1
|
Wang M, He X, Li J, Han D, You P, Yu H, Wang L, Su B. GDI2 deletion alleviates neurodegeneration and memory loss in the 5xFAD mice model of Alzheimer's disease. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167093. [PMID: 38382624 DOI: 10.1016/j.bbadis.2024.167093] [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: 11/15/2023] [Revised: 02/15/2024] [Accepted: 02/16/2024] [Indexed: 02/23/2024]
Abstract
Accumulation of insoluble deposits of amyloid β-peptide (Aβ), derived from amyloid precursor protein (APP) processing, represents one of the major pathological hallmarks of Alzheimer's disease (AD). Perturbations in APP transport and hydrolysis could lead to increased Aβ production. However, the precise mechanisms underlying APP transport remain elusive. The GDP dissociation inhibitor2 (GDI2), a crucial regulator of Rab GTPase activity and intracellular vesicle and membrane trafficking, was investigated for its impact on AD pathogenesis through neuron-specific knockout of GDI2 in 5xFAD mice. Notably, deficiency of GDI2 significantly ameliorated cognitive impairment, prevented neuronal loss in the subiculum and cortical layer V, reduced senile plaques as well as astrocyte activation in 5xFAD mice. Conversely, increased activated microglia and phagocytosis were observed in GDI2 ko mice. Further investigation revealed that GDI2 knockout led to more APP co-localized with the ER rather than the Golgi apparatus and endosomes in SH-SY5Y cells, resulting in decreased Aβ production. Collectively, these findings suggest that GDI2 may regulate Aβ production by modulating APP intracellular transport and localization dynamics. In summary, our study identifies GDI2 as a pivotal regulator governing APP transport and process implicated in AD pathology; thus highlighting its potential as an attractive pharmacological target for future drug development against AD.
Collapse
Affiliation(s)
- Meitian Wang
- Department of Cell Biology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Xiuqing He
- Department of Cell Biology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Jie Li
- Department of Cell Biology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Daobin Han
- Department of Cell Biology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Pan You
- Department of Cell Biology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Hui Yu
- Department of Cell Biology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Luwen Wang
- Advanced Medical Research Institute, Shandong University, Jinan, China
| | - Bo Su
- Department of Cell Biology, School of Basic Medical Sciences, Shandong University, Jinan, China.
| |
Collapse
|
2
|
Nik Akhtar S, Bunner WP, Brennan E, Lu Q, Szatmari EM. Crosstalk between the Rho and Rab family of small GTPases in neurodegenerative disorders. Front Cell Neurosci 2023; 17:1084769. [PMID: 36779014 PMCID: PMC9911442 DOI: 10.3389/fncel.2023.1084769] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 01/06/2023] [Indexed: 01/28/2023] Open
Abstract
Neurodegeneration is associated with defects in cytoskeletal dynamics and dysfunctions of the vesicular trafficking and sorting systems. In the last few decades, studies have demonstrated that the key regulators of cytoskeletal dynamics are proteins from the Rho family GTPases, meanwhile, the central hub for vesicle sorting and transport between target membranes is the Rab family of GTPases. In this regard, the role of Rho and Rab GTPases in the induction and maintenance of distinct functional and morphological neuronal domains (such as dendrites and axons) has been extensively studied. Several members belonging to these two families of proteins have been associated with many neurodegenerative disorders ranging from dementia to motor neuron degeneration. In this analysis, we attempt to present a brief review of the potential crosstalk between the Rab and Rho family members in neurodegenerative pathologies such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington disease, and amyotrophic lateral sclerosis (ALS).
Collapse
Affiliation(s)
- Shayan Nik Akhtar
- The Harriet and John Wooten Laboratory for Alzheimer’s and Neurodegenerative Diseases Research, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| | - Wyatt P. Bunner
- Laboratory of Neuroscience, Department of Physical Therapy, College of Allied Health Sciences, East Carolina University, Greenville, NC, United States
| | - Elizabeth Brennan
- Laboratory of Neuroscience, Department of Physical Therapy, College of Allied Health Sciences, East Carolina University, Greenville, NC, United States
| | - Qun Lu
- The Harriet and John Wooten Laboratory for Alzheimer’s and Neurodegenerative Diseases Research, Brody School of Medicine, East Carolina University, Greenville, NC, United States,*Correspondence: Erzsebet M. Szatmari Qun Lu
| | - Erzsebet M. Szatmari
- Laboratory of Neuroscience, Department of Physical Therapy, College of Allied Health Sciences, East Carolina University, Greenville, NC, United States,*Correspondence: Erzsebet M. Szatmari Qun Lu
| |
Collapse
|
3
|
Hatstat AK, Pupi MD, Reinhart MC, McCafferty DG. Small Molecule Improvement of Trafficking Defects in Models of Neurodegeneration. ACS Chem Neurosci 2021; 12:3972-3984. [PMID: 34652126 DOI: 10.1021/acschemneuro.1c00524] [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: 11/30/2022] Open
Abstract
Disrupted cellular trafficking and transport processes are hallmarks of many neurodegenerative disorders (NDs). Recently, efforts have been made toward developing and implementing experimental platforms to identify small molecules that may help restore normative trafficking functions. There have been a number of successes in targeting endomembrane trafficking with the identification of compounds that restore cell viability through rescue of protein transport and trafficking. Here, we describe some of the experimental platforms implemented for small molecule screening efforts for rescue of trafficking defects in neurodegeneration. A survey of phenotypically active small molecules identified to date is provided, including a summary of medicinal chemistry efforts and insights into putative targets and mechanisms of action. In particular, emphasis is put on ligands that demonstrate activity in more than one model of neurodegeneration as retention of phenotypic activity across ND models suggests conservation of biological targets across NDs.
