1
|
Niazi SK, Mariam Z, Magoola M. Engineered Antibodies to Improve Efficacy against Neurodegenerative Disorders. Int J Mol Sci 2024; 25:6683. [PMID: 38928395 PMCID: PMC11203520 DOI: 10.3390/ijms25126683] [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: 05/11/2024] [Revised: 06/09/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Antibodies that can selectively remove rogue proteins in the brain are an obvious choice to treat neurodegenerative disorders (NDs), but after decades of efforts, only two antibodies to treat Alzheimer's disease are approved, dozens are in the testing phase, and one was withdrawn, and the other halted, likely due to efficacy issues. However, these outcomes should have been evident since these antibodies cannot enter the brain sufficiently due to the blood-brain barrier (BBB) protectant. However, all products can be rejuvenated by binding them with transferrin, preferably as smaller fragments. This model can be tested quickly and at a low cost and should be applied to bapineuzumab, solanezumab, crenezumab, gantenerumab, aducanumab, lecanemab, donanemab, cinpanemab, and gantenerumab, and their fragments. This paper demonstrates that conjugating with transferrin does not alter the binding to brain proteins such as amyloid-β (Aβ) and α-synuclein. We also present a selection of conjugate designs that will allow cleavage upon entering the brain to prevent their exocytosis while keeping the fragments connected to enable optimal binding to proteins. The identified products can be readily tested and returned to patients with the lowest regulatory cost and delays. These engineered antibodies can be manufactured by recombinant engineering, preferably by mRNA technology, as a more affordable solution to meet the dire need to treat neurodegenerative disorders effectively.
Collapse
Affiliation(s)
| | - Zamara Mariam
- Centre for Health and Life Sciences, Coventry University, Coventry City CV1 5FB, UK;
| | | |
Collapse
|
2
|
Vandendriessche C, Bruggeman A, Foroozandeh J, Van Hoecke L, Dujardin P, Xie J, Van Imschoot G, Van Wonterghem E, Castelein J, Lucci C, De Groef L, Vandenbroucke RE. The Spreading and Effects of Human Recombinant α-Synuclein Preformed Fibrils in the Cerebrospinal Fluid of Mice. eNeuro 2024; 11:ENEURO.0024-23.2024. [PMID: 38383588 PMCID: PMC10925901 DOI: 10.1523/eneuro.0024-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 01/12/2024] [Accepted: 01/17/2024] [Indexed: 02/23/2024] Open
Abstract
Parkinson's disease (PD) patients harbor seeding-competent α-synuclein (α-syn) in their cerebrospinal fluid (CSF), which is mainly produced by the choroid plexus (ChP). Nonetheless, little is known about the role of the CSF and the ChP in PD pathogenesis. To address this question, we used an intracerebroventricular (icv) injection mouse model to assess CSF α-syn spreading and its short- and long-term consequences on the brain. Hereby, we made use of seeding-competent, recombinant α-syn preformed fibrils (PFF) that are known to induce aggregation and subsequent spreading of endogenous α-syn in stereotactic tissue injection models. Here, we show that icv-injected PFF, but not monomers (Mono), are rapidly removed from the CSF by interaction with the ChP. Additionally, shortly after icv injection both Mono and PFF were detected in the olfactory bulb and striatum. This spreading was associated with increased inflammation and complement activation in these tissues as well as leakage of the blood-CSF barrier. Despite these effects, a single icv injection of PFF didn't induce a decline in motor function. In contrast, daily icv injections over the course of 5 days resulted in deteriorated grip strength and formation of phosphorylated α-syn inclusions in the brain 2 months later, whereas dopaminergic neuron levels were not affected. These results point toward an important clearance function of the CSF and the ChP, which could mediate removal of PFF from the brain, whereby chronic exposure to PFF in the CSF may negatively impact blood-CSF barrier functionality and PD pathology.
