1
|
Zhou F, Hu R, Wang Y, Wu X, Chen X, Xi Z, Zeng K. Calsyntenin-1 expression and function in brain tissue of lithium-pilocarpine rat seizure models. Synapse 2024; 78:e22307. [PMID: 39171546 DOI: 10.1002/syn.22307] [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: 04/09/2024] [Revised: 08/05/2024] [Accepted: 08/07/2024] [Indexed: 08/23/2024]
Abstract
To present the expression of calsyntenin-1 (Clstn1) in the brain and investigate the potential mechanism of Clstn1 in lithium-pilocarpine rat seizure models. Thirty-five male SD adult rats were induced to have seizures by intraperitoneal injection of lithium chloride pilocarpine. Rats exhibiting spontaneous seizures were divided into the epilepsy (EP) group (n = 15), whereas those without seizures were divided into the control group (n = 14). Evaluate the expression of Clstn1 in the temporal lobe of two groups using Western blotting, immunohistochemistry, and immunofluorescence. Additionally, 55 male SD rats were subjected to status epilepticus (SE) using the same induction method. Rats experiencing seizures exceeding Racine's level 4 (n = 48) were randomly divided into three groups: SE, SE + control lentivirus (lentiviral vector expressing green fluorescent protein [LV-GFP]), and SE + Clstn1-targeted RNA interference lentivirus (LV-Clstn1-RNAi). The LV-GFP group served as a control for the lentiviral vector, whereas the LV-Clstn1-RNAi group received a lentivirus designed to silence Clstn1 expression. These lentiviral treatments were administered via hippocampal stereotactic injection 2 days after SE induction. Seven days after SE, Western blot analysis was performed to evaluate the expression of Clstn1 in the hippocampus and temporal lobe. Meanwhile, we observed the latency of spontaneous seizures and the frequency of spontaneous seizures within 8 weeks among the three groups. The expression of Clstn1 in the cortex and hippocampus of the EP group was significantly increased compared to the control group (p < .05). Immunohistochemistry and immunofluorescence showed that Clstn1 was widely distributed in the cerebral cortex and hippocampus of rats, and colocalization analysis revealed that it was mainly co expressed with neurons in the cytoplasm. Compared with the SE group (11.80 ± 2.17 days) and the SE + GFP group (12.40 ± 1.67 days), there was a statistically significant difference (p < .05) in the latency period of spontaneous seizures (15.14 ± 2.41 days) in the SE + Clstn1 + RNAi group rats. Compared with the SE group (4.60 ± 1.67 times) and the SE + GFP group (4.80 ± 2.05 times), the SE + Clstn1 + RNAi group (2.0 ± .89 times) showed a significant reduction in the frequency of spontaneous seizures within 2 weeks of chronic phase in rats (p < .05). Elevated Clstn1 expression in EP group suggests its role in EP onset. Targeting Clstn1 may be a potential therapeutic approach for EP management.
Collapse
Affiliation(s)
- Fu Zhou
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Rong Hu
- Department of Neurology, Pizhou People's Hospital, Jiangsu, China
| | - Yuzhu Wang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaohui Wu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xuan Chen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhiqin Xi
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Kebin Zeng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
2
|
Honda K, Takahashi H, Hata S, Abe R, Saito T, Saido TC, Taru H, Sobu Y, Ando K, Yamamoto T, Suzuki T. Suppression of the amyloidogenic metabolism of APP and the accumulation of Aβ by alcadein α in the brain during aging. Sci Rep 2024; 14:18471. [PMID: 39122814 PMCID: PMC11316129 DOI: 10.1038/s41598-024-69400-9] [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: 03/10/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024] Open
Abstract
Generation and accumulation of amyloid-β (Aβ) protein in the brain are the primary causes of Alzheimer's disease (AD). Alcadeins (Alcs composed of Alcα, Alcβ and Alcγ family) are a neuronal membrane protein that is subject to proteolytic processing, as is Aβ protein precursor (APP), by APP secretases. Previous observations suggest that Alcs are involved in the pathophysiology of Alzheimer's disease (AD). Here, we generated new mouse AppNL-F (APP-KI) lines with either Alcα- or Alcβ-deficient background and analyzed APP processing and Aβ accumulation through the aging process. The Alcα-deficient APP-KI (APP-KI/Alcα-KO) mice enhanced brain Aβ accumulation along with increased amyloidogenic β-site cleavage of APP through the aging process whereas Alcβ-deficient APP-KI (APP-KI/Alcβ-KO) mice neither affected APP metabolism nor Aβ accumulation at any age. More colocalization of APP and BACE1 was observed in the endolysosomal pathway in neurons of APP-KI/Alcα-KO mice compared to APP-KI and APP-KI/Alcβ-KO mice. These results indicate that Alcα plays an important role in the neuroprotective function by suppressing the amyloidogenic cleavage of APP by BACE1 in the brain, which is distinct from the neuroprotective function of Alcβ, in which p3-Alcβ peptides derived from Alcβ restores the viability in neurons impaired by toxic Aβ.
Collapse
Affiliation(s)
- Keiko Honda
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan
- Advanced Prevention and Research Laboratory for Dementia, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan
| | - Hiroo Takahashi
- Department of Molecular Neurobiology, Faculty of Medicine, Kagawa University, Miki-cho, 761-0793, Japan
| | - Saori Hata
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan
- Advanced Prevention and Research Laboratory for Dementia, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan
- Bioproduction Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Sapporo, 062-8517, Japan
| | - Ruriko Abe
- Advanced Prevention and Research Laboratory for Dementia, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan
- Bioproduction Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Sapporo, 062-8517, Japan
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, 467-8601, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science Institute, Wako, 351-0198, Japan
| | - Hidenori Taru
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan
- Advanced Prevention and Research Laboratory for Dementia, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan
| | - Yuriko Sobu
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan
- Laboratory of Neuronal Regeneration, Graduate School of Brain Science, Doshisha University, Kyotanabe, 610-0394, Japan
| | - Kanae Ando
- Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Hachioji, Tokyo, 192-0397, Japan
| | - Tohru Yamamoto
- Department of Molecular Neurobiology, Faculty of Medicine, Kagawa University, Miki-cho, 761-0793, Japan.
| | - Toshiharu Suzuki
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan.
- Advanced Prevention and Research Laboratory for Dementia, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan.
- Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Hachioji, Tokyo, 192-0397, Japan.
| |
Collapse
|
3
|
Longobardi A, Bellini S, Nicsanu R, Pilotto A, Geviti A, Facconi A, Tolassi C, Libri I, Saraceno C, Fostinelli S, Borroni B, Padovani A, Binetti G, Ghidoni R. Unveiling New Genetic Variants Associated with Age at Onset in Alzheimer's Disease and Frontotemporal Lobar Degeneration Due to C9orf72 Repeat Expansions. Int J Mol Sci 2024; 25:7457. [PMID: 39000564 PMCID: PMC11242823 DOI: 10.3390/ijms25137457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/04/2024] [Accepted: 07/05/2024] [Indexed: 07/16/2024] Open
Abstract
Alzheimer's disease (AD) and Frontotemporal lobar degeneration (FTLD) represent the most common forms of neurodegenerative dementias with a highly phenotypic variability. Herein, we investigated the role of genetic variants related to the immune system and inflammation as genetic modulators in AD and related dementias. In patients with sporadic AD/FTLD (n = 300) and GRN/C9orf72 mutation carriers (n = 80), we performed a targeted sequencing of 50 genes belonging to the immune system and inflammation, selected based on their high expression in brain regions and low tolerance to genetic variation. The linear regression analyses revealed two genetic variants: (i) the rs1049296 in the transferrin (TF) gene, shown to be significantly associated with age at onset in the sporadic AD group, anticipating the disease onset of 4 years for each SNP allele with respect to the wild-type allele, and (ii) the rs7550295 in the calsyntenin-1 (CLSTN1) gene, which was significantly associated with age at onset in the C9orf72 group, delaying the disease onset of 17 years in patients carrying the SNP allele. In conclusion, our data support the role of genetic variants in iron metabolism (TF) and in the modulation of the calcium signalling/axonal anterograde transport of vesicles (CLSTN1) as genetic modulators in AD and FTLD due to C9orf72 expansions.
Collapse
Affiliation(s)
- Antonio Longobardi
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy; (S.B.); (R.N.); (C.S.); (R.G.)
| | - Sonia Bellini
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy; (S.B.); (R.N.); (C.S.); (R.G.)
| | - Roland Nicsanu
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy; (S.B.); (R.N.); (C.S.); (R.G.)
| | - Andrea Pilotto
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy; (A.P.); (C.T.); (I.L.); (B.B.); (A.P.)
