1
|
de Jager C, Soliman E, Theus MH. Interrogating mediators of single-cell transcriptional changes in the acute damaged cerebral cortex: Insights into endothelial-astrocyte interactions. Mol Cell Neurosci 2025; 133:104003. [PMID: 40090391 PMCID: PMC12146052 DOI: 10.1016/j.mcn.2025.104003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 03/09/2025] [Accepted: 03/11/2025] [Indexed: 03/18/2025] Open
Abstract
Traumatic brain injury (TBI) induces complex cellular and molecular changes, challenging recovery and therapeutic development. Although molecular pathways have been implicated in TBI pathology, the cellular specificity of these mechanisms remains underexplored. Here, we investigate the role of endothelial cell (EC) EphA4, a receptor tyrosine kinase receptor involved in axonal guidance, in modulating cell-specific transcriptomic changes within the damaged cerebral cortex. Utilizing single-cell RNA sequencing (scRNA-seq) in an experimental TBI model, we mapped transcriptional changes across various cell types, with a focus on astrocytes and ECs. Our analysis reveals that EC-specific knockout (KO) of EphA4 triggers significant alterations in astrocyte gene expression and shifts predominate subclusters. We identified six distinct astrocyte clusters (C0-C5) in the damaged cortex including as C0-Mobp/Plp1+; C1-Slc1a3/Clu+; C2-Hbb-bs/Hba-a1/Ndrg2+; C3-GFAP/Lcn2+; C4-Gli3/Mertk+, and C5-Cox8a+. We validate a new Sox9+ cluster expressing Mertk and Gas, which mediates efferocytosis to facilitate apoptotic cell clearance and anti-inflammatory responses. Transcriptomic and CellChat analyses of EC-KO cells highlights upregulation of neuroprotective pathways, including increased amyloid precursor protein (APP) and Gas6. Key pathways predicted to be modulated in astrocytes from EC-KO mice include oxidative phosphorylation and FOXO signaling, mitochondrial dysfunction and ephrin B signaling. Concurrently, metabolic and signaling pathways in endothelial cells-such as ceramide and sphingosine phosphate metabolism and NGF-stimulated transcription-indicate an adaptive response to a metabolically demanding post-injury hypoxic environment. These findings elucidate potential interplay between astrocytic and endothelial responses as well as transcriptional networks underlying cortical tissue damage.
Collapse
Affiliation(s)
- Caroline de Jager
- Translational Biology Medicine and Health Graduate Program, Blacksburg, VA 24061, USA
| | - Eman Soliman
- Department of Biomedical Sciences and Pathobiology, Blacksburg, VA 24061, USA
| | - Michelle H Theus
- Department of Biomedical Sciences and Pathobiology, Blacksburg, VA 24061, USA; Center for Engineered Health, Virginia Tech, Blacksburg, VA 24061, USA.
| |
Collapse
|
2
|
Rodkin S, Nwosu C, Kirichenko E. The Role of Hydrogen Sulfide in iNOS and APP Localization and Expression in Neurons and Glial Cells Under Traumatic Effects: An Experimental Study with Bioinformatics Analysis and Biomodeling. Int J Mol Sci 2024; 25:11892. [PMID: 39595962 PMCID: PMC11593695 DOI: 10.3390/ijms252211892] [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: 10/15/2024] [Revised: 11/01/2024] [Accepted: 11/03/2024] [Indexed: 11/28/2024] Open
Abstract
Hydrogen sulfide (H2S) donors are emerging as promising candidates for neuroprotective agents. However, H2S-dependent neuroprotective mechanisms are not yet fully understood. We have demonstrated that an H2S donor (sodium sulfide, Na2S) reduces the expression of inducible NO synthase (iNOS) and amyloid-beta precursor protein (APP) in damaged neural tissue at 24 h and 7 days following traumatic brain injury (TBI). The application of aminooxyacetic acid (AOAA), an inhibitor of cystathionine β-synthase (CBS), produced the opposite effect. Seven days after TBI, iNOS expression was observed not only in the cytoplasm but also in some neuronal nuclei, while APP was exclusively localized in the cytoplasm and axons of damaged neurons. It was also shown that iNOS and APP were present in the cytoplasm of mechanoreceptor neurons (MRNs) in the crayfish, in axons, as well as in certain glial cells 8 h after axotomy. Na2S and AOAA had opposing effects on axotomized MRNs and ganglia in the ventral nerve cord (VNC). Multiple sequence alignments revealed a high degree of identity among iNOS and APP amino acid residues in various vertebrate and invertebrate species. In the final stage of this study, biomodeling identified unique binding sites for H2S, hydrosulfide anion (HS-), and thiosulfate (S2O32-) with iNOS and APP.
Collapse
Affiliation(s)
- Stanislav Rodkin
- Research Laboratory "Medical Digital Images Based on the Basic Model", Department of Bioengineering, Faculty of Bioengineering and Veterinary Medicine, Don State Technical University, Rostov-on-Don 344000, Russia
| | | | | |
Collapse
|
3
|
König S, Schmidt N, Bechberger K, Morris S, Priego M, Zaky H, Song Y, Pielage J, Brunholz S, Brady ST, Kins S, Morfini G. Axon-Autonomous Effects of the Amyloid Precursor Protein Intracellular Domain (AICD) on Kinase Signaling and Fast Axonal Transport. Cells 2023; 12:2403. [PMID: 37830617 PMCID: PMC10572015 DOI: 10.3390/cells12192403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/26/2023] [Accepted: 09/27/2023] [Indexed: 10/14/2023] Open
Abstract
The amyloid precursor protein (APP) is a key molecular component of Alzheimer's disease (AD) pathogenesis. Proteolytic APP processing generates various cleavage products, including extracellular amyloid beta (Aβ) and the cytoplasmic APP intracellular domain (AICD). Although the role of AICD in the activation of kinase signaling pathways is well established in the context of full-length APP, little is known about intracellular effects of the AICD fragment, particularly within discrete neuronal compartments. Deficits in fast axonal transport (FAT) and axonopathy documented in AD-affected neurons prompted us to evaluate potential axon-autonomous effects of the AICD fragment for the first time. Vesicle motility assays using the isolated squid axoplasm preparation revealed inhibition of FAT by AICD. Biochemical experiments linked this effect to aberrant activation of selected axonal kinases and heightened phosphorylation of the anterograde motor protein conventional kinesin, consistent with precedents showing phosphorylation-dependent regulation of motors proteins powering FAT. Pharmacological inhibitors of these kinases alleviated the AICD inhibitory effect on FAT. Deletion experiments indicated this effect requires a sequence encompassing the NPTY motif in AICD and interacting axonal proteins containing a phosphotyrosine-binding domain. Collectively, these results provide a proof of principle for axon-specific effects of AICD, further suggesting a potential mechanistic framework linking alterations in APP processing, FAT deficits, and axonal pathology in AD.
Collapse
Affiliation(s)
- Svenja König
- Department for Human Biology and Human Genetics, University of Kaiserslautern-Landau, 67663 Kaiserslautern, Germany (K.B.); (S.K.)
| | - Nadine Schmidt
- Department for Human Biology and Human Genetics, University of Kaiserslautern-Landau, 67663 Kaiserslautern, Germany (K.B.); (S.K.)
| | - Karin Bechberger
- Department for Human Biology and Human Genetics, University of Kaiserslautern-Landau, 67663 Kaiserslautern, Germany (K.B.); (S.K.)
| | - Sarah Morris
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612, USA (S.T.B.)
- Marine Biological Laboratory, Woods Hole, MA 02543, USA
| | - Mercedes Priego
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612, USA (S.T.B.)
| | - Hannah Zaky
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612, USA (S.T.B.)
| | - Yuyu Song
- Marine Biological Laboratory, Woods Hole, MA 02543, USA
- Department of Neurology, Harvard Medical School and Massachusetts General Hospital, Boston, MA 02129, USA
| | - Jan Pielage
- Department of Zoology, University of Kaiserslautern-Landau, 67663 Kaiserslautern, Germany;
| | - Silke Brunholz
- Department for Human Biology and Human Genetics, University of Kaiserslautern-Landau, 67663 Kaiserslautern, Germany (K.B.); (S.K.)
| | - Scott T. Brady
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612, USA (S.T.B.)
- Marine Biological Laboratory, Woods Hole, MA 02543, USA
| | - Stefan Kins
- Department for Human Biology and Human Genetics, University of Kaiserslautern-Landau, 67663 Kaiserslautern, Germany (K.B.); (S.K.)
| | - Gerardo Morfini
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612, USA (S.T.B.)
- Marine Biological Laboratory, Woods Hole, MA 02543, USA
| |
Collapse
|
4
|
Jang J, Yeo S, Baek S, Jung HJ, Lee MS, Choi SH, Choe Y. Abnormal accumulation of extracellular vesicles in hippocampal dystrophic axons and regulation by the primary cilia in Alzheimer's disease. Acta Neuropathol Commun 2023; 11:142. [PMID: 37667395 PMCID: PMC10478284 DOI: 10.1186/s40478-023-01637-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 08/15/2023] [Indexed: 09/06/2023] Open
Abstract
Dystrophic neurites (DNs) are abnormal axons and dendrites that are swollen or deformed in various neuropathological conditions. In Alzheimer's disease (AD), DNs play a crucial role in impairing neuronal communication and function, and they may also contribute to the accumulation and spread of amyloid beta (Aβ) in the brain of AD patients. However, it is still a challenge to understand the DNs of specific neurons that are vulnerable to Aβ in the pathogenesis of AD. To shed light on the development of radiating DNs, we examined enriched dystrophic hippocampal axons in a mouse model of AD using a three-dimensional rendering of projecting neurons. We employed the anterograde spread of adeno-associated virus (AAV)1 and conducted proteomic analysis of synaptic compartments obtained from hippocampo-septal regions. Our findings revealed that DNs were formed due to synaptic loss at the axon terminals caused by the accumulation of extracellular vesicle (EV). Abnormal EV-mediated transport and exocytosis were identified in association with primary cilia, indicating their involvement in the accumulation of EVs at presynaptic terminals. To further address the regulation of DNs by primary cilia, we conducted knockdown of the Ift88 gene in hippocampal neurons, which impaired EV-mediated secretion of Aβ and promoted accumulation of axonal spheroids. Using single-cell RNA sequencing, we identified the septal projecting hippocampal somatostatin neurons (SOM) as selectively vulnerable to Aβ with primary cilia dysfunction and vesicle accumulation. Our study suggests that DNs in AD are initiated by the ectopic accumulation of EVs at the neuronal axon terminals, which is affected by neuronal primary cilia.
Collapse
Affiliation(s)
| | - Seungeun Yeo
- Korea Brain Research Institute, Daegu, 41068, Korea
| | | | | | - Mi Suk Lee
- Korea Brain Research Institute, Daegu, 41068, Korea
| | | | - Youngshik Choe
- Korea Brain Research Institute, Daegu, 41068, Korea.
- , Daegu, Korea.
| |
Collapse
|
5
|
Cheng GWY, Ma IWT, Huang J, Yeung SHS, Ho P, Chen Z, Mak HKF, Herrup K, Chan KWY, Tse KH. Cuprizone drives divergent neuropathological changes in different mouse models of Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.24.547147. [PMID: 37546935 PMCID: PMC10402084 DOI: 10.1101/2023.07.24.547147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Myelin degradation is a normal feature of brain aging that accelerates in Alzheimer's disease (AD). To date, however, the underlying biological basis of this correlation remains elusive. The amyloid cascade hypothesis predicts that demyelination is caused by increased levels of the β-amyloid (Aβ) peptide. Here we report on work supporting the alternative hypothesis that early demyelination is upstream of amyloid. We challenged two different mouse models of AD (R1.40 and APP/PS1) using cuprizone-induced demyelination and tracked the responses with both neuroimaging and neuropathology. In oppose to amyloid cascade hypothesis, R1.40 mice, carrying only a single human mutant APP (Swedish; APP SWE ) transgene, showed a more abnormal changes of magnetization transfer ratio and diffusivity than in APP/PS1 mice, which carry both APP SWE and a second PSEN1 transgene (delta exon 9; PSEN1 dE9 ). Although cuprizone targets oligodendrocytes (OL), magnetic resonance spectroscopy and targeted RNA-seq data in R1.40 mice suggested a possible metabolic alternation in axons. In support of alternative hypotheses, cuprizone induced significant intraneuronal amyloid deposition in young APP/PS1, but not in R1.40 mice, and it suggested the presence of PSEN deficiencies, may accelerate Aβ deposition upon demyelination. In APP/PS1, mature OL is highly vulnerable to cuprizone with significant DNA double strand breaks (53BP1 + ) formation. Despite these major changes in myelin, OLs, and Aβ immunoreactivity, no cognitive impairment or hippocampal pathology was detected in APP/PS1 mice after cuprizone treatment. Together, our data supports the hypothesis that myelin loss can be the cause, but not the consequence, of AD pathology. SIGNIFICANCE STATEMENT The causal relationship between early myelin loss and the progression of Alzheimer's disease remains unclear. Using two different AD mouse models, R1.40 and APP/PS1, our study supports the hypothesis that myelin abnormalities are upstream of amyloid production and deposition. We find that acute demyelination initiates intraneuronal amyloid deposition in the frontal cortex. Further, the loss of oligodendrocytes, coupled with the accelerated intraneuronal amyloid deposition, interferes with myelin tract diffusivity at a stage before any hippocampus pathology or cognitive impairments occur. We propose that myelin loss could be the cause, not the consequence, of amyloid pathology during the early stages of Alzheimer's disease.