Collapse
Affiliation(s)
- Charysse Vandendriessche
- VIB Center for Inflammation Research, VIB, 9000, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9000, Ghent, Belgium
| | - Arnout Bruggeman
- VIB Center for Inflammation Research, VIB, 9000, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9000, Ghent, Belgium
- Department of Neurology, Ghent University Hospital, 9000, Ghent, Belgium
| | - Joyce Foroozandeh
- VIB Center for Inflammation Research, VIB, 9000, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9000, Ghent, Belgium
- VIB Center for Brain & Disease Research, VIB, 3000, Leuven, Belgium
- Department of Neurosciences, Brain Institute KU Leuven, 3000, Leuven, Belgium
| | - Lien Van Hoecke
- VIB Center for Inflammation Research, VIB, 9000, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9000, Ghent, Belgium
| | - Pieter Dujardin
- VIB Center for Inflammation Research, VIB, 9000, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9000, Ghent, Belgium
| | - Junhua Xie
- VIB Center for Inflammation Research, VIB, 9000, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9000, Ghent, Belgium
| | - Griet Van Imschoot
- VIB Center for Inflammation Research, VIB, 9000, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9000, Ghent, Belgium
| | - Elien Van Wonterghem
- VIB Center for Inflammation Research, VIB, 9000, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9000, Ghent, Belgium
| | - Jonas Castelein
- VIB Center for Inflammation Research, VIB, 9000, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9000, Ghent, Belgium
| | - Cristiano Lucci
- Cellular Communication and Neurodegeneration Research Group, Department of Biology, Leuven Brain Institute, KU Leuven, 3000, Leuven, Belgium
| | - Lies De Groef
- Cellular Communication and Neurodegeneration Research Group, Department of Biology, Leuven Brain Institute, KU Leuven, 3000, Leuven, Belgium
| | - Roosmarijn E Vandenbroucke
- VIB Center for Inflammation Research, VIB, 9000, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9000, Ghent, Belgium
| |
Collapse
|
3
|
Loeffler DA. Antibody-Mediated Clearance of Brain Amyloid-β: Mechanisms of Action, Effects of Natural and Monoclonal Anti-Aβ Antibodies, and Downstream Effects. J Alzheimers Dis Rep 2023; 7:873-899. [PMID: 37662616 PMCID: PMC10473157 DOI: 10.3233/adr-230025] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 07/05/2023] [Indexed: 09/05/2023] Open
Abstract
Immunotherapeutic efforts to slow the clinical progression of Alzheimer's disease (AD) by lowering brain amyloid-β (Aβ) have included Aβ vaccination, intravenous immunoglobulin (IVIG) products, and anti-Aβ monoclonal antibodies. Neither Aβ vaccination nor IVIG slowed disease progression. Despite conflicting phase III results, the monoclonal antibody Aducanumab received Food and Drug Administration (FDA) approval for treatment of AD in June 2021. The only treatments unequivocally demonstrated to slow AD progression to date are the monoclonal antibodies Lecanemab and Donanemab. Lecanemab received FDA approval in January 2023 based on phase II results showing lowering of PET-detectable Aβ; phase III results released at that time indicated slowing of disease progression. Topline results released in May 2023 for Donanemab's phase III trial revealed that primary and secondary end points had been met. Antibody binding to Aβ facilitates its clearance from the brain via multiple mechanisms including promoting its microglial phagocytosis, activating complement, dissolving fibrillar Aβ, and binding of antibody-Aβ complexes to blood-brain barrier receptors. Antibody binding to Aβ in peripheral blood may also promote cerebral efflux of Aβ by a peripheral sink mechanism. According to the amyloid hypothesis, for Aβ targeting to slow AD progression, it must decrease downstream neuropathological processes including tau aggregation and phosphorylation and (possibly) inflammation and oxidative stress. This review discusses antibody-mediated mechanisms of Aβ clearance, findings in AD trials involving Aβ vaccination, IVIG, and anti-Aβ monoclonal antibodies, downstream effects reported in those trials, and approaches which might improve the Aβ-clearing ability of monoclonal antibodies.