- Neurology Unit, Department of Continuity of Care and Frailty, ASST Spedali Civili Hospital, 25123 Brescia, Italy
- Neurobiorepository and Laboratory of Advanced Biological Markers, University of Brescia and ASST Spedali Civili Hospital, 25123 Brescia, Italy
| | - Andrea Geviti
- Service of Statistics, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy; (A.G.); (A.F.)
| | - Alessandro Facconi
- Service of Statistics, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy; (A.G.); (A.F.)
| | - Chiara Tolassi
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy; (A.P.); (C.T.); (I.L.); (B.B.); (A.P.)
- Neurology Unit, Department of Continuity of Care and Frailty, ASST Spedali Civili Hospital, 25123 Brescia, Italy
- Neurobiorepository and Laboratory of Advanced Biological Markers, University of Brescia and ASST Spedali Civili Hospital, 25123 Brescia, Italy
| | - Ilenia Libri
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy; (A.P.); (C.T.); (I.L.); (B.B.); (A.P.)
| | - Claudia Saraceno
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy; (S.B.); (R.N.); (C.S.); (R.G.)
| | - Silvia Fostinelli
- MAC-Memory Clinic and Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy;
| | - Barbara Borroni
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy; (A.P.); (C.T.); (I.L.); (B.B.); (A.P.)
- Cognitive and Behavioural Neurology, ASST Spedali Civili Hospital, 25123 Brescia, Italy
| | - Alessandro Padovani
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy; (A.P.); (C.T.); (I.L.); (B.B.); (A.P.)
- Neurology Unit, Department of Continuity of Care and Frailty, ASST Spedali Civili Hospital, 25123 Brescia, Italy
- Neurobiorepository and Laboratory of Advanced Biological Markers, University of Brescia and ASST Spedali Civili Hospital, 25123 Brescia, Italy
- Brain Health Center, University of Brescia, 25123 Brescia, Italy
| | - Giuliano Binetti
- MAC-Memory Clinic and Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy;
| | - Roberta Ghidoni
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy; (S.B.); (R.N.); (C.S.); (R.G.)
| |
Collapse
|
4
|
Shiraki Y, Mitsuma M, Takada R, Hata S, Kitamura A, Takada S, Kinjo M, Taru H, Müller UC, Yamamoto T, Sobu Y, Suzuki T. Axonal transport of Frizzled5 by Alcadein α-containing vesicles is associated with kinesin-1. Mol Biol Cell 2023; 34:ar110. [PMID: 37585286 PMCID: PMC10559311 DOI: 10.1091/mbc.e22-10-0495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 08/07/2023] [Accepted: 08/10/2023] [Indexed: 08/18/2023] Open
Abstract
Alcadein α (Alcα) and amyloid-β protein precursor (APP) are cargo receptors that associate vesicles with kinesin-1. These vesicles, which contain either Alcα or APP, transport various proteins/cargo molecules into axon nerve terminals. Here, we analyzed immune-isolated Alcα- and APP-containing vesicles of adult mouse brains with LC-MS/MS and identified proteins present in vesicles that contained either Alcα or APP. Among these proteins, Frizzled-5 (Fzd5), a Wnt receptor, was detected mainly in Alcα vesicles. Although colocalization ratios of Fzd5 with Alcα are low in the neurites of differentiating neurons by a low expression of Fzd5 in embryonic brains, the suppression of Alcα expression decreased the localization of Fzd5 in neurites of primary cultured neurons. Furthermore, Fzd5-EGFP expressed in primary cultured neurons was preferentially transported in axons with the transport velocities of Alcα vesicles. In synaptosomal fractions of adult-mice brains that express higher levels of Fzd5, the amount of Fzd5 and the phosphorylation level of calcium/calmodulin-dependent protein kinase-II were reduced in the Alcα-deficient mice. These results suggest that reduced transport of Fzd5 by Alcα-containing vesicles associated with kinesin-1 in axon terminals may impair the response to Wnt ligands in the noncanonical Ca2+-dependent signal transduction pathway at nerve terminals of mature neurons.
Collapse
Affiliation(s)
- Yuzuha Shiraki
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Monet Mitsuma
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Ritsuko Takada
- Exploratory Research Center on Life and Living Systems (ExCELLS), Okazaki, Aichi 444-8787, Japan
- National Institute for Basic Biology, National Institute of Natural Sciences, Okazaki, Aichi 444-8787, Japan
| | - Saori Hata
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
- Bioproduction Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Sapporo 062-8517, Japan
- Advanced Prevention and Research Laboratory for Dementia, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Akira Kitamura
- Laboratory of Molecular Cell Dynamics, Faculty of Advanced Life Science, Hokkaido University, Sapporo 001-0021, Japan
- AMED-PRIME, Japan Agency for Medical Research and Development, 1-7-1 Otemachi, Chiyoda-ku, Tokyo 100-0004 Japan
| | - Shinji Takada
- Exploratory Research Center on Life and Living Systems (ExCELLS), Okazaki, Aichi 444-8787, Japan
- National Institute for Basic Biology, National Institute of Natural Sciences, Okazaki, Aichi 444-8787, Japan
| | - Masataka Kinjo
- Laboratory of Molecular Cell Dynamics, Faculty of Advanced Life Science, Hokkaido University, Sapporo 001-0021, Japan
| | - Hidenori Taru
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
- Advanced Prevention and Research Laboratory for Dementia, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Ulrike C. Müller
- Institute of Pharmacy and Molecular Biotechnology, Im Neuenheimer Feld 364, 69120 Heidelberg, Germany
| | - Tohru Yamamoto
- Department of Molecular Neurobiology, Faculty of Medicine, Kagawa University, Miki-cho, Kagawa 761-0793, Japan
| | - Yuriko Sobu
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
- Advanced Prevention and Research Laboratory for Dementia, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
- Laboratory of Neuronal Regeneration, Graduate School of Brain Science, Doshisha University, Kyotanabe 610-0394, Japan
| | - Toshiharu Suzuki
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
- Advanced Prevention and Research Laboratory for Dementia, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
- Department of Biological Sciences, Tokyo Metropolitan University, Tokyo 192-0397, Japan
| |
Collapse
|
5
|
Kumari D, Ray K. Phosphoregulation of Kinesins Involved in Long-Range Intracellular Transport. Front Cell Dev Biol 2022; 10:873164. [PMID: 35721476 PMCID: PMC9203973 DOI: 10.3389/fcell.2022.873164] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 04/29/2022] [Indexed: 12/28/2022] Open
Abstract
Kinesins, the microtubule-dependent mechanochemical enzymes, power a variety of intracellular movements. Regulation of Kinesin activity and Kinesin-Cargo interactions determine the direction, timing and flux of various intracellular transports. This review examines how phosphorylation of Kinesin subunits and adaptors influence the traffic driven by Kinesin-1, -2, and -3 family motors. Each family of Kinesins are phosphorylated by a partially overlapping set of serine/threonine kinases, and each event produces a unique outcome. For example, phosphorylation of the motor domain inhibits motility, and that of the stalk and tail domains induces cargo loading and unloading effects according to the residue and context. Also, the association of accessory subunits with cargo and adaptor proteins with the motor, respectively, is disrupted by phosphorylation. In some instances, phosphorylation by the same kinase on different Kinesins elicited opposite outcomes. We discuss how this diverse range of effects could manage the logistics of Kinesin-dependent, long-range intracellular transport.
Collapse
|
6
|
Liu Z, Jiang M, Liakath-Ali K, Sclip A, Ko J, Zhang RS, Südhof TC. Deletion of Calsyntenin-3, an atypical cadherin, suppresses inhibitory synapses but increases excitatory parallel-fiber synapses in cerebellum. eLife 2022; 11:e70664. [PMID: 35420982 PMCID: PMC9064300 DOI: 10.7554/elife.70664] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 04/14/2022] [Indexed: 01/11/2023] Open
Abstract
Cadherins contribute to the organization of nearly all tissues, but the functions of several evolutionarily conserved cadherins, including those of calsyntenins, remain enigmatic. Puzzlingly, two distinct, non-overlapping functions for calsyntenins were proposed: As postsynaptic neurexin ligands in synapse formation, or as presynaptic kinesin adaptors in vesicular transport. Here, we show that, surprisingly, acute CRISPR-mediated deletion of calsyntenin-3 in mouse cerebellum in vivo causes a large decrease in inhibitory synapse, but a robust increase in excitatory parallel-fiber synapses in Purkinje cells. As a result, inhibitory synaptic transmission was suppressed, whereas parallel-fiber synaptic transmission was enhanced in Purkinje cells by the calsyntenin-3 deletion. No changes in the dendritic architecture of Purkinje cells or in climbing-fiber synapses were detected. Sparse selective deletion of calsyntenin-3 only in Purkinje cells recapitulated the synaptic phenotype, indicating that calsyntenin-3 acts by a cell-autonomous postsynaptic mechanism in cerebellum. Thus, by inhibiting formation of excitatory parallel-fiber synapses and promoting formation of inhibitory synapses in the same neuron, calsyntenin-3 functions as a postsynaptic adhesion molecule that regulates the excitatory/inhibitory balance in Purkinje cells.