Collapse
|
6
|
Wu MY, Zou WJ, Lee D, Mei L, Xiong WC. APP in the Neuromuscular Junction for the Development of Sarcopenia and Alzheimer's Disease. Int J Mol Sci 2023; 24:ijms24097809. [PMID: 37175515 PMCID: PMC10178513 DOI: 10.3390/ijms24097809] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 04/18/2023] [Accepted: 04/22/2023] [Indexed: 05/15/2023] Open
Abstract
Sarcopenia, an illness condition usually characterized by a loss of skeletal muscle mass and muscle strength or function, is often associated with neurodegenerative diseases, such as Alzheimer's disease (AD), a common type of dementia, leading to memory loss and other cognitive impairment. However, the underlying mechanisms for their associations and relationships are less well understood. The App, a Mendelian gene for early-onset AD, encodes amyloid precursor protein (APP), a transmembrane protein enriched at both the neuromuscular junction (NMJ) and synapses in the central nervous system (CNS). Here, in this review, we highlight APP and its family members' physiological functions and Swedish mutant APP (APPswe)'s pathological roles in muscles and NMJ. Understanding APP's pathophysiological functions in muscles and NMJ is likely to uncover insights not only into neuromuscular diseases but also AD. We summarize key findings from the burgeoning literature, which may open new avenues to investigate the link between muscle cells and brain cells in the development and progression of AD and sarcopenia.
Collapse
Affiliation(s)
- Min-Yi Wu
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Wen-Jun Zou
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Daehoon Lee
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
- Louis Stokes Cleveland Veterans Affairs Medical Center, Northeast Ohio VA Healthcare System, Cleveland, OH 44106, USA
| | - Lin Mei
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Wen-Cheng Xiong
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
- Louis Stokes Cleveland Veterans Affairs Medical Center, Northeast Ohio VA Healthcare System, Cleveland, OH 44106, USA
| |
Collapse
|
7
|
Alhodieb FS, Rahman MA, Barkat MA, Alanezi AA, Barkat HA, Hadi HA, Harwansh RK, Mittal V. Nanomedicine-driven therapeutic interventions of autophagy and stem cells in the management of Alzheimer's disease. Nanomedicine (Lond) 2023; 18:145-168. [PMID: 36938800 DOI: 10.2217/nnm-2022-0108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2023] Open
Abstract
Drug-loaded, brain-targeted nanocarriers could be a promising tool in overcoming the challenges associated with Alzheimer's disease therapy. These nanocargoes are enormously flexible to functionalize and facilitate the delivery of drugs to brain cells by bridging the blood-brain barrier and into brain cells. To date, modifications have included nanoparticles (NPs) coating with tunable surfactants/phospholipids, covalently attaching polyethylene glycol chains (PEGylation), and tethering different targeting ligands to cell-penetrating peptides in a manner that facilitates their entry across the BBB and downregulates various pathological hallmarks as well as intra- and extracellular signaling pathways. This review provides a brief update on drug-loaded, multifunctional nanocarriers and the therapeutic intervention of autophagy and stem cells in the management of Alzheimer's disease.
Collapse
Affiliation(s)
- Fahad Saad Alhodieb
- Department of Clinical Nutrition, College of Applied Health Sciences in Arras, Qassim University, Ar Rass, 51921, Saudi Arabia
| | | | - Muhammad Abul Barkat
- Department of Pharmaceutics, College of Pharmacy, University of Hafr Al-Batin, Al Jamiah, Hafr Al Batin, 39524, Saudi Arabia
| | - Abdulkareem A Alanezi
- Department of Pharmaceutics, College of Pharmacy, University of Hafr Al-Batin, Al Jamiah, Hafr Al Batin, 39524, Saudi Arabia
| | - Harshita Abul Barkat
- Department of Pharmaceutics, College of Pharmacy, University of Hafr Al-Batin, Al Jamiah, Hafr Al Batin, 39524, Saudi Arabia.,Dermatopharmaceutics Research Group, Faculty of Pharmacy, International Islamic University Malaysia, Kuantan, Pahang, 25200, Malaysia
| | - Hazrina Ab Hadi
- Dermatopharmaceutics Research Group, Faculty of Pharmacy, International Islamic University Malaysia, Kuantan, Pahang, 25200, Malaysia
| | - Ranjit K Harwansh
- Institute of Pharmaceutical Research, GLA University, Mathura, 281406, India
| | - Vineet Mittal
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana, 124001, India
| |
Collapse
|
8
|
Lin S, Leitão ADG, Fang S, Gu Y, Barber S, Gilliard-Telefoni R, Castro A, Sung K, Shen R, Florio JB, Mante ML, Ding J, Spencer B, Masliah E, Rissman RA, Wu C. Overexpression of alpha synuclein disrupts APP and Endolysosomal axonal trafficking in a mouse model of synucleinopathy. Neurobiol Dis 2023; 178:106010. [PMID: 36702318 PMCID: PMC10754494 DOI: 10.1016/j.nbd.2023.106010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 01/18/2023] [Accepted: 01/20/2023] [Indexed: 01/24/2023] Open
Abstract
Mutations or triplication of the alpha synuclein (ASYN) gene contribute to synucleinopathies including Parkinson's disease (PD), Dementia with Lewy bodies (DLB) and multiple system atrophy (MSA). Recent evidence suggests that ASYN also plays an important role in amyloid-induced neurotoxicity, although the mechanism(s) remains unknown. One hypothesis is that accumulation of ASYN alters endolysosomal pathways to impact axonal trafficking and processing of the amyloid precursor protein (APP). To define an axonal function for ASYN, we used a transgenic mouse model of synucleinopathy that expresses a GFP-human ASYN (GFP-hASYN) transgene and an ASYN knockout (ASYN-/-) mouse model. Our results demonstrate that expression of GFP-hASYN in primary neurons derived from a transgenic mouse impaired axonal trafficking and processing of APP. In addition, axonal transport of BACE1, Rab5, Rab7, lysosomes and mitochondria were also reduced in these neurons. Interestingly, axonal transport of these organelles was also affected in ASYN-/- neurons, suggesting that ASYN plays an important role in maintaining normal axonal transport function. Therefore, selective impairment of trafficking and processing of APP by ASYN may act as a potential mechanism to induce pathological features of Alzheimer's disease (AD) in PD patients.
Collapse
Affiliation(s)
- Suzhen Lin
- Institute of Neurology, Ruijing Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China; Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - André D G Leitão
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Savannah Fang
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Yingli Gu
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Sophia Barber
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | | | - Alfredo Castro
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Kijung Sung
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Ruinan Shen
- Institute of Neurology, Ruijing Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China; Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Jazmin B Florio
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Michael L Mante
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Jianqing Ding
- Institute of Neurology, Ruijing Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Brian Spencer
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Eliezer Masliah
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Robert A Rissman
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA; VA San Diego Health System, La Jolla, CA, USA.
| | - Chengbiao Wu
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
9
|
Yang Y, Yang J, Liang Y, Zhang G, Cai Z, Zhang Y, Lin H, Tan M. Rab3A interacts with spastin to regulate neurite outgrowth in hippocampal neurons. Biochem Biophys Res Commun 2023; 643:77-87. [PMID: 36587525 DOI: 10.1016/j.bbrc.2022.12.066] [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: 12/07/2022] [Revised: 12/19/2022] [Accepted: 12/22/2022] [Indexed: 12/24/2022]
Abstract
Investigating novel mechanisms of neurite outgrowth via cytoskeleton is critical for developing therapeutic strategies against neural disorders. Rab3A is a vesicle-related protein distributed throughout the nervous system, but the detailed mechanism related to cytoskeleton remains largely unknown. Our previous reports show that spastin serves microtubule to regulate neurite outgrowth. Here, we asked whether Rab3A could function via modulating spastin during neuronal development. The results revealed that Rab3A colocalized with spastin in cultured hippocampal neurons. Immunoprecipitation assays showed that Rab3A physically interacted with spastin in rat brain lysates. Rab3A overexpression significantly induced spastin degradation; this effect was reversed by leupeptin- or MG-132- administration, suggesting the lysosomal and ubiquitin-mediated degradation system. Immunofluorescence staining further confirmed that Rab3A and spastin immune-colocalized with the lysosome marker lysotracker. In COS7 cells, Rab3A overexpression significantly downregulated spastin expression and abolished the spastin-mediated microtubule severing. Furthermore, overexpression inhibited while genetic knockdown of Rab3A promoted neurite outgrowth. However, this inhibitory effect on neurite outgrowth in hippocampal neurons could be reversed via co-transfection of spastin, indicating that Rab3A functions via its interaction protein spastin. In general, our data identify an interaction between Rab3A and spastin, and this interaction affects the protein stability of spastin and eliminates its microtubule severing function, thereby modulating neurite outgrowth.
Collapse
Affiliation(s)
- Yuhao Yang
- Department of Orthopaedics, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, 510630, China
| | - Jie Yang
- Department of Orthopaedics, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, 510630, China
| | - Yaozhong Liang
- Department of Orthopaedics, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, 510630, China
| | - Guowei Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, 510630, China
| | - Zhenbin Cai
- Department of Orthopaedics, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, 510630, China
| | - Yunlong Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, 510630, China
| | - Hongsheng Lin
- Department of Orthopaedics, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, 510630, China.
| | - Minghui Tan
- Department of Orthopaedics, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, 510630, China.
| |
Collapse
|
10
|
Schoeman R, Voges T. Attention-deficit hyperactivity disorder stigma: The silent barrier to care. S Afr J Psychiatr 2022; 28:1865. [PMID: 36569805 PMCID: PMC9772730 DOI: 10.4102/sajpsychiatry.v28i0.1865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 06/09/2022] [Indexed: 12/23/2022] Open
Abstract
Background Attention-deficit hyperactivity disorder (ADHD) is the most common psychiatric disorder in childhood, with symptoms persisting into adulthood in 60% of individuals. If left untreated, the emotional, social and financial consequences can be dire, with many children and adults not reaching their full potential and having a reduced quality of life. Aim The study explored parents' and educators' understanding and experience of stigma in relation to their children's ADHD. Setting Participants were recruited from six schools in the Cape Town metropole, in which the Goldilocks and The Bear Foundation (which delivers mental health services to underprivileged children) were active. Methods A convergent parallel mixed methods research design (consisting of a quantitative survey and an in-depth interview component) was conducted to explore the lack of knowledge about ADHD and stigma as potential barriers to help-seeking behaviour, diagnosis and treatment for children with ADHD. Results Instrumental barriers to care had a bigger impact on practical access to care, while attitudinal and stigma-related resources were found to have a significant impact on well-being of individuals. Core to the themes arising from the interviews were questions of how lack of knowledge influences stigma, how stigma materialises in discriminatory behaviour and how stigma acts as a barrier to care. Conclusion The findings contribute to the literature by exploring parents' and educators' understanding and experience of stigma in relation to their children's ADHD. A collaborative stakeholder approach is needed for effective, comprehensive and relevant interventions to combat stigma and enhance early identification of and interventions for ADHD. Contribution In order to improve access to care, treatment, and well-being of individuals directly or indirectly affected by ADHD, it is crucial that stigma needs to be addressed.
Collapse
Affiliation(s)
- Renata Schoeman
- Stellenbosch Business School, Stellenbosch University, Bellville, South Africa,Private practice, Bellville, South Africa,Goldilocks and The Bear Foundation, Bellville, South Africa
| | - Tawni Voges
- Goldilocks and The Bear Foundation, Bellville, South Africa,Department of Psychology, Faculty of Social Sciences, Stellenbosch University, Cape Town, South Africa
| |
Collapse
|
11
|
Role of Chemokines in the Development and Progression of Alzheimer's Disease. J Mol Neurosci 2022; 72:1929-1951. [PMID: 35821178 PMCID: PMC9392685 DOI: 10.1007/s12031-022-02047-1] [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] [Received: 05/16/2022] [Accepted: 07/04/2022] [Indexed: 11/24/2022]
Abstract
Alzheimer’s disease (AD) is a progressive neurogenerative disorder manifested by gradual memory loss and cognitive decline due to profound damage of cholinergic neurons. The neuropathological hallmarks of AD are intracellular deposits of neurofibrillary tangles (NFTs) and extracellular aggregates of amyloid β (Aβ). Mounting evidence indicates that intensified neuroinflammatory processes play a pivotal role in the pathogenesis of AD. Chemokines serve as signaling molecules in immune cells but also in nerve cells. Under normal conditions, neuroinflammation plays a neuroprotective role against various harmful factors. However, overexpression of chemokines initiates disruption of the integrity of the blood–brain barrier, facilitating immune cells infiltration into the brain. Then activated adjacent glial cells–astrocytes and microglia, release massive amounts of chemokines. Prolonged inflammation loses its protective role and drives an increase in Aβ production and aggregation, impairment of its clearance, or enhancement of tau hyperphosphorylation, contributing to neuronal loss and exacerbation of AD. Moreover, chemokines can be further released in response to growing deposits of toxic forms of Aβ. On the other hand, chemokines seem to exert multidimensional effects on brain functioning, including regulation of neurogenesis and synaptic plasticity in regions responsible for memory and cognitive abilities. Therefore, underexpression or complete genetic ablation of some chemokines can worsen the course of AD. This review covers the current state of knowledge on the role of particular chemokines and their receptors in the development and progression of AD. Special emphasis is given to their impact on forming Aβ and NFTs in humans and in transgenic murine models of AD.