Collapse
Affiliation(s)
- David A. Loeffler
- Beaumont Research Institute, Department of Neurology, Corewell Health, Royal Oak, MI, USA
| |
Collapse
|
4
|
Gu H, Xu Y, Du N, Yu Y, Zheng W, Du Y. Pb Induces MCP-1 in the Choroid Plexus. BIOLOGY 2022; 11:308. [PMID: 35205174 PMCID: PMC8869661 DOI: 10.3390/biology11020308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 02/06/2022] [Accepted: 02/10/2022] [Indexed: 11/16/2022]
Abstract
Lead (Pb) is an environmental element that has been implicated in the development of dementia and Alzheimer's disease (AD). Additionally, innate immune activation contributes to AD pathophysiology. However, the mechanisms involved remain poorly understood. The choroid plexus (CP) is not only the site of cerebrospinal fluid (CSF) production, but also an important location for communication between the circulation and the CSF. In this study, we investigated the involvement of the CP during Pb exposure by evaluating the expression of the monocyte chemoattractant protein-1 (MCP-1). MCP-1 is highly expressed in the CP compared to other CNS tissues. MCP-1 regulates macrophage infiltration and is upregulated in AD brains. Our study revealed that Pb exposure stimulated MCP-1 expression, along with a significantly increased macrophage infiltration into the CP. By using cultured Z310 rat CP cells, Pb exposure stimulated MCP-1 expression in a dose-related fashion and markedly activated both NF-κB and p38 MAP kinase. Interestingly, both SB 203580, a p38 inhibitor, and BAY 11-7082, an NF-κB p65 inhibitor, significantly blocked Pb-induced MCP-1 expression. However, SB203580 did not directly inhibit NF-κB p65 phosphorylation. In conclusion, Pb exposure stimulates MCP-1 expression via the p38 and NF-κB p65 pathways along with macrophage infiltration into the CP.
Collapse
Affiliation(s)
- Huiying Gu
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (H.G.); (Y.X.); (Y.Y.)
| | - Yundan Xu
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (H.G.); (Y.X.); (Y.Y.)
- School of Basic Medical Science, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Nicole Du
- Department of Pediatrics, Children’s National Hospital, Washington, DC 20010, USA;
| | - Yongqi Yu
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (H.G.); (Y.X.); (Y.Y.)
| | - Wei Zheng
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, USA;
| | - Yansheng Du
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (H.G.); (Y.X.); (Y.Y.)
| |
Collapse
|
5
|
Gião T, Teixeira T, Almeida MR, Cardoso I. Choroid Plexus in Alzheimer's Disease-The Current State of Knowledge. Biomedicines 2022; 10:224. [PMID: 35203434 PMCID: PMC8869376 DOI: 10.3390/biomedicines10020224] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/14/2022] [Accepted: 01/19/2022] [Indexed: 01/31/2023] Open
Abstract
The choroid plexus (CP), located in each of the four ventricles of the brain, is formed by a monolayer of epithelial cells that surrounds a highly vascularized connective tissue with permeable capillaries. These cells are joined by tight junctions forming the blood-cerebrospinal fluid barrier (BCSFB), which strictly regulates the exchange of substances between the blood and cerebrospinal fluid (CSF). The primary purpose of the CP is to secrete CSF, but it also plays a role in the immune surveillance of the central nervous system (CNS) and in the removal of neurotoxic compounds from the CSF. According to recent findings, the CP is also involved in the modulation of the circadian cycle and neurogenesis. In diseases such as Alzheimer's disease (AD), the function of the CP is impaired, resulting in an altered secretory, barrier, transport, and immune function. This review describes the current state of knowledge concerning the roles of the CP and BCSFB in the pathophysiology of AD and summarizes recently proposed therapies that aim to restore CP and BCSFB functions.
Collapse
Affiliation(s)
- Tiago Gião
- Molecular Neurobiology Group, i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (T.T.); (M.R.A.)
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Departamento de Biologia Molecular, ICBAS—Instituto de Ciências Biomédicas Abel Salazar, 4050-013 Porto, Portugal
| | - Tiago Teixeira
- Molecular Neurobiology Group, i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (T.T.); (M.R.A.)
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Faculdade de Medicina, Universidade do Porto, 4200-319 Porto, Portugal
| | - Maria Rosário Almeida
- Molecular Neurobiology Group, i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (T.T.); (M.R.A.)
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Departamento de Biologia Molecular, ICBAS—Instituto de Ciências Biomédicas Abel Salazar, 4050-013 Porto, Portugal
| | - Isabel Cardoso
- Molecular Neurobiology Group, i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (T.T.); (M.R.A.)