Collapse
Affiliation(s)
- Zhihui Liu
- Department of Molecular and Cellular Physiology, Stanford UniversityStanfordUnited States
- Howard Hughes Medical Institute, Stanford UniversityStanfordUnited States
| | - Man Jiang
- Department of Molecular and Cellular Physiology, Stanford UniversityStanfordUnited States
| | - Kif Liakath-Ali
- Department of Molecular and Cellular Physiology, Stanford UniversityStanfordUnited States
| | - Alessandra Sclip
- Department of Molecular and Cellular Physiology, Stanford UniversityStanfordUnited States
- Howard Hughes Medical Institute, Stanford UniversityStanfordUnited States
| | - Jaewon Ko
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and TechnologyDaeguRepublic of Korea
| | - Roger Shen Zhang
- Department of Molecular and Cellular Physiology, Stanford UniversityStanfordUnited States
| | - Thomas C Südhof
- Department of Molecular and Cellular Physiology, Stanford UniversityStanfordUnited States
- Howard Hughes Medical Institute, Stanford UniversityStanfordUnited States
| |
Collapse
|
7
|
Ding C, Wu Y, Dabas H, Hammarlund M. Activation of the CaMKII-Sarm1-ASK1-p38 MAP kinase pathway protects against axon degeneration caused by loss of mitochondria. eLife 2022; 11:73557. [PMID: 35285800 PMCID: PMC8920508 DOI: 10.7554/elife.73557] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 01/25/2022] [Indexed: 12/22/2022] Open
Abstract
Mitochondrial defects are tightly linked to axon degeneration, yet the underlying cellular mechanisms remain poorly understood. In Caenorhabditis elegans, PVQ axons that lack mitochondria degenerate spontaneously with age. Using an unbiased genetic screen, we found that cell-specific activation of CaMKII/UNC-43 suppresses axon degeneration due to loss of mitochondria. Unexpectedly, CaMKII/UNC-43 activates the conserved Sarm1/TIR-1-ASK1/NSY-1-p38 MAPK pathway and eventually the transcription factor CEBP-1 to protect against degeneration. In addition, we show that disrupting a trafficking complex composed of calsyntenin/CASY-1, Mint/LIN-10, and kinesin suppresses axon degeneration. Further analysis indicates that disruption of this trafficking complex activates the CaMKII-Sarm1-MAPK pathway through L-type voltage-gated calcium channels. Our findings identify CaMKII as a pivot point between mitochondrial defects and axon degeneration, describe how it is regulated, and uncover a surprising neuroprotective role for the Sarm1-p38 MAPK pathway in this context.
Collapse
Affiliation(s)
- Chen Ding
- Department of Neuroscience, Yale University School of MedicineNew HavenUnited States
| | - Youjun Wu
- Department of Genetics, Yale University School of MedicineNew HavenUnited States
| | - Hadas Dabas
- Department of Genetics, Yale University School of MedicineNew HavenUnited States
| | - Marc Hammarlund
- Department of Neuroscience, Yale University School of MedicineNew HavenUnited States,Department of Genetics, Yale University School of MedicineNew HavenUnited States
| |
Collapse
|
8
|
Lin T, Tjernberg LO, Schedin-Weiss S. Neuronal Trafficking of the Amyloid Precursor Protein-What Do We Really Know? Biomedicines 2021; 9:801. [PMID: 34356865 PMCID: PMC8301342 DOI: 10.3390/biomedicines9070801] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/29/2021] [Accepted: 07/03/2021] [Indexed: 11/23/2022] Open
Abstract
Alzheimer's disease (AD) is the most common type of dementia, contributing to 60-80% of cases. It is a neurodegenerative disease that usually starts symptomless in the first two to three decades and then propagates into a long-term, irreversible disease, resulting in the progressive loss of memory, reasoning, abstraction and language capabilities. It is a complex disease, involving a large number of entangled players, and there is no effective treatment to cure it or alter its progressive course. Therefore, a thorough understanding of the disease pathology and an early diagnosis are both necessary. AD has two significant pathological hallmarks: extracellular senile plaques composed of amyloid β-peptide (Aβ) and intracellular neurofibrillary tangles composed of hyperphosphorylated tau protein, and the aggregation of Aβ, which starts in earlier stages, is usually claimed to be the primary cause of AD. Secretases that cleave Aβ precursor protein (APP) and produce neurotoxic Aβ reside in distinct organelles of the cell, and current concepts suggest that APP moves between distinct intracellular compartments. Obviously, APP transport and processing are intimately related processes that cannot be dissociated from each other, and, thus, how and where APP is transported determines its processing fate. In this review, we summarize critical mechanisms underlying neuronal APP transport, which we divide into separate parts: (1) secretory pathways and (2) endocytic and autophagic pathways. We also include two lipoprotein receptors that play essential roles in APP transport: sorting-related receptor with A-type repeats and sortilin. Moreover, we consider here some major disruptions in the neuronal transport of APP that contribute to AD physiology and pathology. Lastly, we discuss current methods and technical difficulties in the studies of APP transport.
Collapse
Affiliation(s)
| | - Lars O. Tjernberg
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 17164 Stockholm, Sweden;
| | - Sophia Schedin-Weiss
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 17164 Stockholm, Sweden;
| |
Collapse
|
9
|
Aravind P, Bulbule SR, Hemalatha N, Babu R, Devaraju K. Elevation of gene expression of calcineurin, calmodulin and calsyntenin in oxidative stress induced PC12 cells. Genes Dis 2021; 8:87-93. [PMID: 33569517 PMCID: PMC7859428 DOI: 10.1016/j.gendis.2019.09.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 08/01/2019] [Accepted: 09/01/2019] [Indexed: 12/30/2022] Open
Abstract
In normal physiological conditions, reactive oxygen and nitrogen species are used as important signaling molecules in the cell. However, in excess it causes the disruption of cell resulting in their death. Oxidative stress causes influx in intracellular calcium levels leading to higher concentrations of calcium in the cell. This accelerated calcium affects both the mitochondria and nuclei leading to excitotoxicity in neurons. Intracellular calcium levels are controlled by voltage dependent calcium channels located in the plasma membrane, calcium stores like endoplasmic/sarcoplasmic reticulum and majorly by calcium binding proteins. Our study was aimed at analyzing the gene expression of major calcium binding proteins namely calcineurin, calmodulin, calreticulin, synaptotagamin and calsyntenin in stress induced PC 12 cells. Rotenone (1 μM), Peroxynitrite (10 μM), H2O2 (100 μM) and High glucose (33 mM) were used to induce oxidative stress in PC12 cells. Results obtained from the study suggest that calcineurin, calmodulin and calsyntenin gene expression were enhanced compared to the control due to oxidative stress. However, synaptotagmin and calreticulin gene expression were down regulated. Further, Akt protein expression (stress marker) was enhanced in PC12 cells with all other stress inducers except in hyperglycemic condition.
Collapse
Affiliation(s)
- P. Aravind
- Department of Biochemistry, Karnatak University, Pavate Nagar, Dharwad, 580 003, India
| | - Sarojini R. Bulbule
- Department of Biochemistry, Karnatak University, Pavate Nagar, Dharwad, 580 003, India
| | - N. Hemalatha
- Department of Biochemistry and Nutrition, CFTRI, V V Moholla, Mysore, 570 020, India
| | - R.L. Babu
- Department of Bioinformatics and Biotechnology, Akkamahadevi Women's University, Vijayapura, Karnataka, 586 108, India
| | - K.S. Devaraju
- Department of Biochemistry, Karnatak University, Pavate Nagar, Dharwad, 580 003, India
| |
Collapse
|
10
|
Lee ES, Kim WT, Park GY, Lee M, Gen Son T. Calsyntenin 1 mRNA expression sensitivity to ionizing radiation in human hepatocytes and carcinoma cells and blood cells of BALB/c mice. JOURNAL OF RADIATION RESEARCH AND APPLIED SCIENCES 2020. [DOI: 10.1080/16878507.2020.1855911] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Eon-Seok Lee
- Research Center, Dongnam Institute of Radiological and Medical Science, Busan, Republic of Korea
- Department of Biochemistry, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Won-Tae Kim
- Research Center, Dongnam Institute of Radiological and Medical Science, Busan, Republic of Korea
| | - Ga-Young Park
- Research Center, Dongnam Institute of Radiological and Medical Science, Busan, Republic of Korea
| | - Manwoo Lee
- Research Center, Dongnam Institute of Radiological and Medical Science, Busan, Republic of Korea
| | - Tae Gen Son
- Research Center, Dongnam Institute of Radiological and Medical Science, Busan, Republic of Korea
| |
Collapse
|
11
|
Gotoh N, Saito Y, Hata S, Saito H, Ojima D, Murayama C, Shigeta M, Abe T, Konno D, Matsuzaki F, Suzuki T, Yamamoto T. Amyloidogenic processing of amyloid β protein precursor (APP) is enhanced in the brains of alcadein α-deficient mice. J Biol Chem 2020; 295:9650-9662. [PMID: 32467230 PMCID: PMC7363152 DOI: 10.1074/jbc.ra119.012386] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 05/22/2020] [Indexed: 12/25/2022] Open
Abstract
Alzheimer's disease (AD) is a very common neurodegenerative disorder, chiefly caused by increased production of neurotoxic β-amyloid (Aβ) peptide generated from proteolytic cleavage of β-amyloid protein precursor (APP). Except for familial AD arising from mutations in the APP and presenilin (PSEN) genes, the molecular mechanisms regulating the amyloidogenic processing of APP are largely unclear. Alcadein α/calsyntenin1 (ALCα/CLSTN1) is a neuronal type I transmembrane protein that forms a complex with APP, mediated by the neuronal adaptor protein X11-like (X11L or MINT2). Formation of the ALCα-X11L-APP tripartite complex suppresses Aβ generation in vitro, and X11L-deficient mice exhibit enhanced amyloidogenic processing of endogenous APP. However, the role of ALCα in APP metabolism in vivo remains unclear. Here, by generating ALCα-deficient mice and using immunohistochemistry, immunoblotting, and co-immunoprecipitation analyses, we verified the role of ALCα in the suppression of amyloidogenic processing of endogenous APP in vivo We observed that ALCα deficiency attenuates the association of X11L with APP, significantly enhances amyloidogenic β-site cleavage of APP, especially in endosomes, and increases the generation of endogenous Aβ in the brain. Furthermore, we noted amyloid plaque formation in the brains of human APP-transgenic mice in an ALCα-deficient background. These results unveil a potential role of ALCα in protecting cerebral neurons from Aβ-dependent pathogenicity in AD.