Collapse
|
12
|
Chopra H, Bibi S, Singh I, Kamal MA, Islam F, Alhumaydhi FA, Emran TB, Cavalu S. Nanomedicines in the Management of Alzheimer's Disease: Current View and Future Prospects. Front Aging Neurosci 2022; 14:879114. [PMID: 35875806 PMCID: PMC9304964 DOI: 10.3389/fnagi.2022.879114] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 06/17/2022] [Indexed: 12/27/2022] Open
Abstract
Alzheimer's disease (AD) is a kind of dementia that creates serious challenges for sufferers' memory, thinking, and behavior. It commonly targeting the aging population and decay the brain cells, despite attempts have been performed to enhance AD diagnostic and therapeutic techniques. Hence, AD remains incurable owing to its complex and multifactorial consequences and still there is lack of appropriate diagnostics/therapeutics option for this severe brain disorder. Therefore, nanotechnology is currently bringing new tools and insights to improve the previous knowledge of AD and ultimately may provide a novel treatment option and a ray of hope to AD patients. Here in this review, we highlighted the nanotechnologies-based findings for AD, in both diagnostic and therapeutic aspects and explained how advances in the field of nanotechnology/nanomedicine could enhance patient prognosis and quality of life. It is highly expected these emerging technologies could bring a research-based revolution in the field of neurodegenerative disorders and may assist their clinical experiments and develop an efficacious drug for AD also. The main aim of review is to showcase readers the recent advances in nanotechnology-based approaches for treatment and diagnosing of AD.
Collapse
Affiliation(s)
- Hitesh Chopra
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Shabana Bibi
- Department of Biosciences, Shifa Tameer-e-Millat University, Islamabad, Pakistan
- Yunnan Herbal Laboratory, College of Ecology and Environmental Sciences, Yunnan University, Kunming, China
| | - Inderbir Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Mohammad Amjad Kamal
- Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
- Enzymoics, Novel Global Community Educational Foundation, Hebersham, NSW, Australia
| | - Fahadul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Fahad A. Alhumaydhi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Talha Bin Emran
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, Bangladesh
| | - Simona Cavalu
- Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| |
Collapse
|
13
|
Amyloid Precursor Protein in Abusive Head Trauma Suspects. Am J Ophthalmol 2022; 240:58-66. [PMID: 35247337 DOI: 10.1016/j.ajo.2022.02.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 02/14/2022] [Accepted: 02/16/2022] [Indexed: 11/23/2022]
Abstract
PURPOSE To study orthograde axonal transport with amyloid precursor protein (APP-A4) immunohistochemistry (IHC) in the retina and lamina cribrosa (LC) portion of the optic nerve in abusive head trauma (AHT) suspects. DESIGN Retrospective, case-control study. METHODS Seventy-two eyes from suspected AHT victims referred by the Los Angeles Coroner and control eyes from nontraumatized infants were included. IHC was conducted using University of California, Irvine (UCI), Lab Medicine Department's standard protocol and results analyzed by light microcopy after paraffin processing. Quantitation of LC APP-A4 block was estimated in 21 cases with known survival using MetaMorph, a proprietary biomicroscopy imaging software. RESULTS The presence or absence of APP-A4 label accumulations in retinal ganglion cells, nerve fiber layer at the disc margin, and in LC axonal bundles were compared to matching tissues from nontraumatized control eyes with only background staining. Among the globes from AHT suspects with nerve heads available for study, 94% were positive for LC accumulation of marker. Among suspect AHT cases with known survival after injury of 1 to 1588 days, most demonstrated LC APP-A4 accumulations. CONCLUSIONS Our findings reinforce a recent publication based on APP-A4 IHC that demonstrated similar orthograde axonal transport block in the LC in children with AHT and recommend that intraocular pressures be recorded and addressed in these patients.
Collapse
|
14
|
Eggert S, Kins S, Endres K, Brigadski T. Brothers in arms: proBDNF/BDNF and sAPPα/Aβ-signaling and their common interplay with ADAM10, TrkB, p75NTR, sortilin, and sorLA in the progression of Alzheimer's disease. Biol Chem 2022; 403:43-71. [PMID: 34619027 DOI: 10.1515/hsz-2021-0330] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 09/16/2021] [Indexed: 12/22/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) is an important modulator for a variety of functions in the central nervous system (CNS). A wealth of evidence, such as reduced mRNA and protein level in the brain, cerebrospinal fluid (CSF), and blood samples of Alzheimer's disease (AD) patients implicates a crucial role of BDNF in the progression of this disease. Especially, processing and subcellular localization of BDNF and its receptors TrkB and p75 are critical determinants for survival and death in neuronal cells. Similarly, the amyloid precursor protein (APP), a key player in Alzheimer's disease, and its cleavage fragments sAPPα and Aβ are known for their respective roles in neuroprotection and neuronal death. Common features of APP- and BDNF-signaling indicate a causal relationship in their mode of action. However, the interconnections of APP- and BDNF-signaling are not well understood. Therefore, we here discuss dimerization properties, localization, processing by α- and γ-secretase, relevance of the common interaction partners TrkB, p75, sorLA, and sortilin as well as shared signaling pathways of BDNF and sAPPα.
Collapse
Affiliation(s)
- Simone Eggert
- Department of Human Biology and Human Genetics, University of Kaiserslautern, Erwin-Schrödinger-Str. 13, D-67663 Kaiserslautern, Germany
| | - Stefan Kins
- Department of Human Biology and Human Genetics, University of Kaiserslautern, Erwin-Schrödinger-Str. 13, D-67663 Kaiserslautern, Germany
| | - Kristina Endres
- Department of Psychiatry and Psychotherapy, University Medical Center, Johannes Gutenberg-University Mainz, D-55131 Mainz, Germany
| | - Tanja Brigadski
- Department of Informatics and Microsystem Technology, University of Applied Sciences Kaiserslautern, D-66482 Zweibrücken, Germany
| |
Collapse
|
15
|
Armbrust F, Bickenbach K, Marengo L, Pietrzik C, Becker-Pauly C. The Swedish dilemma - the almost exclusive use of APPswe-based mouse models impedes adequate evaluation of alternative β-secretases. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1869:119164. [PMID: 34699873 DOI: 10.1016/j.bbamcr.2021.119164] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 09/30/2021] [Accepted: 10/11/2021] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is the most common form of dementia, however incurable so far. It is widely accepted that aggregated amyloid β (Aβ) peptides play a crucial role for the pathogenesis of AD, as they cause neurotoxicity and deposit as so-called Aβ plaques in AD patient brains. Aβ peptides derive from the amyloid precursor protein (APP) upon consecutive cleavage at the β- and γ-secretase site. Hence, mutations in the APP gene are often associated with autosomal dominant inherited AD. Almost thirty years ago, two mutations at the β-secretase site were observed in two Swedish families (termed Swedish APP (APPswe) mutations), which led to early-onset AD. Consequently, APPswe was established in almost every common AD mouse model, as it contributes to early Aβ plaque formation and cognitive impairments. Analyzing these APPswe-based mouse models, the aspartyl protease BACE1 has been evolving as the prominent β-secretase responsible for Aβ release in AD and as the most important therapeutic target for AD treatment. However, with respect to β-secretase processing, the very rare occurring APPswe variant substantially differs from wild-type APP. BACE1 dominates APPswe processing resulting in the release of Aβ1-x, whereas N-terminally truncated Aβ forms are scarcely generated. However, these N-terminally truncated Aβ species such as Aβ2-x, Aβ3-x and Aβ4-x are elevated in AD patient brains and exhibit an increased potential to aggregate compared to Aβ1-x peptides. Proteases such as meprin β, cathepsin B and ADAMTS4 were identified as alternative β-secretases being capable of generating these N-terminally truncated Aβ species from wild-type APP. However, neither meprin β nor cathepsin B are capable of generating N-terminally truncated Aβ peptides from APPswe. Hence, the role of BACE1 for the Aβ formation during AD might be overrepresented through the excessive use of APPswe mouse models. In this review we critically discuss the consideration of BACE1 as the most promising therapeutic target. Shifting the focus of AD research towards alternative β secretases might unveil promising alternatives to BACE1 inhibitors constantly failing in clinical trials due to ineffectiveness and harmful side effects.
Collapse
Affiliation(s)
- Fred Armbrust
- Biochemical Institute, Unit for Degradomics of the Protease Web, University of Kiel, Kiel, Germany
| | - Kira Bickenbach
- Biochemical Institute, Unit for Degradomics of the Protease Web, University of Kiel, Kiel, Germany
| | - Liana Marengo
- Institute for Pathobiochemistry, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Claus Pietrzik
- Institute for Pathobiochemistry, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.
| | - Christoph Becker-Pauly
- Biochemical Institute, Unit for Degradomics of the Protease Web, University of Kiel, Kiel, Germany.
| |
Collapse
|
16
|
Yu Y, Gao Y, Winblad B, Tjernberg LO, Schedin-Weiss S. A Super-Resolved View of the Alzheimer's Disease-Related Amyloidogenic Pathway in Hippocampal Neurons. J Alzheimers Dis 2021; 83:833-852. [PMID: 34366358 PMCID: PMC8543249 DOI: 10.3233/jad-215008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Processing of the amyloid-β protein precursor (AβPP) is neurophysiologically important due to the resulting fragments that regulate synapse biology, as well as potentially harmful due to generation of the 42 amino acid long amyloid β-peptide (Aβ42), which is a key player in Alzheimer's disease. OBJECTIVE Our aim was to clarify the subcellular locations of the fragments involved in the amyloidogenic pathway in primary neurons with a focus on Aβ42 and its immediate substrate AβPP C-terminal fragment (APP-CTF). To overcome the difficulties of resolving these compartments due to their small size, we used super-resolution microscopy. METHODS Mouse primary hippocampal neurons were immunolabelled and imaged by stimulated emission depletion (STED) microscopy, including three-dimensional three-channel imaging, and quantitative image analyses. RESULTS The first (β-secretase) and second (γ-secretase) cleavages of AβPP were localized to functionally and distally distinct compartments. The β-secretase cleavage was observed in early endosomes in soma, where we were able to show that the liberated N- and C-terminal fragments were sorted into distinct vesicles budding from the early endosomes. Lack of colocalization of Aβ42 and APP-CTF in soma suggested that γ-secretase cleavage occurs in neurites. Indeed, APP-CTF was, in line with Aβ42 in our previous study, enriched in the presynapse but absent from the postsynapse. In contrast, full-length AβPP was not detected in either the pre- or the postsynaptic side of the synapse. Furthermore, we observed that endogenously produced and endocytosed Aβ42 were localized in different compartments. CONCLUSION These findings provide critical super-resolved insight into amyloidogenic AβPP processing in primary neurons.
Collapse
Affiliation(s)
- Yang Yu
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| | - Yang Gao
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| | - Bengt Winblad
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden.,Theme Inflammation and Aging, Karolinska University Hospital, Huddinge, Sweden
| | - Lars O Tjernberg
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| | - Sophia Schedin-Weiss
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| |
Collapse
|
17
|
RNA Localization and Local Translation in Glia in Neurological and Neurodegenerative Diseases: Lessons from Neurons. Cells 2021; 10:cells10030632. [PMID: 33809142 PMCID: PMC8000831 DOI: 10.3390/cells10030632] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/03/2021] [Accepted: 03/09/2021] [Indexed: 12/11/2022] Open
Abstract
Cell polarity is crucial for almost every cell in our body to establish distinct structural and functional domains. Polarized cells have an asymmetrical morphology and therefore their proteins need to be asymmetrically distributed to support their function. Subcellular protein distribution is typically achieved by localization peptides within the protein sequence. However, protein delivery to distinct cellular compartments can rely, not only on the transport of the protein itself but also on the transport of the mRNA that is then translated at target sites. This phenomenon is known as local protein synthesis. Local protein synthesis relies on the transport of mRNAs to subcellular domains and their translation to proteins at target sites by the also localized translation machinery. Neurons and glia specially depend upon the accurate subcellular distribution of their proteome to fulfil their polarized functions. In this sense, local protein synthesis has revealed itself as a crucial mechanism that regulates proper protein homeostasis in subcellular compartments. Thus, deregulation of mRNA transport and/or of localized translation can lead to neurological and neurodegenerative diseases. Local translation has been more extensively studied in neurons than in glia. In this review article, we will summarize the state-of-the art research on local protein synthesis in neuronal function and dysfunction, and we will discuss the possibility that local translation in glia and deregulation thereof contributes to neurological and neurodegenerative diseases.
Collapse
|
18
|
Kedia S, Ramakrishna P, Netrakanti PR, Singh N, Sisodia SS, Jose M, Kumar S, Mahadevan A, Ramanan N, Nadkarni S, Nair D. Alteration in synaptic nanoscale organization dictates amyloidogenic processing in Alzheimer's disease. iScience 2020; 24:101924. [PMID: 33409475 PMCID: PMC7773964 DOI: 10.1016/j.isci.2020.101924] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 11/22/2020] [Accepted: 12/07/2020] [Indexed: 01/08/2023] Open
Abstract
Despite intuitive insights into differential proteolysis of amyloid precursor protein (APP), the stochasticity behind local product formation through amyloidogenic pathway at individual synapses remain unclear. Here, we show that the major components of amyloidogenic machinery namely, APP and secretases are discretely organized into nanodomains of high local concentration compared to their immediate environment in functional zones of the synapse. Additionally, with the aid of multiple models of Alzheimer's disease (AD), we confirm that this discrete nanoscale chemical map of amyloidogenic machinery is altered at excitatory synapses. Furthermore, we provide realistic models of amyloidogenic processing in unitary vesicles originating from the endocytic zone of excitatory synapses. Thus, we show how an alteration in the stochasticity of synaptic nanoscale organization contributes to the dynamic range of C-terminal fragments β (CTFβ) production, defining the heterogeneity of amyloidogenic processing at individual synapses, leading to long-term synaptic deficits as seen in AD. Components of amyloidogenic machinery are organized into nanodomains Assembly of nanodomains differs between functional zones of the synapse Stochasticity of nanoscale organization dictates dynamic range of APP proteolysis Variability in composition of amyloidogenic machinery is associated with AD
Collapse
Affiliation(s)
- Shekhar Kedia
- Centre for Neuroscience, Indian Institute of Science, Bangalore 560012, India
| | | | | | - Nivedita Singh
- Centre for Neuroscience, Indian Institute of Science, Bangalore 560012, India
| | - Sangram S Sisodia
- Center for Molecular Neurobiology, Department of Neurobiology, The University of Chicago, IL 60637, USA
| | - Mini Jose
- Centre for Neuroscience, Indian Institute of Science, Bangalore 560012, India
| | - Sathish Kumar
- Department of Neurology, University of Bonn, Bonn 53127, Germany
| | - Anita Mahadevan
- Department of Neuropathology, NIMHANS, Bangalore 560029, India
| | | | - Suhita Nadkarni
- Indian Institute of Science Education and Research, Pune 411008, India
| | - Deepak Nair
- Centre for Neuroscience, Indian Institute of Science, Bangalore 560012, India
| |
Collapse
|
19
|
Abstract
It is increasingly recognized that local protein synthesis (LPS) contributes to fundamental aspects of axon biology, in both developing and mature neurons. Mutations in RNA-binding proteins (RBPs), as central players in LPS, and other proteins affecting RNA localization and translation are associated with a range of neurological disorders, suggesting disruption of LPS may be of pathological significance. In this review, we substantiate this hypothesis by examining the link between LPS and key axonal processes, and the implicated pathophysiological consequences of dysregulated LPS. First, we describe how the length and autonomy of axons result in an exceptional reliance on LPS. We next discuss the roles of LPS in maintaining axonal structural and functional polarity and axonal trafficking. We then consider how LPS facilitates the establishment of neuronal connectivity through regulation of axonal branching and pruning, how it mediates axonal survival into adulthood and its involvement in neuronal stress responses.