Collapse
Affiliation(s)
- A. Katherine Hatstat
- Department of Chemistry, Duke University, Durham, North Carolina 27708, United States
| | - Michael D. Pupi
- Department of Chemistry, Duke University, Durham, North Carolina 27708, United States
| | - Michaela C. Reinhart
- Department of Chemistry, Duke University, Durham, North Carolina 27708, United States
| | - Dewey G. McCafferty
- Department of Chemistry, Duke University, Durham, North Carolina 27708, United States
| |
Collapse
|
4
|
Deaton CA, Johnson GVW. Presenilin 1 Regulates Membrane Homeostatic Pathways that are Dysregulated in Alzheimer's Disease. J Alzheimers Dis 2021; 77:961-977. [PMID: 32804090 DOI: 10.3233/jad-200598] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Mutations in the PSEN1 gene, encoding presenilin 1 (PS1), are the most common cause of familial Alzheimer's disease (fAD). Since the first mutations in the PSEN1 gene were discovered more than 25 years ago, many postulated functions of PS1 have been investigated. The majority of earlier studies focused on its role as the catalytic component of the γ-secretase complex, which in concert with β site amyloid precursor protein cleaving enzyme 1 (BACE1), mediates the formation of Aβ from amyloid-β protein precursor (AβPP). Though mutant PS1 was originally considered to cause AD by promoting Aβ pathology through its protease function, it is now becoming clear that PS1 is a multifunctional protein involved in regulating membrane dynamics and protein trafficking. Therefore, through loss of these abilities, mutant PS1 has the potential to impair numerous cellular functions such as calcium flux, organization of proteins in different compartments, and protein turnover via vacuolar metabolism. Impaired calcium signaling, vacuolar dysfunction, mitochondrial dysfunction, and increased ER stress, among other related membrane-dependent disturbances, have been considered critical to the development and progression of AD. Given that PS1 plays a key regulatory role in all these processes, this review will describe the role of PS1 in different cellular compartments and provide an integrated view of how PS1 dysregulation (due to mutations or other causes) could result in impairment of various cellular processes and result in a "multi-hit", integrated pathological outcome that could contribute to the etiology of AD.
Collapse
Affiliation(s)
- Carol A Deaton
- Cell Biology of Disease Program and the Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Gail V W Johnson
- Cell Biology of Disease Program and the Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
5
|
Abstract
Defects in membrane trafficking are hallmarks of neurodegeneration. Rab GTPases are key regulators of membrane trafficking. Alterations of Rab GTPases, or the membrane compartments they regulate, are associated with virtually all neuronal activities in health and disease. The observation that many Rab GTPases are associated with neurodegeneration has proven a challenge in the quest for cause and effect. Neurodegeneration can be a direct consequence of a defect in membrane trafficking. Alternatively, changes in membrane trafficking may be secondary consequences or cellular responses. The secondary consequences and cellular responses, in turn, may protect, represent inconsequential correlates or function as drivers of pathology. Here, we attempt to disentangle the different roles of membrane trafficking in neurodegeneration by focusing on selected associations with Alzheimer’s disease, Parkinson’s disease, Huntington’s disease and selected neuropathies. We provide an overview of current knowledge on Rab GTPase functions in neurons and review the associations of Rab GTPases with neurodegeneration with respect to the following classifications: primary cause, secondary cause driving pathology or secondary correlate. This analysis is devised to aid the interpretation of frequently observed membrane trafficking defects in neurodegeneration and facilitate the identification of true causes of pathology.
Collapse
|
6
|
Rab GTPases: Switching to Human Diseases. Cells 2019; 8:cells8080909. [PMID: 31426400 PMCID: PMC6721686 DOI: 10.3390/cells8080909] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 08/13/2019] [Accepted: 08/14/2019] [Indexed: 02/07/2023] Open
Abstract
Rab proteins compose the largest family of small GTPases and control the different steps of intracellular membrane traffic. More recently, they have been shown to also regulate cell signaling, division, survival, and migration. The regulation of these processes generally occurs through recruitment of effectors and regulatory proteins, which control the association of Rab proteins to membranes and their activation state. Alterations in Rab proteins and their effectors are associated with multiple human diseases, including neurodegeneration, cancer, and infections. This review provides an overview of how the dysregulation of Rab-mediated functions and membrane trafficking contributes to these disorders. Understanding the altered dynamics of Rabs and intracellular transport defects might thus shed new light on potential therapeutic strategies.