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Departamento de Biologia Molecular, ICBAS—Instituto de Ciências Biomédicas Abel Salazar, 4050-013 Porto, Portugal
| |
Collapse
|
6
|
Xie J, Gorlé N, Vandendriessche C, Van Imschoot G, Van Wonterghem E, Van Cauwenberghe C, Parthoens E, Van Hamme E, Lippens S, Van Hoecke L, Vandenbroucke RE. Low-grade peripheral inflammation affects brain pathology in the App NL-G-Fmouse model of Alzheimer's disease. Acta Neuropathol Commun 2021; 9:163. [PMID: 34620254 PMCID: PMC8499584 DOI: 10.1186/s40478-021-01253-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 09/01/2021] [Indexed: 12/22/2022] Open
Abstract
Alzheimer’s disease (AD) is a chronic neurodegenerative disease characterized by the accumulation of amyloid β (Aβ) and neurofibrillary tangles. The last decade, it became increasingly clear that neuroinflammation plays a key role in both the initiation and progression of AD. Moreover, also the presence of peripheral inflammation has been extensively documented. However, it is still ambiguous whether this observed inflammation is cause or consequence of AD pathogenesis. Recently, this has been studied using amyloid precursor protein (APP) overexpression mouse models of AD. However, the findings might be confounded by APP-overexpression artifacts. Here, we investigated the effect of low-grade peripheral inflammation in the APP knock-in (AppNL-G-F) mouse model. This revealed that low-grade peripheral inflammation affects (1) microglia characteristics, (2) blood-cerebrospinal fluid barrier integrity, (3) peripheral immune cell infiltration and (4) Aβ deposition in the brain. Next, we identified mechanisms that might cause this effect on AD pathology, more precisely Aβ efflux, persistent microglial activation and insufficient Aβ clearance, neuronal dysfunction and promotion of Aβ aggregation. Our results further strengthen the believe that even low-grade peripheral inflammation has detrimental effects on AD progression and may further reinforce the idea to modulate peripheral inflammation as a therapeutic strategy for AD.![]()
Collapse
|
7
|
Kimura A, Yoshikura N, Hayashi Y, Inuzuka T. Cerebrospinal Fluid C-C Motif Chemokine Ligand 2 Correlates with Brain Atrophy and Cognitive Impairment in Alzheimer's Disease. J Alzheimers Dis 2019; 61:581-588. [PMID: 29171996 DOI: 10.3233/jad-170519] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND Chronic neuroinflammation has been implicated in Alzheimer's disease (AD) pathology. OBJECTIVE To investigate the association between cytokine and anti-amyloid-β (Aβ) autoantibody levels and the degree of brain atrophy and cognitive impairment in AD patients. METHODS Cerebrospinal fluid (CSF) levels of C-C motif chemokine ligand 2 (CCL2), C-X-C motif chemokine ligand 8, C-X-C motif chemokine ligand 10, interleukin 6, and anti-Aβ autoantibody were evaluated in 69 AD patients. Serum levels of CCL2 and anti-Aβ autoantibody were also examined. The degree of brain atrophy was assessed using the voxel-based specific regional analysis system for AD, which targets the volumes of interest (VOI) in medial temporal structures. Cognitive function was evaluated by neuropsychological testing, including the Mini-Mental State Examination (MMSE) and Frontal Assessment Battery (FAB). RESULTS CSF CCL2 levels correlated significantly with the severity (p = 0.023) and the extent (p = 0.022) of VOI atrophy, and with the extent of gray matter atrophy (p = 0.039) in AD patients. CSF anti-Aβ autoantibody levels were inversely correlated with the severity of VOI atrophy (p = 0.020), the extent of VOI atrophy (p = 0.015), and the ratio of VOI/GM atrophy (r = -0.358, p = 0.004). CSF CCL2 levels were also inversely correlated with MMSE (p = 0.0497) and FAB scores (p = 0.016). CONCLUSIONS CSF CCL2 levels are associated with the degree of medial temporal lobe and gray matter atrophy, and cognitive decline in AD.