Collapse
Affiliation(s)
- Naoya Gotoh
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Yuhki Saito
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Saori Hata
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Haruka Saito
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Daiki Ojima
- Department of Molecular Neurobiology, Faculty of Medicine, Kagawa University, Takamatsu, Japan
| | - Chiaki Murayama
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
- Department of Molecular Neurobiology, Faculty of Medicine, Kagawa University, Takamatsu, Japan
| | - Mayo Shigeta
- Laboratory for Animal Resource Development, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Takaya Abe
- Laboratory for Animal Resource Development, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
- Laboratory for Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Daijiro Konno
- Laboratory for Cell Asymmetry, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Fumio Matsuzaki
- Laboratory for Cell Asymmetry, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Toshiharu Suzuki
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Tohru Yamamoto
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
- Department of Molecular Neurobiology, Faculty of Medicine, Kagawa University, Takamatsu, Japan
| |
Collapse
|
12
|
Mórotz GM, Glennon EB, Greig J, Lau DHW, Bhembre N, Mattedi F, Muschalik N, Noble W, Vagnoni A, Miller CCJ. Kinesin light chain-1 serine-460 phosphorylation is altered in Alzheimer's disease and regulates axonal transport and processing of the amyloid precursor protein. Acta Neuropathol Commun 2019; 7:200. [PMID: 31806024 PMCID: PMC6896704 DOI: 10.1186/s40478-019-0857-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 11/25/2019] [Indexed: 12/14/2022] Open
Abstract
Damage to axonal transport is an early pathogenic event in Alzheimer’s disease. The amyloid precursor protein (APP) is a key axonal transport cargo since disruption to APP transport promotes amyloidogenic processing of APP. Moreover, altered APP processing itself disrupts axonal transport. The mechanisms that regulate axonal transport of APP are therefore directly relevant to Alzheimer’s disease pathogenesis. APP is transported anterogradely through axons on kinesin-1 motors and one route for this transport involves calsyntenin-1, a type-1 membrane spanning protein that acts as a direct ligand for kinesin-1 light chains (KLCs). Thus, loss of calsyntenin-1 disrupts APP axonal transport and promotes amyloidogenic processing of APP. Phosphorylation of KLC1 on serine-460 has been shown to reduce anterograde axonal transport of calsyntenin-1 by inhibiting the KLC1-calsyntenin-1 interaction. Here we demonstrate that in Alzheimer’s disease frontal cortex, KLC1 levels are reduced and the relative levels of KLC1 serine-460 phosphorylation are increased; these changes occur relatively early in the disease process. We also show that a KLC1 serine-460 phosphomimetic mutant inhibits axonal transport of APP in both mammalian neurons in culture and in Drosophila neurons in vivo. Finally, we demonstrate that expression of the KLC1 serine-460 phosphomimetic mutant promotes amyloidogenic processing of APP. Together, these results suggest that increased KLC1 serine-460 phosphorylation contributes to Alzheimer’s disease.
Collapse
|
13
|
Hata S, Omori C, Kimura A, Saito H, Kimura N, Gupta V, Pedrini S, Hone E, Chatterjee P, Taddei K, Kasuga K, Ikeuchi T, Waragai M, Nishimura M, Hu A, Nakaya T, Meijer L, Maeda M, Yamamoto T, Masters CL, Rowe CC, Ames D, Yamamoto K, Martins RN, Gandy S, Suzuki T. Decrease in p3-Alcβ37 and p3-Alcβ40, products of Alcadein β generated by γ-secretase cleavages, in aged monkeys and patients with Alzheimer's disease. ALZHEIMERS & DEMENTIA-TRANSLATIONAL RESEARCH & CLINICAL INTERVENTIONS 2019; 5:740-750. [PMID: 31754625 PMCID: PMC6854065 DOI: 10.1016/j.trci.2019.09.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Introduction Neuronal p3-Alcβ peptides are generated from the precursor protein Alcadein β (Alcβ) through cleavage by α- and γ-secretases of the amyloid β (Aβ) protein precursor (APP). To reveal whether p3-Alcβ is involved in Alzheimer's disease (AD) contributes for the development of novel therapy and/or drug targets. Methods We developed new sandwich enzyme-linked immunosorbent assay (sELISA) systems to quantitate levels of p3-Alcβ in the cerebrospinal fluid (CSF). Results In monkeys, CSF p3-Alcβ decreases with age, and the aging is also accompanied by decreased brain expression of Alcβ. In humans, CSF p3-Alcβ levels decrease to a greater extent in those with AD than in age-matched controls. Subjects carrying presenilin gene mutations show a significantly lower CSF p3-Alcβ level. A cell study with an inverse modulator of γ-secretase remarkably reduces the generation of p3-Alcβ37 while increasing the production of Aβ42. Discussion Aging decreases the generation of p3-Alcβ, and further significant decrease of p3-Alcβ caused by aberrant γ-secretase activity may accelerate pathogenesis in AD.
Collapse
Affiliation(s)
- Saori Hata
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
- Corresponding author. Tel.:+81-11-706-3250; Fax: +81-11-706-4991.
| | - Chiori Omori
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, University of Tokyo, Kashiwa, Japan
| | - Ayano Kimura
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Haruka Saito
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Nobuyuki Kimura
- Section of Cell Biology and Pathology, Department of Alzheimer's Disease Research, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan
- Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, Tsukuba, Japan
| | - Veer Gupta
- Centre of Excellence for Alzheimer's Disease Research and Care, Sir James McCusker Alzheimer's Disease Research Unit, Edith Cowan University, Joodalup, WA, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
- Co-operative Research Centre for Mental Health, Carlton, VIC, Australia
| | - Steve Pedrini
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
- Co-operative Research Centre for Mental Health, Carlton, VIC, Australia
| | - Eugene Hone
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
- Co-operative Research Centre for Mental Health, Carlton, VIC, Australia
| | - Pratishtha Chatterjee
- Department of Biomedical Sciences, Faculty of Medical and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Kevin Taddei
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - Kensaku Kasuga
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | - Takeshi Ikeuchi
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | - Masaaki Waragai
- Department of Neurology, Higashi Matsudo Municipal Hospital, Matsudo, Japan
| | - Masaki Nishimura
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Otsu, Japan
| | - Anqi Hu
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Tadashi Nakaya
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Laurent Meijer
- ManRos Therapeutics, Centre de Perharidy, Roscoff, Bretagne, France
| | - Masahiro Maeda
- Immuno-Biological Laboratories Co., Ltd. (IBL), Fujioka, Japan
| | - Tohru Yamamoto
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
- Department of Molecular Neurobiology, Faculty of Medicine, Kagawa University, Miki-cho, Kagawa, Japan
| | - Colin L. Masters
- Neurodegeneration Division, The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Chris C. Rowe
- Department of Nuclear Medicine and Centre for PET, Austin Health, Heidelberg, VIC, Australia
| | - David Ames
- National Ageing Research Institute, Parkville, VIC, Australia
- Academic Unit for Psychiatry of Old age, St. George's Hospital, The University of Melbourne, Parkville, VIC, Australia
| | - Kazuo Yamamoto
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, University of Tokyo, Kashiwa, Japan
| | - Ralph N. Martins
- Centre of Excellence for Alzheimer's Disease Research and Care, Sir James McCusker Alzheimer's Disease Research Unit, Edith Cowan University, Joodalup, WA, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
- Co-operative Research Centre for Mental Health, Carlton, VIC, Australia
- Department of Biomedical Sciences, Faculty of Medical and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Sam Gandy
- Mount Sinai Center for Cognitive Health and NFL Neurological Care, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Toshiharu Suzuki
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
- Corresponding author. Tel.:+81-11-706-3250; Fax: +81-11-706-4991.