Collapse
|
7
|
Qu L, Pan C, He SM, Lang B, Gao GD, Wang XL, Wang Y. The Ras Superfamily of Small GTPases in Non-neoplastic Cerebral Diseases. Front Mol Neurosci 2019; 12:121. [PMID: 31213978 PMCID: PMC6555388 DOI: 10.3389/fnmol.2019.00121] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 04/25/2019] [Indexed: 12/22/2022] Open
Abstract
The small GTPases from the Ras superfamily play crucial roles in basic cellular processes during practically the entire process of neurodevelopment, including neurogenesis, differentiation, gene expression, membrane and protein traffic, vesicular trafficking, and synaptic plasticity. Small GTPases are key signal transducing enzymes that link extracellular cues to the neuronal responses required for the construction of neuronal networks, as well as for synaptic function and plasticity. Different subfamilies of small GTPases have been linked to a number of non-neoplastic cerebral diseases such as Alzheimer’s disease (AD), Parkinson’s disease (PD), intellectual disability, epilepsy, drug addiction, Huntington’s disease (HD), amyotrophic lateral sclerosis (ALS) and a large number of idiopathic cerebral diseases. Here, we attempted to make a clearer illustration of the relationship between Ras superfamily GTPases and non-neoplastic cerebral diseases, as well as their roles in the neural system. In future studies, potential treatments for non-neoplastic cerebral diseases which are based on small GTPase related signaling pathways should be explored further. In this paper, we review all the available literature in support of this possibility.
Collapse
Affiliation(s)
- Liang Qu
- Department of Neurosurgery, Tangdu Hospital, Air Force Military Medical University, Xi'an, China
| | - Chao Pan
- Beijing Institute of Biotechnology, Beijing, China
| | - Shi-Ming He
- Department of Neurosurgery, Tangdu Hospital, Air Force Military Medical University, Xi'an, China.,Department of Neurosurgery, Xi'an International Medical Center, Xi'an, China
| | - Bing Lang
- The School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom.,Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Guo-Dong Gao
- Department of Neurosurgery, Tangdu Hospital, Air Force Military Medical University, Xi'an, China
| | - Xue-Lian Wang
- Department of Neurosurgery, Tangdu Hospital, Air Force Military Medical University, Xi'an, China
| | - Yuan Wang
- Department of Neurosurgery, Tangdu Hospital, Air Force Military Medical University, Xi'an, China
| |
Collapse
|
8
|
Zhang X, Huang TY, Yancey J, Luo H, Zhang YW. Role of Rab GTPases in Alzheimer's Disease. ACS Chem Neurosci 2019; 10:828-838. [PMID: 30261139 DOI: 10.1021/acschemneuro.8b00387] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) comprises two major pathological hallmarks: extraneuronal deposition of β-amyloid (Aβ) peptides ("senile plaques") and intraneuronal aggregation of the microtubule-associated protein tau ("neurofibrillary tangles"). Aβ is derived from sequential cleavage of the β-amyloid precursor protein by β- and γ-secretases, while aggregated tau is hyperphosphorylated in AD. Mounting evidence suggests that dysregulated trafficking of these AD-related proteins contributes to AD pathogenesis. Rab proteins are small GTPases that function as master regulators of vesicular transport and membrane trafficking. Multiple Rab GTPases have been implicated in AD-related protein trafficking, and their expression has been observed to be altered in postmortem AD brain. Here we review current implicated roles of Rab GTPase dysregulation in AD pathogenesis. Further elucidation of the pathophysiological role of Rab GTPases will likely reveal novel targets for AD therapeutics.
Collapse
Affiliation(s)
- Xian Zhang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Medical College of Xiamen University, Xiamen, Fujian 361102, China
| | - Timothy Y. Huang
- Neuroscience Initiative, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, California 92037, United States
| | - Joel Yancey
- Neuroscience Initiative, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, California 92037, United States
| | - Hong Luo
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Medical College of Xiamen University, Xiamen, Fujian 361102, China
| | - Yun-wu Zhang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Medical College of Xiamen University, Xiamen, Fujian 361102, China
| |
Collapse
|
9
|
Kametani F, Hasegawa M. Reconsideration of Amyloid Hypothesis and Tau Hypothesis in Alzheimer's Disease. Front Neurosci 2018; 12:25. [PMID: 29440986 PMCID: PMC5797629 DOI: 10.3389/fnins.2018.00025] [Citation(s) in RCA: 566] [Impact Index Per Article: 80.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 01/12/2018] [Indexed: 12/20/2022] Open
Abstract
The so-called amyloid hypothesis, that the accumulation and deposition of oligomeric or fibrillar amyloid β (Aβ) peptide is the primary cause of Alzheimer's disease (AD), has been the mainstream concept underlying AD research for over 20 years. However, all attempts to develop Aβ-targeting drugs to treat AD have ended in failure. Here, we review recent findings indicating that the main factor underlying the development and progression of AD is tau, not Aβ, and we describe the deficiencies of the amyloid hypothesis that have supported the emergence of this idea.