Collapse
Affiliation(s)
- Akio Kimura
- Departments of Neurology and Geriatrics, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Nobuaki Yoshikura
- Departments of Neurology and Geriatrics, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Yuichi Hayashi
- Departments of Neurology and Geriatrics, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Takashi Inuzuka
- Departments of Neurology and Geriatrics, Gifu University Graduate School of Medicine, Gifu, Japan
| |
Collapse
|
8
|
Xiao R, Yuan L, He W, Yang X. Zinc ions regulate opening of tight junction favouring efflux of macromolecules via the GSK3β/snail-mediated pathway. Metallomics 2019; 10:169-179. [PMID: 29292464 DOI: 10.1039/c7mt00288b] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Zinc is an essential trace element presenting in particularly high concentration in the brain. In some regions, e.g. lateral amygdala, subiculum and hippocampus, rapidly-exchangeable zinc may transiently reach even up to 600 μM. To explore the possible roles of high-concentration Zn2+ in regulating the blood-brain barrier (BBB), we investigated the effects of Zn2+ on the functions and structures of the tight junction (TJ) with an in vitro model of a Madin-Darby canine kidney (MDCK) cell monolayer. The experimental results indicated that high concentrations (>200 μM) of Zn2+ can affect the TJ integrity in a polarized manner. Basolateral addition of Zn2+ led to reversible TJ opening with pore paths of r ∼ 2 nm or more depending on Zn2+ concentration. The efflux/influx ratios of different sized probes were found to be ∼4.6 for FD4 (MW 4000) and ∼1.8 for Eu-DTPA (MW 560), suggesting that the Zn2+-induced paracelluar channels favour efflux especially for macromolecules. Further mechanistic studies revealed that the elevated intracellular Zn2+ taken from the basolateral side can increase phosphorylation of glycogen synthase kinase (GSK) 3β, primarily due to the inhibition of calcineurin (CaN), thus resulting in the elevation of the snail transcriptional repressors. Subsequently, Zn2+ can cause the down-regulation of claudin-1, breakage of occludin and ZO-1 rings, and collapse of basolateral F-actin structures. These overall factors result in the formation of a trumpet-like paracellular channel, which allows asymmetric solute permeation. The ERK1/2 and JNK1/2 pathways may also be involved in the Zn2+-induced TJ opening process, while the activation of matrix metalloproteinase was not observed. Our results may suggest a potential role of zinc in regulation of BBB permeability associated with brain clearance of metabolites through the glymphatic system.
Collapse
Affiliation(s)
- Ruyue Xiao
- State Key laboratories of Natural and Mimetic Drugs and Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing 100191, China.
| | | | | | | |
Collapse
|
9
|
Gu H, Kirchhein Y, Zhu T, Zhao G, Peng H, Du E, Liu J, Mastrianni JA, Farlow MR, Dodel R, Du Y. IVIG Delays Onset in a Mouse Model of Gerstmann-Sträussler-Scheinker Disease. Mol Neurobiol 2018; 56:2353-2361. [PMID: 30027340 DOI: 10.1007/s12035-018-1228-0] [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: 03/09/2018] [Accepted: 07/08/2018] [Indexed: 10/28/2022]
Abstract
Our previous studies showed that intravenous immunoglobulin (IVIG) contained anti-Aβ autoantibodies that might be able to treat Alzheimer's disease (AD). Recently, we identified and characterized naturally occurring autoantibodies against PrP from IVIG. Although autoantibodies in IVIG blocked PrP fibril formation and PrP neurotoxicity in vitro, it remained unknown whether IVIG could reduce amyloid plaque pathology in vivo and be used to effectively treat animals with prion diseases. In this study, we used Gerstmann-Sträussler-Scheinker (GSS)-Tg (PrP-A116V) transgenic mice to test IVIG efficacy since amyloid plaque formation played an important role in GSS pathogenesis. Here, we provided strong evidence that demonstrates how IVIG could significantly delay disease onset, elongate survival, and improve clinical phenotype in Tg (PrP-A116V) mice. Additionally, in treated animals, IVIG could markedly inhibit PrP amyloid plaque formation and attenuate neuronal apoptosis at the age of 120 days in mice. Our results indicate that IVIG may be a potential, effective therapeutic treatment for GSS and other prion diseases.
Collapse
Affiliation(s)
- Huiying Gu
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Yvonne Kirchhein
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.,Department of Neurology, Philipps-University Marburg, Marburg, Germany
| | - Timothy Zhu
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Gang Zhao
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.,School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan, People's Republic of China
| | - Hongjun Peng
- Department of Pediatrics, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210002, China
| | - Eileen Du
- Department of Psychology, Boston College, Chestnut Hill, MA, 02467, USA
| | - Junyi Liu
- Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | | | - Martin R Farlow
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Richard Dodel
- Department of Neurology, Philipps-University Marburg, Marburg, Germany
| | - Yansheng Du
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA. .,Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China.