Collapse
Affiliation(s)
- Julie Qiaojin Lin
- UK Dementia Research Institute at University of Cambridge, Department of Clinical Neurosciences, Island Research Building, Cambridge Biomedical Campus, Cambridge, UK
| | | | - Christine E Holt
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| |
Collapse
|
20
|
Korneva A, Schaub J, Jefferys J, Kimball E, Pease ME, Nawathe M, Johnson TV, Pitha I, Quigley H. A method to quantify regional axonal transport blockade at the optic nerve head after short term intraocular pressure elevation in mice. Exp Eye Res 2020; 196:108035. [PMID: 32353427 PMCID: PMC7335019 DOI: 10.1016/j.exer.2020.108035] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 03/28/2020] [Accepted: 04/08/2020] [Indexed: 01/07/2023]
Abstract
Axonal transport blockade is an initial step in retinal ganglion cell (RGC) degeneration in glaucoma and targeting maintenance of normal axonal transport could confer neuroprotection. We present an objective, quantitative method for assessing axonal transport blockade in mouse glaucoma models. Intraocular pressure (IOP) was elevated unilaterally in CD1 mice for 3 days using intracameral microbead injection. Longitudinal sections of optic nerve head (ONH) were immunofluorescently labeled for myelin basic protein (MBP) and amyloid precursor protein (APP), which is transported predominantly orthograde by neurons. The beginning of the myelin transition zone, visualized with the MBP label, was more posterior with elevated IOP, 288.8 ± 40.9 μm, compared to normotensive control eyes, 228.7 ± 32.7 μm (p = 0.030, N = 6 pairs). Glaucomatous regional APP accumulations in retina, prelaminar ONH, unmyelinated ONH, and myelinated optic nerve were identified by objective qualification of pixels with fluorescent intensity greater than the 97.5th percentile value of control eyes (suprathreshold pixels). This method segregated images with APP blockade from those with normal transport of APP. The fraction of suprathreshold pixels was significantly higher following IOP elevation than in normotensive controls in the unmyelinated ONH and myelinated nerve regions (paired analyses, p = 0.02 and 0.003, respectively, N = 12), but not in retina or prelaminar ONH (p = 0.91 and 0.08, respectively). The mean intensity of suprathreshold pixels was also significantly greater in glaucoma than in normotensive controls in prelaminar ONH, unmyelinated ONH and myelinated optic nerve (p = 0.01, 0.01, 0.002, respectively). Using this method, subconjunctival glyceraldehyde, which is known to worsen long-term RGC loss with IOP elevation, also produced greater APP blockade, but not statistically significant compared to glaucoma alone. Systemic losartan, which aids RGC axonal survival in glaucoma, reduced APP blockade, but not statistically significant compared to glaucoma alone. The method provides a short-term assessment of axonal injury for use in initial tests of neuroprotective therapies that may beneficially affect RGC transport in animal models of glaucoma.
Collapse
Affiliation(s)
- Arina Korneva
- From the Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Julie Schaub
- From the Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Joan Jefferys
- From the Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elizabeth Kimball
- From the Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mary Ellen Pease
- From the Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Manasi Nawathe
- From the Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Thomas V Johnson
- From the Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ian Pitha
- From the Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Harry Quigley
- From the Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
21
|
Wilson B, Geetha KM. Neurotherapeutic applications of nanomedicine for treating Alzheimer's disease. J Control Release 2020; 325:25-37. [PMID: 32473177 DOI: 10.1016/j.jconrel.2020.05.044] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 05/25/2020] [Accepted: 05/26/2020] [Indexed: 01/30/2023]
Abstract
Alzheimer's disease (AD) is a progressive, irreversible, fatal brain disease which disturbs cognitive functions. It affects 35 million people worldwide and the number of people suffering may increase to 100 million by 2050 if no effective treatments are available. The present treatment improves cognitive functions and provide temporary symptomatic relief, but do not stop or delay the disease progression. Moreover, they are mainly available as conventional oral dosage forms and these conventional oral medications lack brain specificity and also produce side effects which leads to poor patient compliance. Brain drug targeting by nanomedicines is a promising approach to improve brain targeting specificity, brain bioavailability and patient compliance. The present review discuses about the currently available pharmacotherapy for AD and the neurotherapeutic applications as well as the advancements of nanomedicine for treating AD. It also highlights the recent advancements of various nanomedicines containing phytopharmaceuticals for treating AD. It is believed that nanomedicines containing approved drugs can be transformed into the clinics hence improve the life style of AD patients.
Collapse
Affiliation(s)
- Barnabas Wilson
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Dayananda Sagar University, Kumaraswamy Layout, Bangalore, Karnataka 560078, India.
| | - Kannoth Mukundan Geetha
- Department of Pharmacology, College of Pharmaceutical Sciences, Dayananda Sagar University, Kumaraswamy Layout, Bangalore, Karnataka 560078, India
| |
Collapse
|
22
|
Bruyère J, Abada YS, Vitet H, Fontaine G, Deloulme JC, Cès A, Denarier E, Pernet-Gallay K, Andrieux A, Humbert S, Potier MC, Delatour B, Saudou F. Presynaptic APP levels and synaptic homeostasis are regulated by Akt phosphorylation of huntingtin. eLife 2020; 9:56371. [PMID: 32452382 PMCID: PMC7269668 DOI: 10.7554/elife.56371] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 05/11/2020] [Indexed: 02/06/2023] Open
Abstract
Studies have suggested that amyloid precursor protein (APP) regulates synaptic homeostasis, but the evidence has not been consistent. In particular, signaling pathways controlling APP transport to the synapse in axons and dendrites remain to be identified. Having previously shown that Huntingtin (HTT), the scaffolding protein involved in Huntington’s disease, regulates neuritic transport of APP, we used a microfluidic corticocortical neuronal network-on-a-chip to examine APP transport and localization to the pre- and post-synaptic compartments. We found that HTT, upon phosphorylation by the Ser/Thr kinase Akt, regulates APP transport in axons but not dendrites. Expression of an unphosphorylatable HTT decreased axonal anterograde transport of APP, reduced presynaptic APP levels, and increased synaptic density. Ablating in vivo HTT phosphorylation in APPPS1 mice, which overexpress APP, reduced presynaptic APP levels, restored synapse number and improved learning and memory. The Akt-HTT pathway and axonal transport of APP thus regulate APP presynaptic levels and synapse homeostasis.
Collapse
Affiliation(s)
- Julie Bruyère
- Univ. Grenoble Alpes, Inserm, U1216, CHU Grenoble Alpes, CEA, Grenoble Institut Neurosciences, Grenoble, France
| | - Yah-Se Abada
- Institut du Cerveau et de la Moelle épinière, ICM, Inserm U1127, CNRS UMR 7225, Sorbonne Université, Paris, France
| | - Hélène Vitet
- Univ. Grenoble Alpes, Inserm, U1216, CHU Grenoble Alpes, CEA, Grenoble Institut Neurosciences, Grenoble, France
| | - Gaëlle Fontaine
- Institut du Cerveau et de la Moelle épinière, ICM, Inserm U1127, CNRS UMR 7225, Sorbonne Université, Paris, France
| | - Jean-Christophe Deloulme
- Univ. Grenoble Alpes, Inserm, U1216, CHU Grenoble Alpes, CEA, Grenoble Institut Neurosciences, Grenoble, France
| | - Aurélia Cès
- Institut du Cerveau et de la Moelle épinière, ICM, Inserm U1127, CNRS UMR 7225, Sorbonne Université, Paris, France
| | - Eric Denarier
- Univ. Grenoble Alpes, Inserm, U1216, CHU Grenoble Alpes, CEA, Grenoble Institut Neurosciences, Grenoble, France
| | - Karin Pernet-Gallay
- Univ. Grenoble Alpes, Inserm, U1216, CHU Grenoble Alpes, CEA, Grenoble Institut Neurosciences, Grenoble, France
| | - Annie Andrieux
- Univ. Grenoble Alpes, Inserm, U1216, CHU Grenoble Alpes, CEA, Grenoble Institut Neurosciences, Grenoble, France
| | - Sandrine Humbert
- Univ. Grenoble Alpes, Inserm, U1216, CHU Grenoble Alpes, CEA, Grenoble Institut Neurosciences, Grenoble, France
| | - Marie-Claude Potier
- Institut du Cerveau et de la Moelle épinière, ICM, Inserm U1127, CNRS UMR 7225, Sorbonne Université, Paris, France
| | - Benoît Delatour
- Institut du Cerveau et de la Moelle épinière, ICM, Inserm U1127, CNRS UMR 7225, Sorbonne Université, Paris, France
| | - Frédéric Saudou
- Univ. Grenoble Alpes, Inserm, U1216, CHU Grenoble Alpes, CEA, Grenoble Institut Neurosciences, Grenoble, France
| |
Collapse
|
23
|
Hermey G, Hoffmeister-Ullerich SA, Merz B, Groß D, Kuhl D, Kins S. Amyloidosis causes downregulation of SorLA, SorCS1 and SorCS3 expression in mice. Biol Chem 2020; 400:1181-1189. [PMID: 31095505 DOI: 10.1515/hsz-2019-0146] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 04/03/2019] [Indexed: 12/30/2022]
Abstract
Accumulation of β-amyloid peptide (Aβ) is regarded as a primary cause of Alzheimer's disease (AD). Aβ is derived by sequential cleavage of the amyloid precursor protein (APP). Alterations in the subcellular targeting of APP are thought to affect the degree of Aβ production. Sorting receptors, such as SorLA, convey subcellular targeting of APP. Dysfunction of SorLA, and likely of the related receptors SorCS1 and SorCS3, cause AD. Nevertheless, disease progression could also provoke altered expression of the receptors. Here, we assessed if Aβ plaque formation promotes altered expression of SorLA, SorCS1 and SorCS3. We analyzed transcript levels during aging and after amyloidosis in brain areas characterized by early amyloid plaque formation in an AD mouse model (APPPS1) and wild types. We observed stable expression levels during aging (1-12 months). After plaque formation, SorCS1 and SorLA expression were markedly reduced in the frontal cerebral cortex and to a minor extent in the hippocampus, whereas SorCS3 expression was solely reduced in the frontal cerebral cortex. Our results indicate that disease progression, associated with Aβ accumulation, can negatively regulate expression of the receptors.
Collapse
Affiliation(s)
- Guido Hermey
- Institute for Molecular and Cellular Cognition, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, D-20251 Hamburg, Germany
| | - Sabine A Hoffmeister-Ullerich
- Bioanalytics, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, D-20251 Hamburg, Germany
| | - Barbara Merz
- Institute for Molecular and Cellular Cognition, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, D-20251 Hamburg, Germany
| | - Dagmar Groß
- Division of Human Biology and Human Genetics, University of Kaiserslautern, Erwin-Schrödinger-Straße 13, D-67663 Kaiserslautern, Germany
| | - Dietmar Kuhl
- Institute for Molecular and Cellular Cognition, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, D-20251 Hamburg, Germany
| | - Stefan Kins
- Division of Human Biology and Human Genetics, University of Kaiserslautern, Erwin-Schrödinger-Straße 13, D-67663 Kaiserslautern, Germany
| |
Collapse
|
24
|
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease caused by eventually aggregated amyloid β (Aβ) plaques in degenerating neurons of the aging brain. These aggregated protein plaques mainly consist of Aβ fibrils and neurofibrillary tangles (NFTs) of phosphorylated tau protein. Even though some cholinesterase inhibitors, NMDA receptor antagonist, and monoclonal antibodies were developed to inhibit neurodegeneration or activate neural regeneration or clear off the Aβ deposits, none of the treatment is effective in improving the cognitive and memory dysfunctions of the AD patients. Thus, stem cell therapy represents a powerful tool for the treatment of AD. In addition to discussing the advents in molecular pathogenesis and animal models of this disease and the treatment approaches using small molecules and immunoglobulins against AD, we will focus on the stem cell sources for AD using neural stem cells (NSCs); embryonic stem cells (ESCs); and mesenchymal stem cells (MSCs) from bone marrow, umbilical cord, and umbilical cord blood. In particular, patient-specific-induced pluripotent stem cells (iPS cells) are proposed as a future prospective and the challenges for the treatment of AD.