Collapse
Affiliation(s)
- Fuyuki Kametani
- Department of Dementia and Higher Brain Function, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Masato Hasegawa
- Department of Dementia and Higher Brain Function, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| |
Collapse
|
10
|
The Role of Presenilin in Protein Trafficking and Degradation—Implications for Metal Homeostasis. J Mol Neurosci 2016; 60:289-297. [DOI: 10.1007/s12031-016-0826-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 08/17/2016] [Indexed: 12/13/2022]
|
11
|
Udayar V, Buggia-Prévot V, Guerreiro RL, Siegel G, Rambabu N, Soohoo AL, Ponnusamy M, Siegenthaler B, Bali J, Simons M, Ries J, Puthenveedu MA, Hardy J, Thinakaran G, Rajendran L. A paired RNAi and RabGAP overexpression screen identifies Rab11 as a regulator of β-amyloid production. Cell Rep 2013; 5:1536-51. [PMID: 24373285 PMCID: PMC4004174 DOI: 10.1016/j.celrep.2013.12.005] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Revised: 11/07/2013] [Accepted: 12/03/2013] [Indexed: 12/16/2022] Open
Abstract
Alzheimer's disease (AD) is characterized by cerebral deposition of β-amyloid (Aβ) peptides, which are generated from amyloid precursor protein (APP) by β- and γ-secretases. APP and the secretases are membrane associated, but whether membrane trafficking controls Aβ levels is unclear. Here, we performed an RNAi screen of all human Rab-GTPases, which regulate membrane trafficking, complemented with a Rab-GTPase-activating protein screen, and present a road map of the membrane-trafficking events regulating Aβ production. We identify Rab11 and Rab3 as key players. Although retromers and retromer-associated proteins control APP recycling, we show that Rab11 controlled β-secretase endosomal recycling to the plasma membrane and thus affected Aβ production. Exome sequencing revealed a significant genetic association of Rab11A with late-onset AD, and network analysis identified Rab11A and Rab11B as components of the late-onset AD risk network, suggesting a causal link between Rab11 and AD. Our results reveal trafficking pathways that regulate Aβ levels and show how systems biology approaches can unravel the molecular complexity underlying AD.
Collapse
Affiliation(s)
- Vinod Udayar
- Systems and Cell Biology of Neurodegeneration, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland; Division of Psychiatry Research, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland; Graduate Program of the Zurich Neuroscience Center, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland; Erasmus Mundus Joint Doctorate Program, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
| | - Virginie Buggia-Prévot
- Departments of Neurobiology, Neurology and Pathology, The University of Chicago, Chicago, IL 60637, USA
| | - Rita L Guerreiro
- Alzheimer's Exome Sequencing Group, University College London, London WC1E 6BT, UK; Reta Lila Weston Research Laboratories, Department of Molecular Neuroscience, UCL Institute of Neurology, London WC1N 3BG, UK
| | - Gabriele Siegel
- Systems and Cell Biology of Neurodegeneration, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland; Division of Psychiatry Research, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
| | - Naresh Rambabu
- Raise.Rural Foundation for Promoting Research Awareness in Student Environment in Rural India, 60045 Chennai, India
| | - Amanda L Soohoo
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Moorthi Ponnusamy
- Raise.Rural Foundation for Promoting Research Awareness in Student Environment in Rural India, 60045 Chennai, India
| | - Barbara Siegenthaler
- Systems and Cell Biology of Neurodegeneration, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland; Division of Psychiatry Research, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
| | - Jitin Bali
- Systems and Cell Biology of Neurodegeneration, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland; Division of Psychiatry Research, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland; Graduate Program of the Zurich Neuroscience Center, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
| | - Mikael Simons
- Department of Neurology, University of Göttingen, 37075 Göttingen, Germany; Max-Planck Institute for Experimental Medicine, 37075 Göttingen, Germany
| | - Jonas Ries
- Cell Biology and Biophysics, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | | | - John Hardy
- Alzheimer's Exome Sequencing Group, University College London, London WC1E 6BT, UK; Reta Lila Weston Research Laboratories, Department of Molecular Neuroscience, UCL Institute of Neurology, London WC1N 3BG, UK
| | - Gopal Thinakaran
- Departments of Neurobiology, Neurology and Pathology, The University of Chicago, Chicago, IL 60637, USA
| | - Lawrence Rajendran
- Systems and Cell Biology of Neurodegeneration, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland; Division of Psychiatry Research, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland; Graduate Program of the Zurich Neuroscience Center, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland; Graduate Program of the Zurich Center for Integrative Human Physiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland.
| |
Collapse
|
12
|
Qi X, Cai J, Ruan Q, Liu L, Boye SL, Chen Z, Hauswirth WW, Ryals RC, Shaw L, Caballero S, Grant MB, Boulton ME. γ-Secretase inhibition of murine choroidal neovascularization is associated with reduction of superoxide and proinflammatory cytokines. Invest Ophthalmol Vis Sci 2012; 53:574-85. [PMID: 22205609 DOI: 10.1167/iovs.11-8728] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
PURPOSE This study aimed to determine whether upregulation of γ-secretase could inhibit laser-induced choroidal neovascularization (CNV) and if this was associated with a reduction in both oxidative stress and proinflammatory cytokines. METHODS γ-Secretase, or its catalytic subunit presenilin 1 (PS1), were upregulated by exposure to either pigment epithelial derived factor (PEDF) or an AAV2 vector containing a PS1 gene driven by a vascular endothelial-cadherin promoter. Retinal endothelial cells were infected with AAV2 or exposed to PEDF in the presence or absence of VEGF and in vitro angiogenesis determined. Mouse eyes either received intravitreal injection of PEDF, DAPT (a γ-secretase inhibitor) or PEDF + DAPT at the time of laser injury, or AAV2 infection 3 weeks before receiving laser burns. Lesion volume was determined 14 days post laser injury. Superoxide generation, antioxidant activity and the production of proinflammatory mediators were assessed. Knockdown of γ-secretase was achieved using siRNA. RESULTS γ-Secretase upregulation and PS1 overexpression suppressed VEGF-induced in vitro angiogenesis and in vivo laser-induced CNV. This was associated with a reduction in the expression of VEGF and angiogenin 1 together with reduced superoxide anion generation and an increase in MnSOD compared with untreated CNV eyes. PS1 overexpression reduced proinflammatory factors and microglial activation in eyes with CNV compared with control. siRNA inhibition of γ-secretase resulted in increased angiogenesis. CONCLUSIONS γ-Secretase, and in particular PS1 alone, are potent regulators of angiogenesis and this is due in part to stabilizing endogenous superoxide generation and reducing proinflammatory cytokine expression during CNV.