| |
Collapse
|
10
|
Shinohara M, Tachibana M, Kanekiyo T, Bu G. Role of LRP1 in the pathogenesis of Alzheimer's disease: evidence from clinical and preclinical studies. J Lipid Res 2017; 58:1267-1281. [PMID: 28381441 DOI: 10.1194/jlr.r075796] [Citation(s) in RCA: 197] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 04/02/2017] [Indexed: 12/16/2022] Open
Abstract
Among the LDL receptor (LDLR) family members, the roles of LDLR-related protein (LRP)1 in the pathogenesis of Alzheimer's disease (AD), especially late-onset AD, have been the most studied by genetic, neuropathological, and biomarker analyses (clinical studies) or cellular and animal model systems (preclinical studies) over the last 25 years. Although there are some conflicting reports, accumulating evidence from preclinical studies indicates that LRP1 not only regulates the metabolism of amyloid-β peptides (Aβs) in the brain and periphery, but also maintains brain homeostasis, impairment of which likely contributes to AD development in Aβ-independent manners. Several preclinical studies have also demonstrated an involvement of LRP1 in regulating the pathogenic role of apoE, whose gene is the strongest genetic risk factor for AD. Nonetheless, evidence from clinical studies is not sufficient to conclude how LRP1 contributes to AD development. Thus, despite very promising results from preclinical studies, the role of LRP1 in AD pathogenesis remains to be further clarified. In this review, we discuss the potential mechanisms underlying how LRP1 affects AD pathogenesis through Aβ-dependent and -independent pathways by reviewing both clinical and preclinical studies. We also discuss potential therapeutic strategies for AD by targeting LRP1.
Collapse
Affiliation(s)
| | | | | | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL
| |
Collapse
|
11
|
Wang G, Manaenko A, Shao A, Ou Y, Yang P, Budbazar E, Nowrangi D, Zhang JH, Tang J. Low-density lipoprotein receptor-related protein-1 facilitates heme scavenging after intracerebral hemorrhage in mice. J Cereb Blood Flow Metab 2017; 37:1299-1310. [PMID: 27317656 PMCID: PMC5453452 DOI: 10.1177/0271678x16654494] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 04/14/2016] [Accepted: 05/10/2016] [Indexed: 11/16/2022]
Abstract
Heme-degradation after erythrocyte lysis plays an important role in the pathophysiology of intracerebral hemorrhage. Low-density lipoprotein receptor-related protein-1 is a receptor expressed predominately at the neurovascular interface, which facilitates the clearance of the hemopexin and heme complex. In the present study, we investigated the role of low-density lipoprotein receptor-related protein-1 in heme removal and neuroprotection in a mouse model of intracerebral hemorrhage. Endogenous low-density lipoprotein receptor-related protein-1 and hemopexin were increased in ipsilateral brain after intracerebral hemorrhage, accompanied by increased hemoglobin levels, brain water content, blood-brain barrier permeability and neurological deficits. Exogenous human recombinant low-density lipoprotein receptor-related protein-1 protein reduced hematoma volume, brain water content surrounding hematoma, blood-brain barrier permeability and improved neurological function three days after intracerebral hemorrhage. The expression of malondialdehyde, fluoro-Jade C positive cells and cleaved caspase 3 was increased three days after intracerebral hemorrhage in the ipsilateral brain tissues and decreased with recombinant low-density lipoprotein receptor-related protein-1. Intracerebral hemorrhage decreased and recombinant low-density lipoprotein receptor-related protein-1 increased the levels of superoxide dismutase 1. Low-density lipoprotein receptor-related protein-1 siRNA reduced the effect of human recombinant low-density lipoprotein receptor-related protein-1 on all outcomes measured. Collectively, our findings suggest that low-density lipoprotein receptor-related protein-1 contributed to heme clearance and blood-brain barrier protection after intracerebral hemorrhage. The use of low-density lipoprotein receptor-related protein-1 as supplement provides a novel approach to ameliorating intracerebral hemorrhage brain injury via its pleiotropic neuroprotective effects.
