 on Aβ aggregation and cellular uptake by primary human astrocytes in the presence of therapeutic apoE and apoJ mimetic peptides (MPs). Firstly, we demonstrated by CD and FTIR that the molecules used in this work were well folded. Next, interactions between apoE or apoJ-MP, scFv-h3D6 and Aβ were studied by CD. The conformational change induced by the interaction of Aβ with apoE-MP was much bigger than the induced with apoJ-MP, in line with the observed formation of protective worm-like fibrils by the scFv-h3D6/Aβ complex in the presence of apoJ-MP, but not of apoE-MP. ScFv-h3D6, apoJ-MP, and apoE-MP to a different extent reduced Aβ uptake by astrocytes, and apoE-MP partially interfered with the dramatic reduction by scFv-h3D6 while apoJ-MP had no effect on scFv-h3D6 action. As sustained Aβ uptake by astrocytes may impair their normal functions, and ultimately neuronal viability, this work shows another beneficence of scFv-h3D6 treatment, which is not further improved by the use of apoE or apoJ mimetic peptides.
Collapse
Affiliation(s)
- Laia Montoliu-Gaya
- Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Sandra D. Mulder
- Clinical Chemistry Department, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The Netherlands
| | - Robert Veerhuis
- Clinical Chemistry Department, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The Netherlands
- Psychiatry Department, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The Netherlands
- * E-mail: (RV); (SV)
| | - Sandra Villegas
- Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
- * E-mail: (RV); (SV)
| |
Collapse
|
17
|
Montoliu-Gaya L, Murciano-Calles J, Martinez JC, Villegas S. Towards the improvement in stability of an anti-Aβ single-chain variable fragment, scFv-h3D6, as a way to enhance its therapeutic potential. Amyloid 2017; 24:167-175. [PMID: 28699800 DOI: 10.1080/13506129.2017.1348347] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
ScFv-h3D6 is a single-chain variable fragment derived from the monoclonal antibody bapineuzumab that prevents Aβ-induced cytotoxicity by capturing Aβ oligomers. The benefits of scFv-h3D6 treatment in Alzheimer's disease are known at the behavioural, cellular and molecular levels in the 3xTg-AD mouse model. Antibody-based therapeutics are only stable in a limited temperature range, so their benefit in vivo depends on their capability for maintaining the proper fold. Here, we have stabilized the scFv-h3D6 folding by introducing the mutation VH-K64R and combining it with the previously described elongation of the VL domain (C3). The stabilities of the different scFv-h3D6 constructs were calculated from urea and thermal denaturation followed by Trp-fluorescence, CD and DSC and resulted in the order C3 > K64R/C3 > VH-K64R ≥ scFv-h3D6; showing that the combination of both mutations was not additive, instead they partially cancelled each other. The three mutants assayed showed a decreased aggregation tendency but maintained their capability to aggregate in the form of worm-like fibrils, basis of the protective effect of scFv-h3D6. Cytotoxicity assays showed that all the mutants recovered cell viability of Aβ-treated neuroblastoma cell cultures in a dose-dependent manner and with efficiencies that correlated with stability, therefore improving the therapeutic ability of this antibody.
Collapse
Affiliation(s)
- Laia Montoliu-Gaya
- a Protein Folding and Stability Group, Departament de Bioquímica i Biologia Molecular , Unitat de Biociències, Universitat Autònoma de Barcelona , Spain
| | - Javier Murciano-Calles
- b Department of Physical Chemistry and Institute of Biotechnology, Faculty of Sciences , University of Granada, Campus Fuentenueva , Granada , Spain
| | - Jose C Martinez
- b Department of Physical Chemistry and Institute of Biotechnology, Faculty of Sciences , University of Granada, Campus Fuentenueva , Granada , Spain
| | - Sandra Villegas
- a Protein Folding and Stability Group, Departament de Bioquímica i Biologia Molecular , Unitat de Biociències, Universitat Autònoma de Barcelona , Spain
| |
Collapse
|
18
|
Montoliu-Gaya L, Esquerda-Canals G, Bronsoms S, Villegas S. Production of an anti-Aβ antibody fragment in Pichia pastoris and in vitro and in vivo validation of its therapeutic effect. PLoS One 2017; 12:e0181480. [PMID: 28771492 PMCID: PMC5542431 DOI: 10.1371/journal.pone.0181480] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 06/30/2017] [Indexed: 12/31/2022] Open
Abstract
ScFv-h3D6 has been shown as an efficient therapy in the 3xTg-AD mouse model of Alzheimer's Disease. Because one of the major bottlenecks for the therapeutic uses of proteins produced in Escherichia coli is their potential contamination with endotoxins, LPS were extensively removed by a rather low-efficient, expensive, and time-consuming purification step. In addition, disulfide scrambling is favored in the reducing bacterial cytoplasm albeit the use of reductase deficient strains. To overcome these hurdles, as well as to improve the yield, the yeast Pichia pastoris, an endotoxin-free host system for recombinant protein production, has been used to produce scFv-h3D6, both in flask and in a fed-batch bioreactor. Comparison of the thermal stability of the obtained protein with that from E. coli showed no differences. Opposite to the case of the protein obtained from E. coli, no disulfide scrambled conformations or LPS traces were detected in that produced in P. pastoris. Cytotoxicity assays in SH-SY5Y neuroblastoma cell-cultures demonstrated that proteins from both expression systems were similarly efficient in precluding Aβ-induced toxicity. Finally, the 3xTg-AD mouse model was used to test the therapeutic effect of both proteins. Quantification of Aβ levels from cortex and hippocampus protein extracts by ELISA, and Aβ-immunohistochemistry, showed that both proteins reduced Aβ burden. This work demonstrates that scFv-h3D6 obtained from P. pastoris shows the same benefits as those already known for that obtained from E. coli, with multiple advantages in terms of recombinant production and safety.