Collapse
Affiliation(s)
- Xiaoping Qi
- Departments of Anatomy and Cell Biology, University of Florida, Gainesville, Florida 32610-0235, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Cai J, Chen Z, Ruan Q, Han S, Liu L, Qi X, Boye SL, Hauswirth WW, Grant MB, Boulton ME. γ-Secretase and presenilin mediate cleavage and phosphorylation of vascular endothelial growth factor receptor-1. J Biol Chem 2011; 286:42514-42523. [PMID: 22016384 PMCID: PMC3234916 DOI: 10.1074/jbc.m111.296590] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2011] [Revised: 10/14/2011] [Indexed: 01/18/2023] Open
Abstract
We have reported previously that pigment epithelium-derived factor (PEDF) can, via γ-secretase-mediated events, inhibit VEGF-induced angiogenesis in microvascular endothelial cells by both (a) cleavage and intracellular translocation of a C-terminal fragment of VEGF receptor-1 (VEGFR1) and (b) inhibition of VEGF-induced phosphorylation of VEGFR1. Using site-direct mutagenesis and transfection of wild type and mutated receptors into endothelial cells, we showed that transmembrane cleavage of VEGFR1 occurs at valine 767 and that a switch from valine to alanine at this position prevented cleavage and formation of a VEGFR1 intracellular fragment. Using siRNA to selectively knock down protein-tyrosine phosphatases (PTPs) in endothelial cells, we demonstrated that vascular endothelial PTP is responsible for dephosphorylation of activated VEGFR1. PEDF up-regulation of full-length presenilin 1 (Fl.PS1) facilitated the association of vascular endothelial PTP and VEGFR1. Knockdown of Fl.PS1 prevented dephosphorylation of VEGFR1, whereas up-regulation of Fl.PS1 stimulated VEGFR1 dephosphorylation. Fl.PS1 associated with VEGFR1 within 15 min after PEDF treatment. In conclusion, we determined the PEDF-mediated events responsible for VEGFR1 signaling and identified full-length presenilin as a critical adaptor molecule in the dephosphorylation of VEGFR1. This greater understanding of the regulation of VEGFR1 signaling will help identify novel anti-VEGF therapeutic strategies.
Collapse
Affiliation(s)
- Jun Cai
- Departments of Anatomy and Cell Biology, University of Florida, Gainesville, Florida 32610-0235
| | - Zhijuan Chen
- Departments of Anatomy and Cell Biology, University of Florida, Gainesville, Florida 32610-0235
| | - Qing Ruan
- Departments of Anatomy and Cell Biology, University of Florida, Gainesville, Florida 32610-0235
| | - Song Han
- Department of Surgery, University of Florida, Gainesville, Florida 32610-0235
| | - Li Liu
- Departments of Pharmacology and Therapeutics, University of Florida, Gainesville, Florida 32610-0235
| | - Xiaoping Qi
- Departments of Anatomy and Cell Biology, University of Florida, Gainesville, Florida 32610-0235
| | - Sanford L Boye
- Department of Ophthalmology, University of Florida, Gainesville, Florida 32610-0235
| | - William W Hauswirth
- Department of Ophthalmology, University of Florida, Gainesville, Florida 32610-0235
| | - Maria B Grant
- Departments of Pharmacology and Therapeutics, University of Florida, Gainesville, Florida 32610-0235
| | - Michael E Boulton
- Departments of Anatomy and Cell Biology, University of Florida, Gainesville, Florida 32610-0235.
| |
Collapse
|
14
|
Peränen J. Rab8 GTPase as a regulator of cell shape. Cytoskeleton (Hoboken) 2011; 68:527-39. [PMID: 21850707 DOI: 10.1002/cm.20529] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2011] [Revised: 08/08/2011] [Accepted: 08/09/2011] [Indexed: 12/14/2022]
Abstract
Endogenous Rab8 is found in dynamic cell structures like filopodia, lamellipodia, protrusions, ruffles, and primary cilia. Activation of Rab8 is linked to the formation of these actin containing structures, whereas inhibition of Rab8 affects negatively their appearance. The activity of Rab8 is controlled by specific guanine nucleotide exchange factors and GTPase activating proteins. Rab8 regulates a membrane recycling pathway that is linked to Arf6, EHD1, Myo5, and Rab11. A hypothesis is presented on the role of Rab8 in the formation of new cell surface domains. The review focuses on the function of Rab8 in cell migration, epithelial polarization, neuron differentiation, and ciliogenesis.