Collapse
Affiliation(s)
- Gaiqing Wang
- 1 Department of Physiology, Loma Linda University, Loma Linda, CA, USA
- 2 Department of Neurology, The Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Anatol Manaenko
- 1 Department of Physiology, Loma Linda University, Loma Linda, CA, USA
| | - Anwen Shao
- 1 Department of Physiology, Loma Linda University, Loma Linda, CA, USA
| | - Yibo Ou
- 1 Department of Physiology, Loma Linda University, Loma Linda, CA, USA
| | - Peng Yang
- 1 Department of Physiology, Loma Linda University, Loma Linda, CA, USA
| | | | - Derek Nowrangi
- 1 Department of Physiology, Loma Linda University, Loma Linda, CA, USA
| | - John H Zhang
- 1 Department of Physiology, Loma Linda University, Loma Linda, CA, USA
- 3 Department of Anesthesiology, Loma Linda University, Loma Linda, CA, USA
| | - Jiping Tang
- 1 Department of Physiology, Loma Linda University, Loma Linda, CA, USA
| |
Collapse
|
12
|
Zhang C, Lu J, Liu B, Cui Q, Wang Y. Primate-specific miR-603 is implicated in the risk and pathogenesis of Alzheimer's disease. Aging (Albany NY) 2016; 8:272-290. [PMID: 26856603 PMCID: PMC4789582 DOI: 10.18632/aging.100887] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 01/20/2016] [Indexed: 06/05/2023]
Abstract
Alzheimer's disease (AD) is a serious neurodegenerative disease, and microRNAs (miRNAs) have been linked to its pathogenesis. miR-603, a novel primate-specific miRNA and an intronic miRNA of a human brain highly expressed gene KIAA1217, is implicated in the risk and pathogenesis of AD. The rs11014002 single nucleotide polymorphism (SNP) (C/U), which locates in miR-603 precursor (pre-miR-603), exhibits a protective effect towards AD risk. Additionally, the rs11014002 SNP promotes the biogenesis of mature miR-603. miR-603 downregulates LRPAP1 mRNA and protein levels through directly binding the 3' untranslated region (3'UTR) of LRPAP1. Moreover, miR-603 increases LRP1 protein expression. LRPAP1 and LRP1, playing opposite roles, are involved in Aβ clearance and pathogenesis of AD. Strikingly, miR-603 exhibits a relatively higher expression and there is a loss of a negative correlation between miR-603 and LRPAP1/RND1 mRNA levels in the hippocampi of patients with AD. In addition, miR-603 directly downregulates a key neuronal apoptotic component-E2F1, and prevents HeLa cells from undergoing H2O2-induced apoptosis. This work suggests that miR-603 may be a novel AD-relevant miRNA and that its rs11014002 SNP may serve as a protective factor against AD.
Collapse
Affiliation(s)
- Chi Zhang
- Neuroscience Research Institute and Department of Neurobiology, Key Laboratory for Neuroscience of Ministry of Education, National Health and Family Planning Commission, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Jie Lu
- Neuroscience Research Institute and Department of Neurobiology, Key Laboratory for Neuroscience of Ministry of Education, National Health and Family Planning Commission, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Bing Liu
- Brainnetome Center, National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Qinghua Cui
- Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Yun Wang
- Neuroscience Research Institute and Department of Neurobiology, Key Laboratory for Neuroscience of Ministry of Education, National Health and Family Planning Commission, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
- PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| |
Collapse
|
13
|
Mushirobira Y, Mizuta H, Luo W, Todo T, Hara A, Reading BJ, Sullivan CV, Hiramatsu N. Molecular cloning and partial characterization of a low‐density lipoprotein receptor‐related protein 13 (Lrp13) involved in vitellogenin uptake in the cutthroat trout (
Oncorhynchus clarki
). Mol Reprod Dev 2015; 82:986-1000. [DOI: 10.1002/mrd.22579] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 08/30/2015] [Indexed: 01/01/2023]
Affiliation(s)
- Yuji Mushirobira
- Graduate School of Fisheries SciencesHokkaido UniversityHakodateJapan
| | - Hiroko Mizuta
- Graduate School of Fisheries SciencesHokkaido UniversityHakodateJapan
| | - Wenshu Luo
- Graduate School of Fisheries SciencesHokkaido UniversityHakodateJapan
| | - Takashi Todo
- Faculty of Fisheries SciencesHokkaido UniversityHakodateJapan
| | - Akihiko Hara
- Faculty of Fisheries SciencesHokkaido UniversityHakodateJapan
| | - Benjamin J. Reading
- Department of Applied EcologyNorth Carolina State UniversityRaleighNorth Carolina
| | | | | |
Collapse
|
14
|