Collapse
|
25
|
The Endolysosomal System and Proteostasis: From Development to Degeneration. J Neurosci 2019; 38:9364-9374. [PMID: 30381428 DOI: 10.1523/jneurosci.1665-18.2018] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 09/17/2018] [Accepted: 09/20/2018] [Indexed: 12/12/2022] Open
Abstract
How do neurons adapt their endolysosomal system to address the particular challenge of membrane transport across their elaborate cellular landscape and to maintain proteostasis for the lifetime of the organism? Here we review recent findings that address this central question. We discuss the cellular and molecular mechanisms of endolysosomal trafficking and the autophagy pathway in neurons, as well as their role in neuronal development and degeneration. These studies highlight the importance of understanding the basic cell biology of endolysosomal trafficking and autophagy and their roles in the maintenance of proteostasis within the context of neurons, which will be critical for developing effective therapies for various neurodevelopmental and neurodegenerative disorders.
Collapse
|
26
|
Liu S, Sun YP, Gao XL, Sui Y. Knowledge domain and emerging trends in Alzheimer's disease: a scientometric review based on CiteSpace analysis. Neural Regen Res 2019; 14:1643-1650. [PMID: 31089065 PMCID: PMC6557102 DOI: 10.4103/1673-5374.255995] [Citation(s) in RCA: 124] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 04/04/2019] [Indexed: 11/06/2022] Open
Abstract
Alzheimer's disease is the most common cause of dementia. It is an increasingly serious global health problem and has a significant impact on individuals and society. However, the precise cause of Alzheimer's disease is still unknown. In this study, 11,748 Web-of-Science-indexed manuscripts regarding Alzheimer's disease, all published from 2015 to 2019, and their 693,938 references were analyzed. A document co-citation network map was drawn using CiteSpace software. Research frontiers and development trends were determined by retrieving subject headings with apparent changing word frequency trends, which can be used to forecast future research developments in Alzheimer's disease.
Collapse
Affiliation(s)
- Shuo Liu
- The Fourth People's Hospital of Shenyang, Shenyang, Liaoning Province, China
| | - Ya-Ping Sun
- The Fourth People's Hospital of Shenyang, Shenyang, Liaoning Province, China
| | - Xu-Ling Gao
- The Fourth People's Hospital of Shenyang, Shenyang, Liaoning Province, China
| | - Yi Sui
- The First People's Hospital of Shenyang, Shenyang, Liaoning Province, China
| |
Collapse
|
27
|
Fan YG, Guo T, Han XR, Liu JL, Cai YT, Xue H, Huang XS, Li YC, Wang ZY, Guo C. Paricalcitol accelerates BACE1 lysosomal degradation and inhibits calpain-1 dependent neuronal loss in APP/PS1 transgenic mice. EBioMedicine 2019; 45:393-407. [PMID: 31303501 PMCID: PMC6642335 DOI: 10.1016/j.ebiom.2019.07.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 07/01/2019] [Accepted: 07/04/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Recent studies have revealed that vitamin D deficiency may increase the risk of Alzheimer's disease, and vitamin D supplementation may be effective strategy to ameliorate the neurodegenerative process in Alzheimer's disease patients. Paricalcitol (PAL), a low-calcemic vitamin D receptor agonist, is clinically used to treat secondary hyperparathyroidism. However, the potential application of PAL for treating neurodegenerative disorders remains unexplored. METHODS The APP/PS1 mice were intraperitoneally injected with PAL or vehicle every other day for 15 weeks. The β-amyloid (Aβ) production was confirmed using immunostaining and enzyme linked immunosorbent assay. The underlying mechanism was verified by western blot and immunostaining in vivo and in vitro. FINDINGS Long-term PAL treatment clearly reduced β-amyloid (Aβ) generation and neuronal loss in APP/PS1 transgenic mouse brains. PAL stimulated the expression of low-density lipoprotein receptor-related protein 1 (LRP1) possibly through inhibiting sterol regulatory element binding protein-2 (SREBP2); PAL also promoted LRP1-mediated β-site APP cleavage enzyme 1 (BACE1) transport to late endosomes, thus increasing the lysosomal degradation of BACE1. Furthermore, PAL diminished 8-hydroxyguanosine (8-OHdG) generation in neuronal mitochondria via enhancing base excision repair (BER), resulting in the attenuation of calpain-1-mediated neuronal loss. INTERPRETATION The present data demonstrate that PAL can reduce Aβ generation through accelerating BACE1 lysosomal degradation and can inhibit neuronal loss through suppressing mitochondrial 8-OHdG generation. Hence, PAL might be a promising agent for treating Alzheimer's disease. FUND: This study was financially supported by the Natural Science Foundation of China (U1608282).
Collapse
Affiliation(s)
- Yong-Gang Fan
- College of Life and Health Sciences, Northeastern University, NO.195, Chuangxin Road, Hunnan District, Shenyang 110169, China
| | - Tian Guo
- College of Life and Health Sciences, Northeastern University, NO.195, Chuangxin Road, Hunnan District, Shenyang 110169, China
| | - Xiao-Ran Han
- College of Life and Health Sciences, Northeastern University, NO.195, Chuangxin Road, Hunnan District, Shenyang 110169, China
| | - Jun-Lin Liu
- College of Life and Health Sciences, Northeastern University, NO.195, Chuangxin Road, Hunnan District, Shenyang 110169, China
| | - Yu-Ting Cai
- College of Life and Health Sciences, Northeastern University, NO.195, Chuangxin Road, Hunnan District, Shenyang 110169, China
| | - Han Xue
- College of Life and Health Sciences, Northeastern University, NO.195, Chuangxin Road, Hunnan District, Shenyang 110169, China
| | - Xue-Shi Huang
- College of Life and Health Sciences, Northeastern University, NO.195, Chuangxin Road, Hunnan District, Shenyang 110169, China
| | - Yan-Chun Li
- Department of Medicine, the University of Chicago, Chicago, IL 60637, USA
| | - Zhan-You Wang
- College of Life and Health Sciences, Northeastern University, NO.195, Chuangxin Road, Hunnan District, Shenyang 110169, China; Institute of Health Sciences, Key Laboratory of Medical Cell Biology of Ministry of Education, China Medical University, Shenyang 110122, China.
| | - Chuang Guo
- College of Life and Health Sciences, Northeastern University, NO.195, Chuangxin Road, Hunnan District, Shenyang 110169, China.
| |
Collapse
|
28
|
Regulatory role of Golgi brefeldin A resistance factor‐1 in amyloid precursor protein trafficking, cleavage and Aβ formation. J Cell Biochem 2019; 120:15604-15615. [DOI: 10.1002/jcb.28827] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 01/11/2019] [Accepted: 01/14/2019] [Indexed: 01/19/2023]
|
29
|
Carter CJ. Autism genes and the leukocyte transcriptome in autistic toddlers relate to pathogen interactomes, infection and the immune system. A role for excess neurotrophic sAPPα and reduced antimicrobial Aβ. Neurochem Int 2019; 126:36-58. [PMID: 30862493 DOI: 10.1016/j.neuint.2019.03.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 02/22/2019] [Accepted: 03/06/2019] [Indexed: 12/20/2022]
Abstract
Prenatal and early childhood infections have been implicated in autism. Many autism susceptibility genes (206 Autworks genes) are localised in the immune system and are related to immune/infection pathways. They are enriched in the host/pathogen interactomes of 18 separate microbes (bacteria/viruses and fungi) and to the genes regulated by bacterial toxins, mycotoxins and Toll-like receptor ligands. This enrichment was also observed for misregulated genes from a microarray study of leukocytes from autistic toddlers. The upregulated genes from this leukocyte study also matched the expression profiles in response to numerous infectious agents from the Broad Institute molecular signatures database. They also matched genes related to sudden infant death syndrome and autism comorbid conditions (autoimmune disease, systemic lupus erythematosus, diabetes, epilepsy and cardiomyopathy) as well as to estrogen and thyrotropin responses and to those upregulated by different types of stressors including oxidative stress, hypoxia, endoplasmic reticulum stress, ultraviolet radiation or 2,4-dinitrofluorobenzene, a hapten used to develop allergic skin reactions in animal models. The oxidative/integrated stress response is also upregulated in the autism brain and may contribute to myelination problems. There was also a marked similarity between the expression signatures of autism and Alzheimer's disease, and 44 shared autism/Alzheimer's disease genes are almost exclusively expressed in the blood-brain barrier. However, in contrast to Alzheimer's disease, levels of the antimicrobial peptide beta-amyloid are decreased and the levels of the neurotrophic/myelinotrophic soluble APP alpha are increased in autism, together with an increased activity of α-secretase. sAPPα induces an increase in glutamatergic and a decrease in GABA-ergic synapses creating and excitatory/inhibitory imbalance that has also been observed in autism. A literature survey showed that multiple autism genes converge on APP processing and that many are able to increase sAPPalpha at the expense of beta-amyloid production. A genetically programmed tilt of this axis towards an overproduction of neurotrophic/gliotrophic sAPPalpha and underproduction of antimicrobial beta-amyloid may explain the brain overgrowth and myelination dysfunction, as well as the involvement of pathogens in autism.
Collapse
Affiliation(s)
- C J Carter
- PolygenicPathways, 41C Marina, Saint Leonard's on Sea, TN38 0BU, East Sussex, UK.
| |
Collapse
|
30
|
Majolo F, Marinowic DR, Machado DC, Da Costa JC. Important advances in Alzheimer's disease from the use of induced pluripotent stem cells. J Biomed Sci 2019; 26:15. [PMID: 30728025 PMCID: PMC6366077 DOI: 10.1186/s12929-019-0501-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 01/09/2019] [Indexed: 12/14/2022] Open
Abstract
Among the various types of dementia, Alzheimer’s disease (AD) is the most prevalent and is clinically defined as the appearance of progressive deficits in cognition and memory. Considering that AD is a central nervous system disease, getting tissue from the patient to study the disease before death is challenging. The discovery of the technique called induced pluripotent stem cells (iPSCs) allows to reprogram the patient’s somatic cells to a pluripotent state by the forced expression of a defined set of transcription factors. Many studies have shown promising results and made important conclusions beyond AD using iPSCs approach. Due to the accumulating knowledge related to this topic and the important advances obtained until now, we review, using PubMed, and present an update of all publications related to AD from the use of iPSCs. The first iPSCs generated for AD were carried out in 2011 by Yahata et al. (PLoS One 6:e25788, 2011) and Yaqi et al. (Hum Mol Genet 20:4530–9, 2011). Like other authors, both authors used iPSCs as a pre-clinical tool for screening therapeutic compounds. This approach is also essential to model AD, testing early toxicity and efficacy, and developing a platform for drug development. Considering that the iPSCs technique is relatively recent, we can consider that the AD field received valuable contributions from iPSCs models, contributing to our understanding and the treatment of this devastating disorder.
Collapse
Affiliation(s)
- Fernanda Majolo
- Brain Institute of Rio Grande do Sul (BraIns), Postgraduate Program in Medicine and Health Sciences (PUCRS), Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, 90610000, Brazil.
| | - Daniel Rodrigo Marinowic
- Brain Institute of Rio Grande do Sul (BraIns), Postgraduate Program in Medicine and Health Sciences (PUCRS), Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, 90610000, Brazil
| | - Denise Cantarelli Machado
- Brain Institute of Rio Grande do Sul (BraIns), Postgraduate Program in Medicine and Health Sciences (PUCRS), Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, 90610000, Brazil
| | - Jaderson Costa Da Costa
- Brain Institute of Rio Grande do Sul (BraIns), Postgraduate Program in Medicine and Health Sciences (PUCRS), Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, 90610000, Brazil
| |
Collapse
|
31
|
Zhao X, Wang X, Su G, Sun Q, Fu J, Zhang H, Teng J. The effect of early growth response 1 on levels of Amyloid-β 40 peptide in U87MG cells. J Cell Biochem 2018; 120:3514-3519. [PMID: 30548663 DOI: 10.1002/jcb.27627] [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: 06/15/2018] [Accepted: 08/14/2018] [Indexed: 11/11/2022]
Abstract
A recent study has shown that early growth response 1 (EGR1) plays a critical role in the β-amyloid cascade and tau hypotheses. In addition, evidence has suggested that EGR1 can regulate levels of amyloid-beta peptides, key molecules in the pathogenesis of Alzheimer's disease (AD). However, whether EGR1 is a deleterious or protective factor in the AD is still controversial. In this present study, we constructed an overexpression plasmid, CMV-EGFP-EGR1-Kanamycin, and transfected it into U87MG cells to investigate the effects of EGR1 expression on amyloid-β (1-40) peptide (Aβ40) levels. U87MG cells transfected by CMV-EGFP-EGR1-Kanamycin and CMV-EGFP-Kanamycin were assigned, respectively, to experimental and control groups. Fluorescence microscopy was used to observe transfection efficiencies of the plasmids after 6 hours. EGR1 messenger RNA levels were measured by quantitative reverse transcription polymerase chain reaction. Aβ40 secretion was analyzed by enzyme-linked immunosorbent assay. Expression of the amyloid precursor protein, beta-secretase enzyme, and presenilin 1 proteins were analyzed by Western blot analysis. The results showed that EGR1 overexpression increased Aβ40 secretion in vitro, possibly through increasing BACE1 expression. Based on these results, EGR1 might be a promising therapeutic target for the AD.