| |
Collapse
|
14
|
Colás-Algora N, Millán J. How many cadherins do human endothelial cells express? Cell Mol Life Sci 2019; 76:1299-1317. [PMID: 30552441 PMCID: PMC11105309 DOI: 10.1007/s00018-018-2991-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 11/16/2018] [Accepted: 12/06/2018] [Indexed: 12/13/2022]
Abstract
The vasculature is the paradigm of a compartment generated by parallel cellular barriers that aims to transport oxygen, nutrients and immune cells in complex organisms. Vascular barrier dysfunction leads to fatal acute and chronic inflammatory diseases. The endothelial barrier lines the inner side of vessels and is the main regulator of vascular permeability. Cadherins comprise a superfamily of 114 calcium-dependent adhesion proteins that contain conserved cadherin motifs and form cell-cell junctions in metazoans. In mature human endothelial cells, only VE (vascular endothelial)-cadherin and N (neural)-cadherin have been investigated in detail. Although both cadherins are essential for regulating endothelial permeability, no comprehensive expression studies to identify which other family members could play a relevant role in endothelial cells has so far been performed. Here, we have reviewed gene and protein expression databases to analyze cadherin expression in mature human endothelium and found that at least 24 cadherin superfamily members are significantly expressed. Based on data obtained from other cell types, organisms and experimental models, we discuss their potential functions, many of them unrelated to the formation of endothelial cell-cell junctions. The expression of this new set of endothelial cadherins highlights the important but still poorly defined roles of planar cell polarity, the Hippo pathway and mitochondria metabolism in human vascular homeostasis.
Collapse
Affiliation(s)
- Natalia Colás-Algora
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, C/Nicolás Cabrera 1, Cantoblanco, 28049, Madrid, Spain
| | - Jaime Millán
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, C/Nicolás Cabrera 1, Cantoblanco, 28049, Madrid, Spain.
| |
Collapse
|
15
|
Thapliyal S, Vasudevan A, Dong Y, Bai J, Koushika SP, Babu K. The C-terminal of CASY-1/Calsyntenin regulates GABAergic synaptic transmission at the Caenorhabditis elegans neuromuscular junction. PLoS Genet 2018. [PMID: 29529030 PMCID: PMC5864096 DOI: 10.1371/journal.pgen.1007263] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The C. elegans ortholog of mammalian calsyntenins, CASY-1, is an evolutionarily conserved type-I transmembrane protein that is highly enriched in the nervous system. Mammalian calsyntenins are strongly expressed at inhibitory synapses, but their role in synapse development and function is still elusive. Here, we report a crucial role for CASY-1 in regulating GABAergic synaptic transmission at the C. elegans neuromuscular junction (NMJ). The shorter isoforms of CASY-1; CASY-1B and CASY-1C, express and function in GABA motor neurons where they regulate GABA neurotransmission. Using pharmacological, behavioral, electrophysiological, optogenetic and imaging approaches we establish that GABA release is compromised at the NMJ in casy-1 mutants. Further, we demonstrate that CASY-1 is required to modulate the transport of GABAergic synaptic vesicle (SV) precursors through a possible interaction with the SV motor protein, UNC-104/KIF1A. This study proposes a possible evolutionarily conserved model for the regulation of GABA synaptic functioning by calsyntenins. GABA acts as a major inhibitory neurotransmitter in both vertebrate and invertebrate nervous systems. Despite the potential deregulation of GABA signaling in several neurological disorders, our understanding of the genetic factors that regulate GABAergic synaptic transmission has just started to evolve. Here, we identify a role for a cell adhesion molecule, CASY-1, in regulating GABA signaling at the C. elegans NMJ. We show that the mutants in casy-1 have reduced number of GABA vesicles at the synapse resulting in less GABA release from the presynaptic GABAergic motor neurons. Further, we show that the shorter isoforms of the casy-1 gene; casy-1b and casy-1c that carry a potential kinesin-motor binding domain are responsible for maintaining GABAergic signaling at the synapse. We show a novel interaction of the CASY-1 isoforms with the C- terminal of the UNC-104/KIF1A motor protein that mediates the trafficking of GABAergic synaptic vesicle precursors to the synapse, thus maintaining normal inhibitory signaling at the NMJ.
Collapse
Affiliation(s)
- Shruti Thapliyal
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Mohali, Knowledge City, Sector 81, SAS Nagar, Manauli, Punjab, India
| | - Amruta Vasudevan
- Department of Biological Sciences, Tata Institute of Fundamental Research, Colaba, Mumbai, India
| | - Yongming Dong
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109 and Department of Biochemistry, University of Washington, Seattle, WA, United Sttaes of America
| | - Jihong Bai
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109 and Department of Biochemistry, University of Washington, Seattle, WA, United Sttaes of America
| | - Sandhya P. Koushika
- Department of Biological Sciences, Tata Institute of Fundamental Research, Colaba, Mumbai, India
| | - Kavita Babu
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Mohali, Knowledge City, Sector 81, SAS Nagar, Manauli, Punjab, India
- * E-mail: ,
| |
Collapse
|
16
|
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: 5.7] [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
|
17
|
Sobu Y, Furukori K, Chiba K, Nairn AC, Kinjo M, Hata S, Suzuki T. Phosphorylation of multiple sites within an acidic region of Alcadein α is required for kinesin-1 association and Golgi exit of Alcadein α cargo. Mol Biol Cell 2017; 28:3844-3856. [PMID: 29093024 PMCID: PMC5739299 DOI: 10.1091/mbc.e17-05-0301] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 09/18/2017] [Accepted: 10/26/2017] [Indexed: 11/16/2022] Open
Abstract
Alcadein a (Alca) is reported to function as a cargo receptor when associated with kinesin-1. Phosphorylation of three serine residues in the acidic region located between the two WD motifs of Alca is required for interaction with kinesin-1 and Golgi exit of Alca cargo. Alcadein α (Alcα) is a major cargo of kinesin-1 that is subjected to anterograde transport in neuronal axons. Two tryptophan- and aspartic acid-containing (WD) motifs located in its cytoplasmic domain directly bind the tetratricopeptide repeat (TPR) motifs of the kinesin light chain (KLC), which activate kinesin-1 and recruit kinesin-1 to Alcα cargo. We found that phosphorylation of three serine residues in the acidic region located between the two WD motifs is required for interaction with KLC. Phosphorylation of these serine residues may alter the disordered structure of the acidic region to induce direct association with KLC. Replacement of these serines with Ala results in a mutant that is unable to bind kinesin-1, which impairs exit of Alcα cargo from the Golgi. Despite this deficiency, the compromised Alcα mutant was still transported, albeit improperly by vesicles following missorting of the Alcα mutant with amyloid β-protein precursor (APP) cargo. This suggests that APP partially compensates for defective Alcα in anterograde transport by providing an alternative cargo receptor for kinesin-1.
Collapse
Affiliation(s)
- Yuriko Sobu
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Keiko Furukori
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan.,Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06508
| | - Kyoko Chiba
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Angus C Nairn
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06508
| | - Masataka Kinjo
- Laboratory of Molecular Cell Dynamics, Faculty of Advanced Life Science, Hokkaido University, Sapporo, 001-0021, Japan
| | - Saori Hata
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Toshiharu Suzuki
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| |
Collapse
|
18
|
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: 51] [Impact Index Per Article: 7.3] [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
|
19
|
de Ramon Francàs G, Alther T, Stoeckli ET. Calsyntenins Are Expressed in a Dynamic and Partially Overlapping Manner during Neural Development. Front Neuroanat 2017; 11:76. [PMID: 28912692 PMCID: PMC5582071 DOI: 10.3389/fnana.2017.00076] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 08/17/2017] [Indexed: 01/26/2023] Open
Abstract
Calsyntenins form a family of linker proteins between distinct populations of vesicles and kinesin motors for axonal transport. They were implicated in synapse formation and synaptic plasticity by findings in worms, mice and humans. These findings were in accordance with the postsynaptic localization of the Calsyntenins in the adult brain. However, they also affect the formation of neural circuits, as loss of Calsyntenin-1 (Clstn1) was shown to interfere with axonal branching and axon guidance. Despite the fact that Calsyntenins were discovered originally in embryonic chicken motoneurons, their distribution in the developing nervous system has not been analyzed in detail so far. Here, we summarize our analysis of the temporal and spatial expression patterns of the cargo-docking proteins Clstn1, Clstn2 and Clstn3 during neural development by comparing the dynamic distribution of their mRNAs by in situ hybridization in the spinal cord, the cerebellum, the retina and the tectum, as well as in the dorsal root ganglia (DRG).