Collapse
Affiliation(s)
- Johan Peränen
- Institute of Biotechnology, University of Helsinki, Finland.
| |
Collapse
|
15
|
Abstract
AD (Alzheimer's disease) is a neurodegenerative disease characterized by a gradual loss of neurons and the accumulation of neurotoxic Aβ (amyloid β-peptide) and hyperphosphorylated tau. The discovery of mutations in three genes, PSEN1 (presenilin 1), PSEN2 (presenilin 2) and APP (amyloid precursor protein), in patients with FAD (familial AD) has made an important contribution towards an understanding of the disease aetiology; however, a complete molecular mechanism is still lacking. Both presenilins belong to the γ-secretase complex, and serve as the catalytic entity needed for the final cleavage of APP into Aβ. PSEN only functions within the γ-secretase complex through intra- and inter-molecular interactions with three other membrane components, including nicastrin, Aph-1 (anterior pharynx defective-1) and Pen-2 (PSEN enhancer-2). However, although the list of γ-secretase substrates is still expanding, other non-catalytic activities of presenilins are also increasing the complexity behind its molecular contribution towards AD. These γ-secretase-independent roles are so far mainly attributed to PSEN1, including the transport of membrane proteins, cell adhesion, ER (endoplasmic reticulum) Ca(2+) regulation and cell signalling. In the present minireview, we discuss the current understanding of the γ-secretase-independent roles of PSENs and their possible implications in respect of AD.
Collapse
|
16
|
Hass MR, Sato C, Kopan R, Zhao G. Presenilin: RIP and beyond. Semin Cell Dev Biol 2008; 20:201-10. [PMID: 19073272 DOI: 10.1016/j.semcdb.2008.11.014] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2008] [Revised: 11/19/2008] [Accepted: 11/19/2008] [Indexed: 12/22/2022]
Abstract
Over the years the presenilins (PSENs), a family of multi-transmembrane domain proteins, have been ascribed a number of diverse potential functions. Recent in vivo evidence has supported the existence of PSEN functions beyond its well-established role in regulated intramembrane proteolysis. In this review, we will briefly discuss the ability of PSEN to modulate cellular signaling pathways through gamma-secretase cleavage of transmembrane proteins. Additionally, we will critically examine the proposed roles of PSEN in the regulation of beta-catenin function, protein trafficking, calcium regulation, and apoptosis.
Collapse
Affiliation(s)
- Matthew R Hass
- Department of Developmental Biology, Washington University School of Medicine, Saint Louis, MO, United States
| | | | | | | |
Collapse
|
17
|
Reid PC, Urano Y, Kodama T, Hamakubo T. Alzheimer's disease: cholesterol, membrane rafts, isoprenoids and statins. J Cell Mol Med 2007; 11:383-92. [PMID: 17635634 PMCID: PMC3922347 DOI: 10.1111/j.1582-4934.2007.00054.x] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is a heterogeneous neurodegenerative disorder and the most prevalent form of dementia worldwide. AD is characterized pathologically by amyloid-β plaques, neurofibrillary tangles and neuronal loss, and clinically by a progressive loss of cognitive abilities. At present, the fundamental molecular mechanisms underlying the disease are unclear and no treatment for AD is known. Epidemiological evidence continues to mount linking vascular diseases, such as hypertension and diabetes, and hypercholesterolaemia with an increased risk for developing AD. A growing amount of evidence suggests a mechanistic link between cholesterol metabolism in the brain and the formation of amyloid plaques in AD development. Cholesterol and statins clearly modulate β-amyloid precursor protein (βAPP) processing in cell culture and animal models. Statins not only reduce endogenous cholesterol synthesis but also exert other various pleiotrophic effects, such as the reduction in protein isoprenylation. Through these effects statins modulate a variety of cellular functions involving both cholesterol (and membrane rafts) and isoprenylation. Although clearly other factors, such as vascular inflammation, oxidative stress and genetic factors, are intimately linked with the progression of AD, this review focuses on the present research findings describing the effect of cholesterol, membrane rafts and isoprenylation in regulating βAPP processing and in particular γ-secretase complex assembly and function and AD progression, along with consideration for the potential role statins may play in modulating these events.