Collapse
Affiliation(s)
- Xinyu Zhao
- Department of Neurology, The Frist Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaojie Wang
- Department of Neurology, The Frist Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Gang Su
- Department of Neurology, The Frist Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qi Sun
- Department of Neurology, The Frist Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jitong Fu
- Department of Neurology, The Frist Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Huili Zhang
- Department of Neurology, The Frist Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Junfang Teng
- Department of Neurology, The Frist Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
32
|
Kuznetsov IA, Kuznetsov AV. How the formation of amyloid plaques and neurofibrillary tangles may be related: a mathematical modelling study. Proc Math Phys Eng Sci 2018; 474:20170777. [PMID: 29507520 PMCID: PMC5832841 DOI: 10.1098/rspa.2017.0777] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 01/12/2018] [Indexed: 12/12/2022] Open
Abstract
We develop a mathematical model that enables us to investigate possible mechanisms by which two primary markers of Alzheimer's disease (AD), extracellular amyloid plaques and intracellular tangles, may be related. Our model investigates the possibility that the decay of anterograde axonal transport of amyloid precursor protein (APP), caused by toxic tau aggregates, leads to decreased APP transport towards the synapse and APP accumulation in the soma. The developed model thus couples three processes: (i) slow axonal transport of tau, (ii) tau misfolding and agglomeration, which we simulated by using the Finke-Watzky model and (iii) fast axonal transport of APP. Because the timescale for tau agglomeration is much larger than that for tau transport, we suggest using the quasi-steady-state approximation for formulating and solving the governing equations for these three processes. Our results suggest that misfolded tau most likely accumulates in the beginning of the axon. The analysis of APP transport suggests that APP will also likely accumulate in the beginning of the axon, causing an increased APP concentration in this region, which could be interpreted as a 'traffic jam'. The APP flux towards the synapse is significantly reduced by tau misfolding, but not due to the APP traffic jam, which can be viewed as a symptom, but rather due to the reduced affinity of kinesin-1 motors to APP-transporting vesicles.
Collapse
Affiliation(s)
- I. A. Kuznetsov
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - A. V. Kuznetsov
- Department of Mechanical and Aerospace Engineering, North Carolina State University, Raleigh, NC 27695–7910, USA
| |
Collapse
|
33
|
Sharoar MG, Yan R. Effects of altered RTN3 expression on BACE1 activity and Alzheimer's neuritic plaques. Rev Neurosci 2018; 28:145-154. [PMID: 27883331 DOI: 10.1515/revneuro-2016-0054] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Accepted: 09/20/2016] [Indexed: 12/15/2022]
Abstract
Reticulon 3 (RTN3), which is a member of the reticulon family of proteins, has a biochemical function of shaping tubular endoplasmic reticulum. RTN3 has also been found to interact with β-site amyloid precursor protein cleaving enzyme 1 (BACE1), which initiates the generation of β-amyloid peptides (Aβ) from amyloid precursor protein. Aβ is the major proteinaceous component in neuritic plaques, which constitute one of the major pathological features in brains of Alzheimer's disease (AD) patients. Mice deficient in or overexpressing RTN3 have altered amyloid deposition through effects on BACE1 expression and activity. In this review, we will summarize the current findings concerning the role of RTN3 in AD pathogenesis and demonstrate that RTN3 protein levels act as age-dependent modulators of BACE1 activity and Aβ deposition during the pathogenic progression of AD.
Collapse
|
34
|
Eggert S, Gonzalez AC, Thomas C, Schilling S, Schwarz SM, Tischer C, Adam V, Strecker P, Schmidt V, Willnow TE, Hermey G, Pietrzik CU, Koo EH, Kins S. Dimerization leads to changes in APP (amyloid precursor protein) trafficking mediated by LRP1 and SorLA. Cell Mol Life Sci 2018; 75:301-322. [PMID: 28799085 PMCID: PMC11105302 DOI: 10.1007/s00018-017-2625-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 07/17/2017] [Accepted: 08/08/2017] [Indexed: 12/22/2022]
Abstract
Proteolytic cleavage of the amyloid precursor protein (APP) by α-, β- and γ-secretases is a determining factor in Alzheimer's disease (AD). Imbalances in the activity of all three enzymes can result in alterations towards pathogenic Aβ production. Proteolysis of APP is strongly linked to its subcellular localization as the secretases involved are distributed in different cellular compartments. APP has been shown to dimerize in cis-orientation, affecting Aβ production. This might be explained by different substrate properties defined by the APP oligomerization state or alternatively by altered APP monomer/dimer localization. We investigated the latter hypothesis using two different APP dimerization systems in HeLa cells. Dimerization caused a decreased localization of APP to the Golgi and at the plasma membrane, whereas the levels in the ER and in endosomes were increased. Furthermore, we observed via live cell imaging and biochemical analyses that APP dimerization affects its interaction with LRP1 and SorLA, suggesting that APP dimerization modulates its interplay with sorting molecules and in turn its localization and processing. Thus, pharmacological approaches targeting APP oligomerization properties might open novel strategies for treatment of AD.
Collapse
Affiliation(s)
- Simone Eggert
- Department of Human Biology and Human Genetics, University of Kaiserslautern, Erwin-Schrödinger-Str. 13, 67663, Kaiserslautern, Germany.
| | - A C Gonzalez
- Department of Human Biology and Human Genetics, University of Kaiserslautern, Erwin-Schrödinger-Str. 13, 67663, Kaiserslautern, Germany
- Institute for Biochemistry, Christian Albrechts University Kiel, 24118, Kiel, Germany
| | - C Thomas
- Department of Human Biology and Human Genetics, University of Kaiserslautern, Erwin-Schrödinger-Str. 13, 67663, Kaiserslautern, Germany
| | - S Schilling
- Department of Human Biology and Human Genetics, University of Kaiserslautern, Erwin-Schrödinger-Str. 13, 67663, Kaiserslautern, Germany
| | - S M Schwarz
- Department of Human Biology and Human Genetics, University of Kaiserslautern, Erwin-Schrödinger-Str. 13, 67663, Kaiserslautern, Germany
- Institute for Medical Virology, University of Frankfurt, 60596, Frankfurt, Germany
| | | | - V Adam
- Department of Human Biology and Human Genetics, University of Kaiserslautern, Erwin-Schrödinger-Str. 13, 67663, Kaiserslautern, Germany
| | - P Strecker
- Department of Human Biology and Human Genetics, University of Kaiserslautern, Erwin-Schrödinger-Str. 13, 67663, Kaiserslautern, Germany
| | - V Schmidt
- Max Delbrueck Center for Molecular Medicine, Berlin, Germany
| | - T E Willnow
- Max Delbrueck Center for Molecular Medicine, Berlin, Germany
| | - G Hermey
- Institute for Molecular and Cellular Cognition, Center for Molecular University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - C U Pietrzik
- Institute for Pathobiochemistry, Molecular Neurodegeneration, University Medical Center of the Johannes Gutenberg-University Mainz, 55099, Mainz, Germany
| | - E H Koo
- Department of Neuroscience, University of California San Diego (UCSD), La Jolla, CA, 92093-0662, USA
| | - Stefan Kins
- Department of Human Biology and Human Genetics, University of Kaiserslautern, Erwin-Schrödinger-Str. 13, 67663, Kaiserslautern, Germany.
| |
Collapse
|
35
|
Musardo S, Marcello E. Synaptic dysfunction in Alzheimer's disease: From the role of amyloid β-peptide to the α-secretase ADAM10. Eur J Pharmacol 2017. [DOI: 10.1016/j.ejphar.2017.06.018] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
36
|
Trafficking in Alzheimer's Disease: Modulation of APP Transport and Processing by the Transmembrane Proteins LRP1, SorLA, SorCS1c, Sortilin, and Calsyntenin. Mol Neurobiol 2017; 55:5809-5829. [PMID: 29079999 DOI: 10.1007/s12035-017-0806-x] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 10/17/2017] [Indexed: 12/11/2022]
Abstract
The amyloid precursor protein (APP), one key player in Alzheimer's disease (AD), is extensively processed by different proteases. This leads to the generation of diverging fragments including the amyloid β (Aβ) peptide, which accumulates in brains of AD patients. Subcellular trafficking of APP is an important aspect for its proteolytic conversion, since the various secretases which cleave APP are located in different cellular compartments. As a consequence, altered subcellular targeting of APP is thought to directly affect the degree to which Aβ is generated. The mechanisms underlying intracellular APP transport are critical to understand AD pathogenesis and can serve as a target for future pharmacological interventions. In the recent years, a number of APP interacting proteins were identified which are implicated in sorting of APP, thereby influencing APP processing at different angles of the secretory or endocytic pathway. This review provides an update on the proteolytic processing of APP and the interplay of the transmembrane proteins low-density lipoprotein receptor-related protein 1, sortilin-receptor with A-type repeats, SorCS1c, sortilin, and calsyntenin. We discuss the specific interactions with APP, the capacity to modulate the intracellular itinerary and the proteolytic conversion of APP, a possible involvement in the clearance of Aβ, and the implications of these transmembrane proteins in AD and other neurodegenerative diseases.
Collapse
|
37
|
Li JM, Huang LL, Liu F, Tang BS, Yan XX. Can brain impermeable BACE1 inhibitors serve as anti-CAA medicine? BMC Neurol 2017; 17:163. [PMID: 28841840 PMCID: PMC5574137 DOI: 10.1186/s12883-017-0942-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 08/14/2017] [Indexed: 12/21/2022] Open
Abstract
Background Cerebral amyloid angiopathy (CAA) is characterized by the deposition of ß-amyloid peptides (Aß) in and surrounding the wall of microvasculature in the central nervous system, together with parenchymal amyloid plaques collectively referred to as cerebral amyloidosis, which occurs in the brain commonly among the elderly and more frequently in patients with Alzheimer’s disease (AD). CAA is associated with vascular injury and may cause devastating neurological outcomes. No therapeutic approach is available for this lesion to date. Main body ß-Secretase 1 (BACE1) is the enzyme initiating Aß production. Brain permeable BACE1 inhibitors targeting primarily at the parenchymal plaque pathology are currently evaluated in clinical trials. This article presents findings in support of a role of BACE1 elevation in the development of CAA, in addition to plaque pathogenesis. The rationale, feasibility, benefit and strategic issues for developing BACE1 inhibitors against CAA are discussed. Brain impermeable compounds are considered preferable as they might exhibit sufficient anti-CAA efficacy without causing significant neuronal/synaptic side effects. Conclusion Early pharmacological intervention to the pathogenesis of CAA is expected to provide significant protection for cerebral vascular health and hence brain health. Brain impermeable BACE1 inhibitors should be optimized and tested as potential anti-CAA therapeutics.
Collapse
Affiliation(s)
- Jian-Ming Li
- Department of Neurology & Key Laboratory of Hunan Province in Neurodegenerative Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.,Neuroscience Research Center, Changsha Medical University, Changsha, Hunan, 410219, China
| | - Li-Ling Huang
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Science, Changsha, Hunan, 410013, China
| | - Fei Liu
- Department of Neurosurgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
| | - Bei-Sha Tang
- Department of Neurology & Key Laboratory of Hunan Province in Neurodegenerative Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Xiao-Xin Yan
- Department of Neurology & Key Laboratory of Hunan Province in Neurodegenerative Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China. .,Department of Anatomy and Neurobiology, Central South University School of Basic Medical Science, Changsha, Hunan, 410013, China.
| |
Collapse
|
38
|
Sosa LJ, Cáceres A, Dupraz S, Oksdath M, Quiroga S, Lorenzo A. The physiological role of the amyloid precursor protein as an adhesion molecule in the developing nervous system. J Neurochem 2017; 143:11-29. [PMID: 28677143 DOI: 10.1111/jnc.14122] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 06/28/2017] [Accepted: 06/29/2017] [Indexed: 12/12/2022]
Abstract
The amyloid precursor protein (APP) is a type I transmembrane glycoprotein better known for its participation in the physiopathology of Alzheimer disease as the source of the beta amyloid fragment. However, the physiological functions of the full length protein and its proteolytic fragments have remained elusive. APP was first described as a cell-surface receptor; nevertheless, increasing evidence highlighted APP as a cell adhesion molecule. In this review, we will focus on the current knowledge of the physiological role of APP as a cell adhesion molecule and its involvement in key events of neuronal development, such as migration, neurite outgrowth, growth cone pathfinding, and synaptogenesis. Finally, since APP is over-expressed in Down syndrome individuals because of the extra copy of chromosome 21, in the last section of the review, we discuss the potential contribution of APP to the neuronal and synaptic defects described in this genetic condition. Read the Editorial Highlight for this article on page 9. Cover Image for this issue: doi. 10.1111/jnc.13817.
Collapse
Affiliation(s)
- Lucas J Sosa
- Departamento de Química Biológica Ranwell Caputto, Facultad de Ciencias Químicas, CIQUIBIC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Alfredo Cáceres
- Laboratorio Neurobiología, Instituto Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina.,Instituto Universitario Ciencias Biomédicas Córdoba, Córdoba, Argentina
| | - Sebastián Dupraz
- Axonal Growth and Regeneration, German Center for Neurodegenarative Diseases, Bonn, Germany
| | - Mariana Oksdath
- Departamento de Química Biológica Ranwell Caputto, Facultad de Ciencias Químicas, CIQUIBIC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Santiago Quiroga
- Departamento de Química Biológica Ranwell Caputto, Facultad de Ciencias Químicas, CIQUIBIC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Alfredo Lorenzo
- Laboratorio de Neuropatología Experimental, Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
39
|
Hefter D, Draguhn A. APP as a Protective Factor in Acute Neuronal Insults. Front Mol Neurosci 2017; 10:22. [PMID: 28210211 PMCID: PMC5288400 DOI: 10.3389/fnmol.2017.00022] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 01/16/2017] [Indexed: 12/25/2022] Open
Abstract
Despite its key role in the molecular pathology of Alzheimer’s disease (AD), the physiological function of amyloid precursor protein (APP) is unknown. Increasing evidence, however, points towards a neuroprotective role of this membrane protein in situations of metabolic stress. A key observation is the up-regulation of APP following acute (stroke, cardiac arrest) or chronic (cerebrovascular disease) hypoxic-ischemic conditions. While this mechanism may increase the risk or severity of AD, APP by itself or its soluble extracellular fragment APPsα can promote neuronal survival. Indeed, different animal models of acute hypoxia-ischemia, traumatic brain injury (TBI) and excitotoxicity have revealed protective effects of APP or APPsα. The underlying mechanisms involve APP-mediated regulation of calcium homeostasis via NMDA receptors (NMDAR), voltage-gated calcium channels (VGCC) or internal calcium stores. In addition, APP affects the expression of survival- or apoptosis-related genes as well as neurotrophic factors. In this review, we summarize the current understanding of the neuroprotective role of APP and APPsα and possible implications for future research and new therapeutic strategies.