Collapse
Affiliation(s)
- Laia Montoliu-Gaya
- Protein Folding and Stability Group, Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Gisela Esquerda-Canals
- Protein Folding and Stability Group, Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
- Departament de Biologia Cel·lular, de Fisiologia i d’Immunologia, Unitat de Citologia i d’Histologia, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Silvia Bronsoms
- Servei de Proteòmica i Biologia Estructural, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Sandra Villegas
- Protein Folding and Stability Group, Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| |
Collapse
|
19
|
Esquerda-Canals G, Martí-Clúa J, Roda AR, Villegas S. An Intracellular Amyloid-β/AβPP Epitope Correlates with Neurodegeneration in those Neuronal Populations Early Involved in Alzheimer’s Disease. J Alzheimers Dis 2017; 59:1079-1096. [DOI: 10.3233/jad-170218] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- Gisela Esquerda-Canals
- Departament de Bioquímica i Biologia Molecular, Protein Folding and Stability Group, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
- Departament de Biologia Cellular, Protein Folding and Stability Group, de Fisiologia i d’Immunologia, Unitat de Citologia i d’Histologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Joaquim Martí-Clúa
- Departament de Biologia Cellular, Protein Folding and Stability Group, de Fisiologia i d’Immunologia, Unitat de Citologia i d’Histologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Alejandro R. Roda
- Departament de Bioquímica i Biologia Molecular, Protein Folding and Stability Group, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Sandra Villegas
- Departament de Bioquímica i Biologia Molecular, Protein Folding and Stability Group, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| |
Collapse
|
20
|
Montoliu-Gaya L, Martínez JC, Villegas S. Understanding the contribution of disulfide bridges to the folding and misfolding of an anti-Aβ scFv. Protein Sci 2017; 26:1138-1149. [PMID: 28340507 DOI: 10.1002/pro.3164] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 03/20/2017] [Accepted: 03/21/2017] [Indexed: 12/13/2022]
Abstract
ScFv-h3D6 is a single chain variable fragment that precludes Aβ peptide-induced cytotoxicity by withdrawing Aβ oligomers from the amyloid pathway to the worm-like pathway. Production of scFv molecules is not a straightforward procedure because of the occurrence of disulfide scrambled conformations generated in the refolding process. Here, we separately removed the disulfide bond of each domain and solved the scrambling problem; and then, we intended to compensate the loss of thermodynamic stability by adding three C-terminal elongation mutations, previously described to stabilize the native fold of scFv-h3D6. Such stabilization occurred through stabilization of the intermediate state in the folding pathway and destabilization of a different, β-rich, intermediate state driving to worm-like fibrils. Elimination of the disulfide bridge of the less stable domain, VL , deeply compromised the yield and increased the aggregation tendency, but elimination of the disulfide bridge of the more stable domain, VH , solved the scrambling problem and doubled the production yield. Notably, it also changed the aggregation pathway from the protective worm-like morphology to an amyloid one. This was so because a partially unfolded intermediate driving to amyloid aggregation was present, instead of the β-rich intermediate driving to worm-like fibrils. When combining with the elongation mutants, stabilization of the partially unfolded intermediate driving to amyloid fibrils was the only effect observed. Therefore, the same mutations drove to completely different scenarios depending on the presence of disulfide bridges and this illustrates the relevance of such linkages in the stability of different intermediate states for folding and misfolding.