Collapse
Affiliation(s)
- Patrick C Reid
- Laboratory for Systems Biology and Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
- PeptiDream Inc., Tokyo, Japan
- *Correspondence to: Takao HAMAKUBO Department of Molecular Biology and Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, #35 4-6-1 Komaba, Meguro-ku, Tokyo 153-8904, Japan. Tel.: +81-3-5452-5231; Fax: +81-3-5452-5232 E-mail:
| | - Yasuomi Urano
- Laboratory for Systems Biology and Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
- Department of Biochemistry, Dartmouth Medical School, Hanover, NH, USA
| | - Tatsuhiko Kodama
- Laboratory for Systems Biology and Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Takao Hamakubo
- Laboratory for Systems Biology and Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
- Department of Molecular Biology and Medicine, The University of Tokyo, Tokyo, Japan
- *Correspondence to: Takao HAMAKUBO Department of Molecular Biology and Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, #35 4-6-1 Komaba, Meguro-ku, Tokyo 153-8904, Japan. Tel.: +81-3-5452-5231; Fax: +81-3-5452-5232 E-mail:
| |
Collapse
|
18
|
Longitudinal, quantitative assessment of amyloid, neuroinflammation, and anti-amyloid treatment in a living mouse model of Alzheimer's disease enabled by positron emission tomography. J Neurosci 2007; 27:10957-68. [PMID: 17928437 DOI: 10.1523/jneurosci.0673-07.2007] [Citation(s) in RCA: 223] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
We provide the first evidence for the capability of a high-resolution positron emission tomographic (PET) imaging system in quantitatively mapping amyloid accumulation in living amyloid precursor protein transgenic (Tg) mice. After the intravenous administration of N-[11C]methyl-2-(4'-methylaminophenyl)-6-hydroxybenzothiazole (or [11C]PIB for "Pittsburgh Compound-B") with high-specific radioactivity, the Tg mice exhibited high-level retention of radioactivity in amyloid-rich regions. PET investigation for Tg mice over an extended range of ages, including longitudinal assessments, demonstrated age-dependent increase in radioligand binding consistent with progressive amyloid accumulation. Reduction in amyloid levels in the hippocampus of Tg mice was also successfully monitored by multiple PET scans along the time course of anti-amyloid treatment using an antibody against amyloid beta peptide (Abeta). Moreover, PET scans with [18F]fluoroethyl-DAA1106, a radiotracer for activated glia, were conducted for these individuals parallel to amyloid imaging, revealing treatment-induced neuroinflammatory responses, the magnitude of which intimately correlated with the levels of pre-existing amyloid estimated by [11C]PIB. It is also noteworthy that the localization and abundance of [11C]PIB autoradiographic signals were closely associated with those of N-terminally truncated and modified Abeta, AbetaN3-pyroglutamate, in Alzheimer's disease (AD) and Tg mouse brains, implying that the detectability of amyloid by [11C]PIB positron emission tomography is dependent on the accumulation of specific Abeta subtypes. Our results support the usefulness of the small animal-dedicated PET system in conjunction with high-specific radioactivity probes and appropriate Tg models not only for clarifying the mechanistic properties of amyloidogenesis in mouse models but also for preclinical tests of emerging diagnostic and therapeutic approaches to AD.
Collapse
|
19
|
Uno T, Nakada T, Okamaoto S, Nakamura M, Matsubara M, Imaishi H, Yamagata H, Kanamaru K, Takagi M. Determination of phosphorylated amino acid residues of Rab8 from Bombyx mori. ARCHIVES OF INSECT BIOCHEMISTRY AND PHYSIOLOGY 2007; 66:89-97. [PMID: 17879235 DOI: 10.1002/arch.20201] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
The Rab family of small GTPases are key regulators of membrane trafficking. Partially purified Rab8 from Bombyx mori (BRab8) was phosphorylated by protein kinase C in mammalian cells in vitro. To determine which of the seven serines and four threonines are phosphorylated, we generated deletion and site-directed mutants of BRab8, inserted them in Escherichia coli, partially purified the encoded fusion proteins by affinity chromatography, and examined their phosphorylation by protein kinase C in vitro. We found that Ser-132 of BRab8 was specifically phosphorylated by protein kinase C. In addition, Western blotting using an antiserum against BRab8 and in-gel staining for phosphorylated proteins revealed that BRab8 is phosphorylated in vivo.
Collapse
Affiliation(s)
- Tomohide Uno
- Laboratory of Biological Chemistry, Department of Biofunctional Chemistry, Faculty of Agriculture, Kobe University, Kobe, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Hu Z, Zeng L, Huang Z, Zhang J, Li T. The Study of Golgi Apparatus in Alzheimer’s Disease. Neurochem Res 2007; 32:1265-77. [PMID: 17401657 DOI: 10.1007/s11064-007-9302-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2006] [Accepted: 01/30/2007] [Indexed: 10/23/2022]
Abstract
Alzheimer's disease is an irreversible, progressive neurodegenerative disorder leading invariably to death, usually within 7-10 years after diagnosis and is the leading cause of dementia in the elderly. Not only is Alzheimer's disease a tragic disease in which people suffer from neurodegeneration in the years to come, it also becomes an incredible burden on the public health system. However, there is currently no effective treatment to halt the progression or prevent the onset of Alzheimer's disease. This is partly due to the fact that the complex pathophysiology of Alzheimer's disease is not yet completely understood. Recently, Golgi apparatus is found to play an important role in Alzheimer's disease. In this review, we discuss the changes of Golgi apparatus during clinical progression and pathological development of Alzheimer's disease. First, changes of Golgi apparatus size in Alzheimer's disease are summarized. We then address the role of Golgi apparatus in the neuropathology of Alzheimer's disease. Finally, the role of Golgi apparatus in the pathogenesis of Alzheimer's disease is discussed. Understanding the contribution of Golgi apparatus dysfunction to Alzheimer's disease and its pathophysiological basis will significantly impact our ability to develop more effective therapies for Alzheimer's disease.