A human monoclonal IgG that binds aβ assemblies and diverse amyloids exhibits anti-amyloid activities in vitro and in vivo. J Neurosci 2015; 35:6265-76. [PMID: 25904780 DOI: 10.1523/jneurosci.5109-14.2015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Alzheimer's disease (AD) and familial Danish dementia (FDD) are degenerative neurological diseases characterized by amyloid pathology. Normal human sera contain IgG antibodies that specifically bind diverse preamyloid and amyloid proteins and have shown therapeutic potential in vitro and in vivo. We cloned one of these antibodies, 3H3, from memory B cells of a healthy individual using a hybridoma method. 3H3 is an affinity-matured IgG that binds a pan-amyloid epitope, recognizing both Aβ and λ Ig light chain (LC) amyloids, which are associated with AD and primary amyloidosis, respectively. The pan-amyloid-binding properties of 3H3 were demonstrated using ELISA, immunohistochemical studies, and competition binding assays. Functional studies showed that 3H3 inhibits both Aβ and LC amyloid formation in vitro and abrogates disruption of hippocampal synaptic plasticity by AD-patient-derived soluble Aβ in vivo. A 3H3 single-chain variable fragment (scFv) retained the binding specificity of the 3H3 IgG and, when expressed in the brains of transgenic mice using an adeno-associated virus (AAV) vector, decreased parenchymal Aβ amyloid deposition in TgCRND8 mice and ADan (Danish Amyloid) cerebral amyloid angiopathy in the mouse model of FDD. These data indicate that naturally occurring human IgGs can recognize a conformational, amyloid-specific epitope and have potent anti-amyloid activities, providing a rationale to test their potential as antibody therapeutics for diverse neurological and other amyloid diseases.
Collapse
|
15
|
González-Marrero I, Giménez-Llort L, Johanson CE, Carmona-Calero EM, Castañeyra-Ruiz L, Brito-Armas JM, Castañeyra-Perdomo A, Castro-Fuentes R. Choroid plexus dysfunction impairs beta-amyloid clearance in a triple transgenic mouse model of Alzheimer's disease. Front Cell Neurosci 2015; 9:17. [PMID: 25705176 PMCID: PMC4319477 DOI: 10.3389/fncel.2015.00017] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 01/12/2015] [Indexed: 01/10/2023] Open
Abstract
Compromised secretory function of choroid plexus (CP) and defective cerebrospinal fluid (CSF) production, along with accumulation of beta-amyloid (Aβ) peptides at the blood-CSF barrier (BCSFB), contribute to complications of Alzheimer’s disease (AD). The AD triple transgenic mouse model (3xTg-AD) at 16 month-old mimics critical hallmarks of the human disease: β-amyloid (Aβ) plaques and neurofibrillary tangles (NFT) with a temporal- and regional- specific profile. Currently, little is known about transport and metabolic responses by CP to the disrupted homeostasis of CNS Aβ in AD. This study analyzed the effects of highly-expressed AD-linked human transgenes (APP, PS1 and tau) on lateral ventricle CP function. Confocal imaging and immunohistochemistry revealed an increase only of Aβ42 isoform in epithelial cytosol and in stroma surrounding choroidal capillaries; this buildup may reflect insufficient clearance transport from CSF to blood. Still, there was increased expression, presumably compensatory, of the choroidal Aβ transporters: the low density lipoprotein receptor-related protein 1 (LRP1) and the receptor for advanced glycation end product (RAGE). A thickening of the epithelial basal membrane and greater collagen-IV deposition occurred around capillaries in CP, probably curtailing solute exchanges. Moreover, there was attenuated expression of epithelial aquaporin-1 and transthyretin (TTR) protein compared to Non-Tg mice. Collectively these findings indicate CP dysfunction hypothetically linked to increasing Aβ burden resulting in less efficient ion transport, concurrently with reduced production of CSF (less sink action on brain Aβ) and diminished secretion of TTR (less neuroprotection against cortical Aβ toxicity). The putative effects of a disabled CP-CSF system on CNS functions are discussed in the context of AD.
Collapse
Affiliation(s)
| | - Lydia Giménez-Llort
- Institute of Neurosciences and Department of Psychiatry and Forensic Medicine, Autonomous University of Barcelona Barcelona, Spain
| | - Conrad E Johanson
- Department of Neurosurgery, Alpert Medical School at Brown University Providence, Rhode Island, USA
| | | | | | | | | | - Rafael Castro-Fuentes
- Department of Physiology, School of Medicine, University of La Laguna Tenerife, Spain
| |
Collapse
|