Collapse
Affiliation(s)
- Laia Montoliu-Gaya
- Protein Folding and Stability Group, Departament de Bioquímica i Biologia Molecular, Unitat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Jose C Martínez
- Department of Physical Chemistry and Institute of Biotechnology, Faculty of Sciences, University of Granada, Granada, Spain
| | - Sandra Villegas
- Protein Folding and Stability Group, Departament de Bioquímica i Biologia Molecular, Unitat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Spain
| |
Collapse
|
21
|
Aβ-Immunotherapeutic strategies: a wide range of approaches for Alzheimer's disease treatment. Expert Rev Mol Med 2016; 18:e13. [PMID: 27357999 DOI: 10.1017/erm.2016.11] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Current therapies to treat Alzheimer's disease (AD) are focused on ameliorating symptoms instead of treating the underlying causes of AD. The accumulation of amyloid β (Aβ) oligomers, whether by an increase in production or by a decrease in clearance, has been described as the seed that initiates the pathological cascade in AD. Developing therapies to target these species is a vital step in improving AD treatment. Aβ-immunotherapy, especially passive immunotherapy, is a promising approach to reduce the Aβ burden. Up to now, several monoclonal antibodies (mAbs) have been tested in clinical trials on humans, but none of them have passed Phase III. In all likelihood, these trials failed mainly because patients with mild-to-moderate AD were recruited, and thus treatment may have been too late to be effective. Therefore, many ongoing clinical trials are being conducted in patients at the prodromal stage. New structures based on antibody fragments have been engineered intending to improve efficacy and safety. This review presents the properties of this variety of developing treatments and provides a perspective on state-of-the-art of passive Aβ-immunotherapy in AD.
Collapse
|
22
|
Congdon EE, Krishnaswamy S, Sigurdsson EM. Harnessing the immune system for treatment and detection of tau pathology. J Alzheimers Dis 2015; 40 Suppl 1:S113-21. [PMID: 24603943 DOI: 10.3233/jad-132435] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The tau protein is an attractive target for therapy and diagnosis. We started a tau immunotherapy program about 13 years ago and have since demonstrated that active and passive immunotherapies diminish tau pathology and improve function, including cognition, in different mouse models. These findings have been confirmed and extended by several groups. We routinely detect neuronal, and to a lesser extent microglial, antibody uptake correlating with tau pathology. Antibodies bind tau aggregates in the endosomal/lysosomal system, enhancing clearance presumably by promoting their disassembly. Extracellular clearance has recently been shown by others, using antibodies that apparently are not internalized. As most pathological tau is neuronal, intracellular targeting may be more efficacious. However, extracellular tau may be more accessible to antibodies, with tau-antibody complexes a target for microglial phagocytosis. The extent of involvement of each pathway may depend on numerous factors including antibody properties, degree of pathology, and experimental model. On the imaging front, multiple tau ligands derived from β-sheet dyes have been developed by several groups, some with promising results in clinical PET tests. Postmortem analysis should clarify their tau specificity, as in theory and based on histological staining, those are likely to have some affinity for various amyloids. We are developing antibody-derived tau probes that should be more specific, and have in mouse models shown in vivo detection and binding to pathological tau after peripheral injection. These are exciting times for research on tau therapies and diagnostic agents that hopefully can be applied to humans in the near future.
Collapse
Affiliation(s)
- Erin E Congdon
- Departments of Neuroscience and Physiology, New York University School of Medicine, NY, USA
| | | | - Einar M Sigurdsson
- Departments of Neuroscience and Physiology, New York University School of Medicine, NY, USA Departments of Psychiatry, New York University School of Medicine, NY, USA
| |
Collapse
|
23
|
[Alzheimer's disease: New therapeutic strategies]. Med Clin (Barc) 2014; 145:76-83. [PMID: 25245784 DOI: 10.1016/j.medcli.2014.05.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 05/09/2014] [Accepted: 05/15/2014] [Indexed: 11/21/2022]
Abstract
The rapid increase in prevalence rates of Alzheimer's disease means that treatments to prevent, stop or reverse this devastating disease are urgently needed. Despite advances in understanding its molecular pathology, there are no drugs that can halt its progression. This review takes a tour through phase 2, or higher studies, probing receptor agonist agents interfering with aggregation, inhibitors/modulators of secretases, lipid-lowering agents, and, finally and most extensively, immunotherapy. The fact that phase 3 studies with bapineuzumab and solaneuzumab have recently failed does not invalidate the potential of immunotherapy, as more information is available and new clinical trials are being initiated.
Collapse
|