Collapse
Affiliation(s)
- Gemma de Ramon Francàs
- Department of Molecular Life Sciences and Neuroscience Center Zurich, University of ZurichZurich, Switzerland
| | - Tobias Alther
- Department of Molecular Life Sciences and Neuroscience Center Zurich, University of ZurichZurich, Switzerland
| | - Esther T Stoeckli
- Department of Molecular Life Sciences and Neuroscience Center Zurich, University of ZurichZurich, Switzerland
| |
Collapse
|
20
|
Bettler B, Fakler B. Ionotropic AMPA-type glutamate and metabotropic GABAB receptors: determining cellular physiology by proteomes. Curr Opin Neurobiol 2017; 45:16-23. [DOI: 10.1016/j.conb.2017.02.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Accepted: 02/15/2017] [Indexed: 02/07/2023]
|
21
|
Pin JP, Bettler B. Organization and functions of mGlu and GABAB receptor complexes. Nature 2016; 540:60-68. [DOI: 10.1038/nature20566] [Citation(s) in RCA: 155] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 10/21/2016] [Indexed: 02/08/2023]
|
22
|
Awan Z, Tay ESE, Eyre NS, Wu LE, Beard MR, Boo I, Drummer HE, George J, Douglas MW. Calsyntenin-1 mediates hepatitis C virus replication. J Gen Virol 2016; 97:1877-1887. [PMID: 27221318 DOI: 10.1099/jgv.0.000511] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The hepatitis C virus (HCV) RNA genome of 9.6 kb encodes only 10 proteins, and so is highly dependent on host hepatocyte factors to facilitate replication. We aimed to identify host factors involved in the egress of viral particles. By screening the supernatant of HCV-infected Huh7 cells using SILAC-based proteomics, we identified the transmembrane protein calsyntenin-1 as a factor specifically secreted by infected cells. Calsyntenin-1 has previously been shown to mediate transport of endosomes along microtubules in neurons, through interactions with kinesin light chain-1. Here we demonstrate for the first time, we believe, a similar role for calsyntenin-1 in Huh7 cells, mediating intracellular transport of endosomes. In HCV-infected cells we show that calsyntenin-1 contributes to the early stages of the viral replication cycle and the formation of the replication complex. Importantly, we demonstrate in our model that silencing calsyntenin-1 disrupts the viral replication cycle, confirming the reliance of HCV on this protein as a host factor. Characterizing the function of calsyntenin-1 will increase our understanding of the HCV replication cycle and pathogenesis, with potential application to other viruses sharing common pathways.
Collapse
Affiliation(s)
- Zunaira Awan
- Storr Liver Centre, The Westmead Millennium Institute for Medical Research, The University of Sydney at Westmead Hospital, 176 Hawkesbury Rd, Westmead NSW 2145, Australia
| | - Enoch S E Tay
- Storr Liver Centre, The Westmead Millennium Institute for Medical Research, The University of Sydney at Westmead Hospital, 176 Hawkesbury Rd, Westmead NSW 2145, Australia
| | - Nicholas S Eyre
- Hepatitis C Virus Research Laboratory, School of Molecular and Biomedical Science, University of Adelaide, Adelaide, Australia
| | - Lindsay E Wu
- University of New South Wales, Sydney NSW 2052, Australia
| | - Michael R Beard
- Hepatitis C Virus Research Laboratory, School of Molecular and Biomedical Science, University of Adelaide, Adelaide, Australia
| | - Irene Boo
- Centre for Biomedical Research, Burnet Institute, 85 Commercial Rd, Melbourne VIC 3004, Australia
| | - Heidi E Drummer
- Centre for Biomedical Research, Burnet Institute, 85 Commercial Rd, Melbourne VIC 3004, Australia.,Department of Microbiology, 19 Innovation Walk, Monash University, Victoria, Australia.,Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Jacob George
- Storr Liver Centre, The Westmead Millennium Institute for Medical Research, The University of Sydney at Westmead Hospital, 176 Hawkesbury Rd, Westmead NSW 2145, Australia
| | - Mark W Douglas
- Storr Liver Centre, The Westmead Millennium Institute for Medical Research, The University of Sydney at Westmead Hospital, 176 Hawkesbury Rd, Westmead NSW 2145, Australia.,Centre for Infectious Diseases and Microbiology, Marie Bashir Institute for Infectious Diseases and Biosecurity, University of Sydney at Westmead Hospital, Westmead NSW 2145, Australia
| |
Collapse
|
23
|
D Potdar P, U Shetti A. Molecular Biomarkers for Diagnosis & Therapies of Alzheimer’s Disease. AIMS Neurosci 2016. [DOI: 10.3934/neuroscience.2016.4.433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
24
|
Abstract
β-Amyloid is generated by the sequential cleavage of amyloid precursor protein. Calsyntenin-1 and kinesin light chain-1 splice variant E (KLC1-E) have been proposed to regulate β-amyloid production from amyloid precursor protein. Vesicles containing calsyntenin-1 are transported from the Golgi apparatus to axons by interaction between calsyntenin-1 and KLC1 in their C-terminal regions. However, it is unclear whether KLC1 isoform E influences the interaction between KLC1 and calsyntenin-1, resulting in the impaired axonal transport of calsyntenin-1 vesicles. Here, we show that KLC1-E does not interact with calsyntenin-1 using a pull-down assay, coimmunoprecipitation, and immunocytochemistry. These findings suggest that KLC1-E enrichment may impair the axonal transport of calsyntenin-1 vesicles.
Collapse
|
25
|
Ortiz-Medina H, Emond MR, Jontes JD. Zebrafish calsyntenins mediate homophilic adhesion through their amino-terminal cadherin repeats. Neuroscience 2014; 286:87-96. [PMID: 25463516 DOI: 10.1016/j.neuroscience.2014.11.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 11/07/2014] [Accepted: 11/18/2014] [Indexed: 11/20/2022]
Abstract
The calsyntenins are atypical members of the cadherin superfamily that have been implicated in learning in Caenorhabditis elegans and memory formation in humans. As members of the cadherin superfamily, they could mediate cell-cell adhesion, although their adhesive properties have not been investigated. As an initial step in characterizing the calsyntenins, we have cloned clstn1, clstn2 and clstn3 from the zebrafish and determined their expression in the developing zebrafish nervous system. The three genes each have broad, yet distinct, expression patterns in the zebrafish brain. Each of the ectodomains mediates homophilic interactions through two, amino-terminal cadherin repeats. In bead sorting assays, the calsyntenin ectodomains do not exhibit homophilic preferences. These data support the idea that calsyntenins could either act as adhesion molecules or as diffusible, homophilic or heterophilic ligands in the vertebrate nervous system.
Collapse
Affiliation(s)
- H Ortiz-Medina
- Department of Neuroscience, Ohio State University Medical Center, United States
| | - M R Emond
- Department of Neuroscience, Ohio State University Medical Center, United States
| | - J D Jontes
- Department of Neuroscience, Ohio State University Medical Center, United States.
| |
Collapse
|
26
|
Takei N, Sobu Y, Kimura A, Urano S, Piao Y, Araki Y, Taru H, Yamamoto T, Hata S, Nakaya T, Suzuki T. Cytoplasmic fragment of Alcadein α generated by regulated intramembrane proteolysis enhances amyloid β-protein precursor (APP) transport into the late secretory pathway and facilitates APP cleavage. J Biol Chem 2014; 290:987-95. [PMID: 25406318 DOI: 10.1074/jbc.m114.599852] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The neural type I membrane protein Alcadein α (Alcα), is primarily cleaved by amyloid β-protein precursor (APP) α-secretase to generate a membrane-associated carboxyl-terminal fragment (Alcα CTF), which is further cleaved by γ-secretase to secrete p3-Alcα peptides and generate an intracellular cytoplasmic domain fragment (Alcα ICD) in the late secretory pathway. By association with the neural adaptor protein X11L (X11-like), Alcα and APP form a ternary complex that suppresses the cleavage of both Alcα and APP by regulating the transport of these membrane proteins into the late secretory pathway where secretases are active. However, it has not been revealed how Alcα and APP are directed from the ternary complex formed largely in the Golgi into the late secretory pathway to reach a nerve terminus. Using a novel transgenic mouse line expressing excess amounts of human Alcα CTF (hAlcα CTF) in neurons, we found that expression of hAlcα CTF induced excess production of hAlcα ICD, which facilitated APP transport into the nerve terminus and enhanced APP metabolism, including Aβ generation. In vitro cell studies also demonstrated that excess expression of Alcα ICD released both APP and Alcα from the ternary complex. These results indicate that regulated intramembrane proteolysis of Alcα by γ-secretase regulates APP trafficking and the production of Aβ in vivo.