Collapse
Affiliation(s)
- Zhiping Hu
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China
| | | | | | | | | |
Collapse
|
21
|
Hattula K, Furuhjelm J, Tikkanen J, Tanhuanpää K, Laakkonen P, Peränen J. Characterization of the Rab8-specific membrane traffic route linked to protrusion formation. J Cell Sci 2006; 119:4866-77. [PMID: 17105768 DOI: 10.1242/jcs.03275] [Citation(s) in RCA: 177] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Rab8 has a drastic effect on cell shape, but the membrane trafficking route it regulates is poorly defined. Here, we show that endogenous and ectopically expressed Rab8 is associated with macropinosomes generated at ruffling membrane domains. These macropinosomes fuse or transform into tubules that move toward the cell center, from where they are recycled back to the leading edge. The biogenesis of these tubules is dependent on actin and microtubular dynamics. Expression of dominant-negative Rab8 mutants or depletion of Rab8 by RNA interference inhibit protrusion formation, but promote cell-cell adhesion and actin stress fiber formation, whereas expression of the constitutively active Rab8-Q67L has the opposite effect. Rab8 localization overlaps with both Rab11 and Arf6, and is functionally linked to Arf6. We also demonstrate that Rab8 activity is needed for the transport of transferrin and the transferrin receptor to the pericentriolar region and to cell protrusions, and that Rab8 controls the traffic of cholera toxin B to the Golgi compartment. Finally, Rab8 colocalizes and binds specifically to a synaptotagmin-like protein (Slp1/JFC1), which is involved in controlling Rab8 membrane dynamics. We propose that Rab8 regulates a membrane-recycling pathway that mediates protrusion formation.
Collapse
Affiliation(s)
- Katarina Hattula
- Institute of Biotechnology, PO Box 56 (Viikinkaari 9), FIN-00014 University of Helsinki, Finland
| | | | | | | | | | | |
Collapse
|
22
|
Cole SL, Vassar R. Isoprenoids and Alzheimer's disease: a complex relationship. Neurobiol Dis 2006; 22:209-22. [PMID: 16406223 DOI: 10.1016/j.nbd.2005.11.007] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2005] [Revised: 11/04/2005] [Accepted: 11/13/2005] [Indexed: 11/22/2022] Open
Abstract
Cholesterol metabolism has been linked to Alzheimer's disease (AD) neuropathology, which is characterized by amyloid plaques, neurofibrillary tangles and neuroinflammation. Indeed, the use of statins, which inhibit cholesterol and isoprenoid biosynthesis, as potential AD therapeutics is under investigation. Whether statins offer benefit for AD will be determined by the outcome of large, placebo-controlled, randomized clinical trials. However, their use as pharmacological tools has delineated novel roles for isoprenoids in AD. Protein isoprenylation regulates multiple cellular and molecular events and here we review the complex roles of isoprenoids in AD-relevant processes and carefully evaluate isoprenoid pathways as potential AD therapeutic targets.
Collapse
Affiliation(s)
- S L Cole
- Department of Cell and Molecular Biology, Northwestern University Medical School, Chicago, IL 60611, USA.
| | | |
Collapse
|
23
|
Suga K, Tomiyama T, Mori H, Akagawa K. Syntaxin 5 interacts with presenilin holoproteins, but not with their N- or C-terminal fragments, and affects beta-amyloid peptide production. Biochem J 2004; 381:619-28. [PMID: 15109302 PMCID: PMC1133870 DOI: 10.1042/bj20040618] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2004] [Accepted: 04/27/2004] [Indexed: 12/16/2022]
Abstract
Mutations in presenilins 1 and 2 (PS1 and PS2) account for the majority of cases of early-onset familial Alzheimer's disease. However, the trafficking and interaction of PSs with other proteins in the early secretory pathways are poorly understood. Using co-immunoprecipitation, we found that PS bound to Syx5 (syntaxin 5), which is a target-soluble N-ethylmaleimide-sensitive fusion protein attachment protein receptor involved in endoplasmic reticulum (ER)-Golgi vesicular transport in vivo. Syx5 interacted only with the full-length PS holoproteins and not with the naturally occurring N- or C-terminal fragments. The PS holoproteins co-immunoprecipitated with the mutant Syx5, which localized to the ER and Golgi compartments, despite the substitution of the transmembrane region with that of syntaxin 1A. In contrast, the transmembrane deletion mutant that localized to the cytosol, but not to the ER or Golgi compartments, did not co-immunoprecipitate the PS holoproteins. The PS1 variant linked to familial Alzheimer's disease (PS1DeltaE9), lacking the region that contains the endoproteolytic cleavage site in the cytoplasmic loop, showed markedly decreased binding to Syx5. Immunofluorescence and sucrose-density-gradient fractionation analyses showed that the full-length PS holoproteins co-localized with Syx5 to the ER and cis-Golgi compartments. Furthermore, Syx5 overexpression resulted in the accumulation of PS holoproteins and the beta-amyloid precursor protein, and reduced the secretion of the Abeta (amyloid beta) peptide in COS-7 cells. In summary, these results indicate that Syx5 binds to full-length PSs and affects the processing and trafficking of beta-amyloid precursor protein in the early secretory compartments.
Collapse
Affiliation(s)
- Kei Suga
- Department of Cell Physiology, Kyorin University School of Medicine, 6-20-2, Shinkawa, Mitaka, Tokyo 181-8611, Japan.
| | | | | | | |
Collapse
|