Collapse
Affiliation(s)
- Dimitri Hefter
- Institute of Physiology and Pathophysiology, Heidelberg UniversityHeidelberg, Germany; Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg UniversityMannheim, Germany
| | - Andreas Draguhn
- Institute of Physiology and Pathophysiology, Heidelberg University Heidelberg, Germany
| |
Collapse
|
40
|
Wild K, August A, Pietrzik CU, Kins S. Structure and Synaptic Function of Metal Binding to the Amyloid Precursor Protein and its Proteolytic Fragments. Front Mol Neurosci 2017; 10:21. [PMID: 28197076 PMCID: PMC5281630 DOI: 10.3389/fnmol.2017.00021] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 01/16/2017] [Indexed: 12/19/2022] Open
Abstract
Alzheimer’s disease (AD) is ultimately linked to the amyloid precursor protein (APP). However, current research reveals an important synaptic function of APP and APP-like proteins (APLP1 and 2). In this context various neurotrophic and neuroprotective functions have been reported for the APP proteolytic fragments sAPPα, sAPPβ and the monomeric amyloid-beta peptide (Aβ). APP is a metalloprotein and binds copper and zinc ions. Synaptic activity correlates with a release of these ions into the synaptic cleft and dysregulation of their homeostasis is linked to different neurodegenerative diseases. Metal binding to APP or its fragments affects its structure and its proteolytic cleavage and therefore its physiological function at the synapse. Here, we summarize the current data supporting this hypothesis and provide a model of how these different mechanisms might be intertwined with each other.
Collapse
Affiliation(s)
- Klemens Wild
- Heidelberg University Biochemistry Center (BZH), University of Heidelberg Heidelberg, Germany
| | - Alexander August
- Division of Human Biology and Human Genetics, Technical University of Kaiserslautern Kaiserslautern, Germany
| | - Claus U Pietrzik
- Institute for Pathobiochemistry, University Medical Center of the Johannes Gutenberg-University Mainz Mainz, Germany
| | - Stefan Kins
- Division of Human Biology and Human Genetics, Technical University of Kaiserslautern Kaiserslautern, Germany
| |
Collapse
|
41
|
Barrera-Ocampo A, Arlt S, Matschke J, Hartmann U, Puig B, Ferrer I, Zürbig P, Glatzel M, Sepulveda-Falla D, Jahn H. Amyloid-β Precursor Protein Modulates the Sorting of Testican-1 and Contributes to Its Accumulation in Brain Tissue and Cerebrospinal Fluid from Patients with Alzheimer Disease. J Neuropathol Exp Neurol 2016; 75:903-16. [PMID: 27486134 PMCID: PMC5015660 DOI: 10.1093/jnen/nlw065] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
The mechanisms leading to amyloid-β (Aβ) accumulation in sporadic Alzheimer disease (AD) are unknown but both increased production or impaired clearance likely contribute to aggregation. To understand the potential roles of the extracellular matrix proteoglycan Testican-1 in the pathophysiology of AD, we used samples from AD patients and controls and an in vitro approach. Protein expression analysis showed increased levels of Testican-1 in frontal and temporal cortex of AD patients; histological analysis showed that Testican-1 accumulates and co-aggregates with Aβ plaques in the frontal, temporal and entorhinal cortices of AD patients. Proteomic analysis identified 10 fragments of Testican-1 in cerebrospinal fluid (CSF) from AD patients. HEK293T cells expressing human wild type or mutant Aβ precursor protein (APP) were transfected with Testican-1. The co-expression of both proteins modified the sorting of Testican-1 into the endocytic pathway leading to its transient accumulation in Golgi, which seemed to affect APP processing, as indicated by reduced Aβ40 and Aβ42 levels in APP mutant cells. In conclusion, patient data reflect a clearance impairment that may favor Aβ accumulation in AD brains and our in vitro model supports the notion that the interaction between APP and Testican-1 may be a key step in the production and aggregation of Aβ species.
Collapse
Affiliation(s)
- Alvaro Barrera-Ocampo
- From the Institute of Neuropathology (AB-O, JM, BP, MG, DS-F), Department of Psychiatry and Psychotherapy (SA, HJ), University Medical Center Hamburg-Eppendorf, Hamburg, Germany, Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany (UH), Institute of Neuropathology, Bellvitge University Hospital, CIBERNED, Hospitalet de Llobregat, University of Barcelona, Spain (IF), Mosaiques Diagnostics and Therapeutics AG, Hannover, Germany (PZ), and Department of Pharmaceutical Sciences, Natura Research Group, Faculty of Natural Sciences, ICESI University, Cali, Colombia (AB-O)
| | - Sönke Arlt
- From the Institute of Neuropathology (AB-O, JM, BP, MG, DS-F), Department of Psychiatry and Psychotherapy (SA, HJ), University Medical Center Hamburg-Eppendorf, Hamburg, Germany, Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany (UH), Institute of Neuropathology, Bellvitge University Hospital, CIBERNED, Hospitalet de Llobregat, University of Barcelona, Spain (IF), Mosaiques Diagnostics and Therapeutics AG, Hannover, Germany (PZ), and Department of Pharmaceutical Sciences, Natura Research Group, Faculty of Natural Sciences, ICESI University, Cali, Colombia (AB-O)
| | - Jakob Matschke
- From the Institute of Neuropathology (AB-O, JM, BP, MG, DS-F), Department of Psychiatry and Psychotherapy (SA, HJ), University Medical Center Hamburg-Eppendorf, Hamburg, Germany, Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany (UH), Institute of Neuropathology, Bellvitge University Hospital, CIBERNED, Hospitalet de Llobregat, University of Barcelona, Spain (IF), Mosaiques Diagnostics and Therapeutics AG, Hannover, Germany (PZ), and Department of Pharmaceutical Sciences, Natura Research Group, Faculty of Natural Sciences, ICESI University, Cali, Colombia (AB-O)
| | - Ursula Hartmann
- From the Institute of Neuropathology (AB-O, JM, BP, MG, DS-F), Department of Psychiatry and Psychotherapy (SA, HJ), University Medical Center Hamburg-Eppendorf, Hamburg, Germany, Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany (UH), Institute of Neuropathology, Bellvitge University Hospital, CIBERNED, Hospitalet de Llobregat, University of Barcelona, Spain (IF), Mosaiques Diagnostics and Therapeutics AG, Hannover, Germany (PZ), and Department of Pharmaceutical Sciences, Natura Research Group, Faculty of Natural Sciences, ICESI University, Cali, Colombia (AB-O)
| | - Berta Puig
- From the Institute of Neuropathology (AB-O, JM, BP, MG, DS-F), Department of Psychiatry and Psychotherapy (SA, HJ), University Medical Center Hamburg-Eppendorf, Hamburg, Germany, Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany (UH), Institute of Neuropathology, Bellvitge University Hospital, CIBERNED, Hospitalet de Llobregat, University of Barcelona, Spain (IF), Mosaiques Diagnostics and Therapeutics AG, Hannover, Germany (PZ), and Department of Pharmaceutical Sciences, Natura Research Group, Faculty of Natural Sciences, ICESI University, Cali, Colombia (AB-O)
| | - Isidre Ferrer
- From the Institute of Neuropathology (AB-O, JM, BP, MG, DS-F), Department of Psychiatry and Psychotherapy (SA, HJ), University Medical Center Hamburg-Eppendorf, Hamburg, Germany, Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany (UH), Institute of Neuropathology, Bellvitge University Hospital, CIBERNED, Hospitalet de Llobregat, University of Barcelona, Spain (IF), Mosaiques Diagnostics and Therapeutics AG, Hannover, Germany (PZ), and Department of Pharmaceutical Sciences, Natura Research Group, Faculty of Natural Sciences, ICESI University, Cali, Colombia (AB-O)
| | - Petra Zürbig
- From the Institute of Neuropathology (AB-O, JM, BP, MG, DS-F), Department of Psychiatry and Psychotherapy (SA, HJ), University Medical Center Hamburg-Eppendorf, Hamburg, Germany, Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany (UH), Institute of Neuropathology, Bellvitge University Hospital, CIBERNED, Hospitalet de Llobregat, University of Barcelona, Spain (IF), Mosaiques Diagnostics and Therapeutics AG, Hannover, Germany (PZ), and Department of Pharmaceutical Sciences, Natura Research Group, Faculty of Natural Sciences, ICESI University, Cali, Colombia (AB-O)
| | - Markus Glatzel
- From the Institute of Neuropathology (AB-O, JM, BP, MG, DS-F), Department of Psychiatry and Psychotherapy (SA, HJ), University Medical Center Hamburg-Eppendorf, Hamburg, Germany, Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany (UH), Institute of Neuropathology, Bellvitge University Hospital, CIBERNED, Hospitalet de Llobregat, University of Barcelona, Spain (IF), Mosaiques Diagnostics and Therapeutics AG, Hannover, Germany (PZ), and Department of Pharmaceutical Sciences, Natura Research Group, Faculty of Natural Sciences, ICESI University, Cali, Colombia (AB-O)
| | - Diego Sepulveda-Falla
- From the Institute of Neuropathology (AB-O, JM, BP, MG, DS-F), Department of Psychiatry and Psychotherapy (SA, HJ), University Medical Center Hamburg-Eppendorf, Hamburg, Germany, Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany (UH), Institute of Neuropathology, Bellvitge University Hospital, CIBERNED, Hospitalet de Llobregat, University of Barcelona, Spain (IF), Mosaiques Diagnostics and Therapeutics AG, Hannover, Germany (PZ), and Department of Pharmaceutical Sciences, Natura Research Group, Faculty of Natural Sciences, ICESI University, Cali, Colombia (AB-O)
| | - Holger Jahn
- From the Institute of Neuropathology (AB-O, JM, BP, MG, DS-F), Department of Psychiatry and Psychotherapy (SA, HJ), University Medical Center Hamburg-Eppendorf, Hamburg, Germany, Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany (UH), Institute of Neuropathology, Bellvitge University Hospital, CIBERNED, Hospitalet de Llobregat, University of Barcelona, Spain (IF), Mosaiques Diagnostics and Therapeutics AG, Hannover, Germany (PZ), and Department of Pharmaceutical Sciences, Natura Research Group, Faculty of Natural Sciences, ICESI University, Cali, Colombia (AB-O)
| |
Collapse
|
42
|
Hadavi D, Poot AA. Biomaterials for the Treatment of Alzheimer's Disease. Front Bioeng Biotechnol 2016; 4:49. [PMID: 27379232 PMCID: PMC4909781 DOI: 10.3389/fbioe.2016.00049] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 05/27/2016] [Indexed: 12/26/2022] Open
Abstract
Alzheimer’s disease (AD) as a progressive and fatal neurodegenerative disease represents a huge unmet need for treatment. The low efficacy of current treatment methods is not only due to low drug potency but also due to the presence of various obstacles in the delivery routes. One of the main barriers is the blood–brain barrier. The increasing prevalence of AD and the low efficacy of current therapies have increased the amount of research on unraveling of disease pathways and development of treatment strategies. One of the interesting areas for the latter subject is biomaterials and their applications. This interest originates from the fact that biomaterials are very useful for the delivery of therapeutic agents, such as drugs, proteins, and/or cells, in order to treat diseases and regenerate tissues. Recently, manufacturing of nano-sized delivery systems has increased the efficacy and delivery potential of biomaterials. In this article, we review the latest developments with regard to the use of biomaterials for the treatment of AD, including nanoparticles and liposomes for delivery of therapeutic compounds and scaffolds for cell delivery strategies.
Collapse
Affiliation(s)
- Darya Hadavi
- Department of Biomaterials Science and Technology, Institute for Biomedical Technology and Technical Medicine (MIRA), University of Twente , Enschede , Netherlands
| | - André A Poot
- Department of Biomaterials Science and Technology, Institute for Biomedical Technology and Technical Medicine (MIRA), University of Twente , Enschede , Netherlands
| |
Collapse
|
43
|
Deposition of BACE-1 Protein in the Brains of APP/PS1 Double Transgenic Mice. BIOMED RESEARCH INTERNATIONAL 2016; 2016:8380618. [PMID: 27294139 PMCID: PMC4887654 DOI: 10.1155/2016/8380618] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 04/13/2016] [Accepted: 04/17/2016] [Indexed: 11/30/2022]
Abstract
The main causes of Alzheimer's disease remain elusive. Previous data have implicated the BACE-1 protein as a central player in the pathogenesis of Alzheimer's disease. However, many inhibitors of BACE-1 have failed during preclinical and clinical trials for AD treatment. Therefore, uncovering the exact role of BACE-1 in AD may have significant impact on the future development of therapeutic agents. Three- and six-month-old female APP/PS1 double transgenic mice were used to study abnormal accumulation of BACE-1 protein in brains of mice here. Immunofluorescence, immunohistochemistry, and western blot were performed to measure the distributing pattern and expression level of BACE-1. We found obvious BACE-1 protein accumulation in 3-month-old APP/PS1 mice, which had increased by the time of 6 months. Coimmunostaining results showed BACE-1 surrounded amyloid plaques in brain sections. The abnormal protein expression might not be attributable to the upregulation of BACE-1 protein, as no significant difference of protein expression was observed between wild-type and APP/PS1 mice. With antibodies against BACE-1 and CD31, we found a high immunoreactive density of BACE-1 protein on the outer layer of brain blood vessels. The aberrant distribution of BACE-1 in APP/PS1 mice suggests BACE-1 may be involved in the microvascular abnormality of AD.