Collapse
Affiliation(s)
- Norio Takei
- From the Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita-12 Nishi-6, Kita-ku, Sapporo 060-0812, Japan and
| | - Yuriko Sobu
- From the Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita-12 Nishi-6, Kita-ku, Sapporo 060-0812, Japan and
| | - Ayano Kimura
- From the Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita-12 Nishi-6, Kita-ku, Sapporo 060-0812, Japan and
| | - Satomi Urano
- From the Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita-12 Nishi-6, Kita-ku, Sapporo 060-0812, Japan and
| | - Yi Piao
- From the Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita-12 Nishi-6, Kita-ku, Sapporo 060-0812, Japan and
| | - Yoichi Araki
- From the Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita-12 Nishi-6, Kita-ku, Sapporo 060-0812, Japan and
| | - Hidenori Taru
- From the Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita-12 Nishi-6, Kita-ku, Sapporo 060-0812, Japan and
| | - Tohru Yamamoto
- Department of Molecular Neurobiology, Faculty of Medicine, Kagawa University, Miki-cho 761-0793, Japan
| | - Saori Hata
- From the Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita-12 Nishi-6, Kita-ku, Sapporo 060-0812, Japan and
| | - Tadashi Nakaya
- From the Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita-12 Nishi-6, Kita-ku, Sapporo 060-0812, Japan and
| | - Toshiharu Suzuki
- From the Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita-12 Nishi-6, Kita-ku, Sapporo 060-0812, Japan and
| |
Collapse
|
27
|
Gan KJ, Morihara T, Silverman MA. Atlas stumbled: Kinesin light chain-1 variant E triggers a vicious cycle of axonal transport disruption and amyloid-β generation in Alzheimer's disease. Bioessays 2014; 37:131-41. [DOI: 10.1002/bies.201400131] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- Kathlyn J. Gan
- Department of Molecular Biology and Biochemistry; Simon Fraser University; Burnaby BC Canada
| | - Takashi Morihara
- Department of Psychiatry; Graduate School of Medicine; Osaka University; Osaka Japan
| | - Michael A. Silverman
- Department of Molecular Biology and Biochemistry; Simon Fraser University; Burnaby BC Canada
- Department of Biological Sciences; Simon Fraser University; Burnaby BC Canada
- Brain Research Centre; University of British Columbia; Vancouver BC Canada
| |
Collapse
|
28
|
Calsyntenin-1 regulates axon branching and endosomal trafficking during sensory neuron development in vivo. J Neurosci 2014; 34:9235-48. [PMID: 25009257 DOI: 10.1523/jneurosci.0561-14.2014] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Precise regulation of axon branching is crucial for neuronal circuit formation, yet the mechanisms that control branch formation are not well understood. Moreover, the highly complex morphology of neurons makes them critically dependent on protein/membrane trafficking and transport systems, although the functions for membrane trafficking in neuronal morphogenesis are largely undefined. Here we identify a kinesin adaptor, Calsyntenin-1 (Clstn-1), as an essential regulator of axon branching and neuronal compartmentalization in vivo. We use morpholino knockdown and a Clstn-1 mutant to show that Clstn-1 is required for formation of peripheral but not central sensory axons, and for peripheral axon branching in zebrafish. We used live imaging of endosomal trafficking in vivo to show that Clstn-1 regulates transport of Rab5-containing endosomes from the cell body to specific locations of developing axons. Our results suggest a model in which Clstn-1 patterns separate axonal compartments and define their ability to branch by directing trafficking of specific endosomes.
Collapse
|
29
|
Calsyntenin-1 regulates targeting of dendritic NMDA receptors and dendritic spine maturation in CA1 hippocampal pyramidal cells during postnatal development. J Neurosci 2014; 34:8716-27. [PMID: 24966372 DOI: 10.1523/jneurosci.0144-14.2014] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Calsyntenin-1 is a transmembrane cargo-docking protein important for kinesin-1-mediated fast transport of membrane-bound organelles that exhibits peak expression levels at postnatal day 7. However, its neuronal function during postnatal development remains unknown. We generated a knock-out mouse to characterize calsyntenin-1 function in juvenile mice. In the absence of calsyntenin-1, synaptic transmission was depressed. To address the mechanism, evoked EPSPs were analyzed revealing a greater proportion of synaptic GluN2B subunit-containing receptors typical for less mature synapses. This imbalance was due to a disruption in calsyntenin-1-mediated dendritic transport of NMDA receptor subunits. As a consequence of increased expression of GluN2B subunits, NMDA receptor-dependent LTP was enhanced at Schaffer collateral-CA1 pyramidal cell synapses. Interestingly, these defects were accompanied by a decrease in dendritic arborization and increased proportions of immature filopodia-like dendritic protrusions at the expense of thin-type dendritic spines in CA1 pyramidal cells. Thus, these results highlight a key role for calsyntenin-1 in the transport of NMDA receptors to synaptic targets, which is necessary for the maturation of neuronal circuits during early development.
Collapse
|
30
|
Tam JHK, Seah C, Pasternak SH. The Amyloid Precursor Protein is rapidly transported from the Golgi apparatus to the lysosome and where it is processed into beta-amyloid. Mol Brain 2014; 7:54. [PMID: 25085554 PMCID: PMC4237969 DOI: 10.1186/s13041-014-0054-1] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 07/23/2014] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is characterized by cerebral deposition of β-amyloid peptide (Aβ). Aβ is produced by sequential cleavage of the Amyloid Precursor Protein (APP) by β- and γ-secretases. Many studies have demonstrated that the internalization of APP from the cell surface can regulate Aβ production, although the exact organelle in which Aβ is produced remains contentious. A number of recent studies suggest that intracellular trafficking also plays a role in regulating Aβ production, but these pathways are relatively under-studied. The goal of this study was to elucidate the intracellular trafficking of APP, and to examine the site of intracellular APP processing. RESULTS We have tagged APP on its C-terminal cytoplasmic tail with photoactivatable Green Fluorescent Protein (paGFP). By photoactivating APP-paGFP in the Golgi, using the Golgi marker Galactosyltranferase fused to Cyan Fluorescent Protein (GalT-CFP) as a target, we are able to follow a population of nascent APP molecules from the Golgi to downstream compartments identified with compartment markers tagged with red fluorescent protein (mRFP or mCherry); including rab5 (early endosomes) rab9 (late endosomes) and LAMP1 (lysosomes). Because γ-cleavage of APP releases the cytoplasmic tail of APP including the photoactivated GFP, resulting in loss of fluorescence, we are able to visualize the cleavage of APP in these compartments. Using APP-paGFP, we show that APP is rapidly trafficked from the Golgi apparatus to the lysosome; where it is rapidly cleared. Chloroquine and the highly selective γ-secretase inhibitor, L685, 458, cause the accumulation of APP in lysosomes implying that APP is being cleaved by secretases in the lysosome. The Swedish mutation dramatically increases the rate of lysosomal APP processing, which is also inhibited by chloroquine and L685, 458. By knocking down adaptor protein 3 (AP-3; a heterotetrameric protein complex required for trafficking many proteins to the lysosome) using siRNA, we are able to reduce this lysosomal transport. Blocking lysosomal transport of APP reduces Aβ production by more than a third. CONCLUSION These data suggests that AP-3 mediates rapid delivery of APP to lysosomes, and that the lysosome is a likely site of Aβ production.