Collapse
|
44
|
Post-translational regulation of the β-secretase BACE1. Brain Res Bull 2016; 126:170-177. [PMID: 27086128 DOI: 10.1016/j.brainresbull.2016.04.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 04/08/2016] [Accepted: 04/12/2016] [Indexed: 11/21/2022]
Abstract
β-Secretase, widely known as β-site APP cleaving enzyme 1 (BACE1), is a membrane-associated protease that cleaves amyloid precursor protein (APP) to generate amyloid β-protein (Aβ). As this cleavage is a pathologically relevant event in Alzheimer's disease, BACE1 is considered a viable therapeutic target. BACE1 can be regulated at the transcriptional, post-transcriptional, translational, and post-translational levels. Elucidation of the regulatory pathways of BACE1 is critical, not only for understanding the pathological mechanisms of AD but also developing effective therapeutic strategies to inhibit activity of the protease. This mini-review focuses on the post-translational regulation of BACE1, as modulation at this level is closely associated with both physiological and pathological conditions. Current knowledge on the mechanisms underlying such BACE1 regulation and their implications for therapy are discussed.
Collapse
|
45
|
Plummer S, Van den Heuvel C, Thornton E, Corrigan F, Cappai R. The Neuroprotective Properties of the Amyloid Precursor Protein Following Traumatic Brain Injury. Aging Dis 2016; 7:163-79. [PMID: 27114849 PMCID: PMC4809608 DOI: 10.14336/ad.2015.0907] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Accepted: 09/07/2015] [Indexed: 01/16/2023] Open
Abstract
Despite the significant health and economic burden that traumatic brain injury (TBI) places on society, the development of successful therapeutic agents have to date not translated into efficacious therapies in human clinical trials. Injury to the brain is ongoing after TBI, through a complex cascade of primary and secondary injury events, providing a valuable window of opportunity to help limit and prevent some of the severe consequences with a timely treatment. Of note, it has been suggested that novel treatments for TBI should be multifactorial in nature, mimicking the body's own endogenous repair response. Whilst research has historically focused on the role of the amyloid precursor protein (APP) in the pathogenesis of Alzheimer's disease, recent advances in trauma research have demonstrated that APP offers considerable neuroprotective properties following TBI, suggesting that APP is an ideal therapeutic candidate. Its acute upregulation following TBI has been shown to serve a beneficial role following trauma and has lead to significant advances in understanding the neuroprotective and neurotrophic functions of APP and its metabolites. Research has focused predominantly on the APP derivative sAPPα, which has consistently demonstrated neuroprotective and neurotrophic functions both in vitro and in vivo following various traumatic insults. Its neuroprotective activity has been narrowed down to a 15 amino acid sequence, and this region is linked to both heparan binding and growth-factor-like properties. It has been proposed that APP binds to heparan sulfate proteoglycans to exert its neuroprotective action. APP presents us with a novel therapeutic compound that could overcome many of the challenges that have stalled development of efficacious TBI treatments previously.
Collapse
Affiliation(s)
- Stephanie Plummer
- Adelaide Centre for Neuroscience Research, the University of Adelaide, South Australia, Australia
| | - Corinna Van den Heuvel
- Adelaide Centre for Neuroscience Research, the University of Adelaide, South Australia, Australia
| | - Emma Thornton
- Adelaide Centre for Neuroscience Research, the University of Adelaide, South Australia, Australia
| | - Frances Corrigan
- Adelaide Centre for Neuroscience Research, the University of Adelaide, South Australia, Australia
| | - Roberto Cappai
- Department of Pathology, the University of Melbourne, Victoria, Australia
| |
Collapse
|
46
|
Stevenson JW, Conaty EA, Walsh RB, Poidomani PJ, Samoriski CM, Scollins BJ, DeGiorgis JA. The Amyloid Precursor Protein of Alzheimer's Disease Clusters at the Organelle/Microtubule Interface on Organelles that Bind Microtubules in an ATP Dependent Manner. PLoS One 2016; 11:e0147808. [PMID: 26814888 PMCID: PMC4729464 DOI: 10.1371/journal.pone.0147808] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Accepted: 01/08/2016] [Indexed: 11/18/2022] Open
Abstract
The amyloid precursor protein (APP) is a causal agent in the pathogenesis of Alzheimer's disease and is a transmembrane protein that associates with membrane-limited organelles. APP has been shown to co-purify through immunoprecipitation with a kinesin light chain suggesting that APP may act as a trailer hitch linking kinesin to its intercellular cargo, however this hypothesis has been challenged. Previously, we identified an mRNA transcript that encodes a squid homolog of human APP770. The human and squid isoforms share 60% sequence identity and 76% sequence similarity within the cytoplasmic domain and share 15 of the final 19 amino acids at the C-terminus establishing this highly conserved domain as a functionally import segment of the APP molecule. Here, we study the distribution of squid APP in extruded axoplasm as well as in a well-characterized reconstituted organelle/microtubule preparation from the squid giant axon in which organelles bind microtubules and move towards the microtubule plus-ends. We find that APP associates with microtubules by confocal microscopy and co-purifies with KI-washed axoplasmic organelles by sucrose density gradient fractionation. By electron microscopy, APP clusters at a single focal point on the surfaces of organelles and localizes to the organelle/microtubule interface. In addition, the association of APP-organelles with microtubules is an ATP dependent process suggesting that the APP-organelles contain a microtubule-based motor protein. Although a direct kinesin/APP association remains controversial, the distribution of APP at the organelle/microtubule interface strongly suggests that APP-organelles have an orientation and that APP like the Alzheimer's protein tau has a microtubule-based function.
Collapse
Affiliation(s)
- James W. Stevenson
- Biology Department, Providence College, Providence, Rhode Island, United States of America
- Bell Center, Marine Biological Laboratory, Woods Hole, Massachusetts, United States of America
| | - Eliza A. Conaty
- Biology Department, Providence College, Providence, Rhode Island, United States of America
- Bell Center, Marine Biological Laboratory, Woods Hole, Massachusetts, United States of America
| | - Rylie B. Walsh
- Biology Department, Providence College, Providence, Rhode Island, United States of America
- Bell Center, Marine Biological Laboratory, Woods Hole, Massachusetts, United States of America
| | - Paul J. Poidomani
- Biology Department, Providence College, Providence, Rhode Island, United States of America
- Bell Center, Marine Biological Laboratory, Woods Hole, Massachusetts, United States of America
| | - Colin M. Samoriski
- Biology Department, Providence College, Providence, Rhode Island, United States of America
- Bell Center, Marine Biological Laboratory, Woods Hole, Massachusetts, United States of America
| | - Brianne J. Scollins
- Biology Department, Providence College, Providence, Rhode Island, United States of America
- Bell Center, Marine Biological Laboratory, Woods Hole, Massachusetts, United States of America
| | - Joseph A. DeGiorgis
- Biology Department, Providence College, Providence, Rhode Island, United States of America
- Bell Center, Marine Biological Laboratory, Woods Hole, Massachusetts, United States of America
| |
Collapse
|
47
|
Amyloid-Beta Induced Changes in Vesicular Transport of BDNF in Hippocampal Neurons. Neural Plast 2016; 2016:4145708. [PMID: 26881108 PMCID: PMC4736975 DOI: 10.1155/2016/4145708] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 11/26/2015] [Accepted: 11/29/2015] [Indexed: 12/15/2022] Open
Abstract
The neurotrophin brain derived neurotrophic factor (BDNF) is an important growth factor in the CNS. Deficits in transport of this secretory protein could underlie neurodegenerative diseases. Investigation of disease-related changes in BDNF transport might provide insights into the cellular mechanism underlying, for example, Alzheimer's disease (AD). To analyze the role of BDNF transport in AD, live cell imaging of fluorescently labeled BDNF was performed in hippocampal neurons of different AD model systems. BDNF and APP colocalized with low incidence in vesicular structures. Anterograde as well as retrograde transport of BDNF vesicles was reduced and these effects were mediated by factors released from hippocampal neurons into the extracellular medium. Transport of BDNF was altered at a very early time point after onset of human APP expression or after acute amyloid-beta(1-42) treatment, while the activity-dependent release of BDNF remained unaffected. Taken together, extracellular cleavage products of APP induced rapid changes in anterograde and retrograde transport of BDNF-containing vesicles while release of BDNF was unaffected by transgenic expression of mutated APP. These early transport deficits might lead to permanently impaired brain functions in the adult brain.
Collapse
|
48
|
Killing Me Softly: Connotations to Unfolded Protein Response and Oxidative Stress in Alzheimer's Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:1805304. [PMID: 26881014 PMCID: PMC4736771 DOI: 10.1155/2016/1805304] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 11/28/2015] [Accepted: 12/07/2015] [Indexed: 11/18/2022]
Abstract
This review is focused on the possible causes of mitochondrial dysfunction in AD, underlying molecular mechanisms of this malfunction, possible causes and known consequences of APP, Aβ, and hyperphosphorylated tau presence in mitochondria, and the contribution of altered lipid metabolism (nonsterol isoprenoids) to pathological processes leading to increased formation and accumulation of the aforementioned hallmarks of AD. Abnormal protein folding and unfolded protein response seem to be the outcomes of impaired glycosylation due to metabolic disturbances in geranylgeraniol intermediary metabolism. The origin and consecutive fate of APP, Aβ, and tau are emphasized on intracellular trafficking apparently influenced by inaccurate posttranslational modifications. We hypothesize that incorrect intracellular processing of APP determines protein translocation to mitochondria in AD. Similarly, without obvious reasons, the passage of Aβ and tau to mitochondria is observed. APP targeted to mitochondria blocks the activity of protein translocase complex resulting in poor import of proteins central to oxidative phosphorylation. Besides, APP, Aβ, and neurofibrillary tangles of tau directly or indirectly impair mitochondrial biochemistry and bioenergetics, with concomitant generation of oxidative/nitrosative stress. Limited protective mechanisms are inadequate to prevent the free radical-mediated lesions. Finally, neuronal loss is observed in AD-affected brains typically by pathologic apoptosis.
Collapse
|
49
|
The Association of Amyloid-β Protein Precursor With α- and β-Secretases in Mouse Cerebral Cortex Synapses Is Altered in Early Alzheimer’s Disease. Mol Neurobiol 2015; 53:5710-21. [DOI: 10.1007/s12035-015-9491-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 10/14/2015] [Indexed: 10/22/2022]
|
50
|
Hermey G, Schmidt N, Bluhm B, Mensching D, Ostermann K, Rupp C, Kuhl D, Kins S. SorCS1 variants and amyloid precursor protein (APP) are co-transported in neurons but only SorCS1c modulates anterograde APP transport. J Neurochem 2015; 135:60-75. [PMID: 26119586 DOI: 10.1111/jnc.13221] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Revised: 05/06/2015] [Accepted: 06/22/2015] [Indexed: 12/13/2022]
Abstract
Processing of amyloid precursor protein (APP) into amyloid-β peptide (Aβ) is crucial for the development of Alzheimer's disease (AD). Because this processing is highly dependent on its intracellular itinerary, altered subcellular targeting of APP is thought to directly affect the degree to which Aβ is generated. The sorting receptor SorCS1 has been genetically linked to AD, but the underlying molecular mechanisms are poorly understood. We analyze two SorCS1 variants; one, SorCS1c, conveys internalization of surface-bound ligands whereas the other, SorCS1b, does not. In agreement with previous studies, we demonstrate co-immunoprecipitation and co-localization of both SorCS1 variants with APP. Our results suggest that SorCS1c and APP are internalized independently, although they mostly share a common post-endocytic pathway. We introduce functional Venus-tagged constructs to study SorCS1b and SorCS1c in living cells. Both variants are transported by fast anterograde axonal transport machinery and about 30% of anterograde APP-positive transport vesicles contain SorCS1. Co-expression of SorCS1b caused no change of APP transport kinetics, but SorCS1c reduced the anterograde transport rate of APP and increased the number of APP-positive stationary vesicles. These data suggest that SorCS1 and APP share trafficking pathways and that SorCS1c can retain APP from insertion into anterograde transport vesicles. Altered APP trafficking is thought to modulate its processing. SorCS1 has been suggested to function in APP trafficking. We analyzed if the two SorCS1 variants, SorCS1b and SorCS1c, tie APP to the cell surface or modify its internalization and intracellular targeting. We observed co-localization and vesicular co-transport of APP and SorCS1, but independent internalization and sorting through a common post-endocytic pathway. Co-expression of one variant, SorCS1c, reduced anterograde APP transport. These data demonstrate that SorCS1 and APP share trafficking pathways and that SorCS1c can retain APP from insertion into anterograde transport vesicles.
Collapse
Affiliation(s)
- Guido Hermey
- Institute for Molecular and Cellular Cognition, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nadine Schmidt
- Division of Human Biology and Human Genetics, University of Kaiserslautern, Kaiserslautern, Germany
| | - Björn Bluhm
- Institute for Molecular and Cellular Cognition, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Daniel Mensching
- Institute for Molecular and Cellular Cognition, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kristina Ostermann
- Division of Human Biology and Human Genetics, University of Kaiserslautern, Kaiserslautern, Germany
| | - Carsten Rupp
- Division of Human Biology and Human Genetics, University of Kaiserslautern, Kaiserslautern, Germany
| | - Dietmar Kuhl
- Institute for Molecular and Cellular Cognition, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Kins
- Division of Human Biology and Human Genetics, University of Kaiserslautern, Kaiserslautern, Germany
| |
Collapse
|