Collapse
Affiliation(s)
- Joshua HK Tam
- J. Allyn Taylor Centre for Cell Biology, Molecular Brain Research Group, Robarts Research Institute, 100 Perth Drive, London N6A 5K8, Ontario, Canada
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, the University of Western Ontario, London N6A 5K8, Ontario, Canada
| | - Claudia Seah
- J. Allyn Taylor Centre for Cell Biology, Molecular Brain Research Group, Robarts Research Institute, 100 Perth Drive, London N6A 5K8, Ontario, Canada
| | - Stephen H Pasternak
- J. Allyn Taylor Centre for Cell Biology, Molecular Brain Research Group, Robarts Research Institute, 100 Perth Drive, London N6A 5K8, Ontario, Canada
- Department of Clinical Neurological Sciences, London N6A 5K8, Ontario, Canada
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, the University of Western Ontario, London N6A 5K8, Ontario, Canada
| |
Collapse
|
31
|
Ohno H, Kato S, Naito Y, Kunitomo H, Tomioka M, Iino Y. Role of synaptic phosphatidylinositol 3-kinase in a behavioral learning response in C. elegans. Science 2014; 345:313-7. [DOI: 10.1126/science.1250709] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
32
|
UV irradiation accelerates amyloid precursor protein (APP) processing and disrupts APP axonal transport. J Neurosci 2014; 34:3320-39. [PMID: 24573290 DOI: 10.1523/jneurosci.1503-13.2014] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Overexpression and/or abnormal cleavage of amyloid precursor protein (APP) are linked to Alzheimer's disease (AD) development and progression. However, the molecular mechanisms regulating cellular levels of APP or its processing, and the physiological and pathological consequences of altered processing are not well understood. Here, using mouse and human cells, we found that neuronal damage induced by UV irradiation leads to specific APP, APLP1, and APLP2 decline by accelerating their secretase-dependent processing. Pharmacological inhibition of endosomal/lysosomal activity partially protects UV-induced APP processing implying contribution of the endosomal and/or lysosomal compartments in this process. We found that a biological consequence of UV-induced γ-secretase processing of APP is impairment of APP axonal transport. To probe the functional consequences of impaired APP axonal transport, we isolated and analyzed presumptive APP-containing axonal transport vesicles from mouse cortical synaptosomes using electron microscopy, biochemical, and mass spectrometry analyses. We identified a population of morphologically heterogeneous organelles that contains APP, the secretase machinery, molecular motors, and previously proposed and new residents of APP vesicles. These possible cargoes are enriched in proteins whose dysfunction could contribute to neuronal malfunction and diseases of the nervous system including AD. Together, these results suggest that damage-induced APP processing might impair APP axonal transport, which could result in failure of synaptic maintenance and neuronal dysfunction.
Collapse
|
33
|
Wang J, Shan C, Cao W, Zhang C, Teng J, Chen J. SCG10 promotes non-amyloidogenic processing of amyloid precursor protein by facilitating its trafficking to the cell surface. Hum Mol Genet 2013; 22:4888-900. [DOI: 10.1093/hmg/ddt339] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
|
34
|
Vagnoni A, Glennon EBC, Perkinton MS, Gray EH, Noble W, Miller CCJ. Loss of c-Jun N-terminal kinase-interacting protein-1 does not affect axonal transport of the amyloid precursor protein or Aβ production. Hum Mol Genet 2013; 22:4646-52. [PMID: 23825109 PMCID: PMC3889811 DOI: 10.1093/hmg/ddt313] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Disruption to axonal transport is an early pathological feature in Alzheimer's disease. The amyloid precursor protein (APP) is a key axonal transport cargo in Alzheimer's disease since perturbation of its transport increases APP processing and production of amyloid-β peptide (Aβ) that is deposited in the brains of Alzheimer's disease patients. APP is transported anterogradely through axons on kinesin-1 motors. One favoured route for attachment of APP to kinesin-1 involves the scaffolding protein c-Jun N-terminal kinase-interacting protein-1 (JIP1), which has been shown to bind both APP and kinesin-1 light chain (KLC). However, direct experimental evidence to support a role of JIP1 in APP transport is lacking. Notably, the effect of loss of JIP1 on movement of APP through axons of living neurons, and the impact of such loss on APP processing and Aβ production has not been reported. To address these issues, we monitored how siRNA mediated loss of JIP1 influenced transport of enhanced green fluorescent protein (EGFP)-tagged APP through axons and production of endogenous Aβ in living neurons. Surprisingly, we found that knockdown of JIP1 did not affect either APP transport or Aβ production. These results have important implications for our understanding of APP trafficking in Alzheimer's disease.
Collapse
Affiliation(s)
- Alessio Vagnoni
- Departments of Neuroscience and Clinical Neurosciences, Institute of Psychiatry, King's College London, London, UK
| | | | | | | | | | | |
Collapse
|
35
|
Thouvenot E, Urbach S, Vigy O, Séveno M, Galéotti N, Nguyen G, Bockaert J, Marin P. Quantitative proteomic analysis reveals protein expression changes in the murine neuronal secretome during apoptosis. J Proteomics 2012; 77:394-405. [DOI: 10.1016/j.jprot.2012.09.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Revised: 08/24/2012] [Accepted: 09/12/2012] [Indexed: 01/08/2023]
|
36
|
Abstract
Cadherins are Ca(2+)-dependent cell-cell adhesion molecules that play critical roles in animal morphogenesis. Various cadherin-related molecules have also been identified, which show diverse functions, not only for the regulation of cell adhesion but also for that of cell proliferation and planar cell polarity. During the past decade, understanding of the roles of these molecules in the nervous system has significantly progressed. They are important not only for the development of the nervous system but also for its functions and, in turn, for neural disorders. In this review, we discuss the roles of cadherins and related molecules in neural development and function in the vertebrate brain.
Collapse
Affiliation(s)
- Shinji Hirano
- Department of Neurobiology and Anatomy, Kochi Medical School, Okoh-cho Kohasu, Nankoku-City 783–8505, Japan.
| | | |
Collapse
|
37
|
Steuble M, Diep TM, Schätzle P, Ludwig A, Tagaya M, Kunz B, Sonderegger P. Calsyntenin-1 shelters APP from proteolytic processing during anterograde axonal transport. Biol Open 2012; 1:761-74. [PMID: 23213470 PMCID: PMC3507217 DOI: 10.1242/bio.20121578] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Accepted: 05/22/2012] [Indexed: 11/27/2022] Open
Abstract
Endocytosis of amyloid-β precursor protein (APP) is thought to represent the major source of substrate for the production of the amyloidogenic Aβ peptide by the β-secretase BACE1. The irreversible nature of proteolytic cleavage implies the existence of an efficient replenishment route for APP from its sites of synthesis to the cell surface. We recently found that APP exits the trans-Golgi network in intimate association with calsyntenin-1, a transmembrane cargo-docking protein for Kinesin-1-mediated vesicular transport. Here we characterized the function of calsyntenin-1 in neuronal APP transport using selective immunoisolation of intracellular trafficking organelles, immunocytochemistry, live-imaging, and RNAi. We found that APP is co-transported with calsyntenin-1 along axons to early endosomes in the central region of growth cones in carriers that exclude the α-secretase ADAM10. Intriguingly, calsyntenin-1/APP organelles contained BACE1, suggesting premature cleavage of APP along its anterograde path. However, we found that APP contained in calsyntenin-1/APP organelles was stable. We further analyzed vesicular trafficking of APP in cultured hippocampal neurons, in which calsyntenin-1 was reduced by RNAi. We found a markedly increased co-localization of APP and ADAM10 in axons and growth cones, along with increased proteolytic processing of APP and Aβ secretion in these neurons. This suggested that the reduced capacity for calsyntenin-1-dependent APP transport resulted in mis-sorting of APP into additional axonal carriers and, therefore, the premature encounter of unprotected APP with its ectodomain proteases. In combination, our results characterize calsyntenin-1/APP organelles as carriers for sheltered anterograde axonal transport of APP.
Collapse
Affiliation(s)
- Martin Steuble
- Department of Biochemistry, University of Zurich , Winterthurerstrasse 190, CH-8057 Zurich , Switzerland
| | | | | | | | | | | | | |
Collapse
|
38
|
Claeysen S, Cochet M, Donneger R, Dumuis A, Bockaert J, Giannoni P. Alzheimer culprits: cellular crossroads and interplay. Cell Signal 2012; 24:1831-40. [PMID: 22627093 DOI: 10.1016/j.cellsig.2012.05.008] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2012] [Accepted: 05/09/2012] [Indexed: 12/22/2022]
Abstract
Alzheimer's disease (AD) is the primary cause of dementia in the elderly and one of the major health problems worldwide. Since its first description by Alois Alzheimer in 1907, noticeable but insufficient scientific comprehension of this complex pathology has been achieved. All the research that has been pursued takes origin from the identification of the pathological hallmarks in the forms of amyloid-β (Aβ) deposits (plaques), and aggregated hyperphosphorylated tau protein filaments (named neurofibrillary tangles). Since this discovery, many hypotheses have been proposed to explain the origin of the pathology. The "amyloid cascade hypothesis" is the most accredited theory. The mechanism suggested to be one of the initial causes of AD is an imbalance between the production and the clearance of Aβ peptides. Therefore, Amyloid Precursor Protein (APP) synthesis, trafficking and metabolism producing either the toxic Aβ peptide via the amyloidogenic pathway or the sAPPα fragment via the non amyloidogenic pathway have become appealing subjects of study. Being able to reduce the formation of the toxic Aβ peptides is obviously an immediate approach in the trial to prevent AD. The following review summarizes the most relevant discoveries in the field of the last decades.
Collapse
Affiliation(s)
- Sylvie Claeysen
- CNRS, UMR-5203, Institut de Génomique Fonctionnelle, F-34000 Montpellier, France.
| | | | | | | | | | | |
Collapse
|
39
|
Kawano T, Araseki M, Araki Y, Kinjo M, Yamamoto T, Suzuki T. A Small Peptide Sequence is Sufficient for Initiating Kinesin-1 Activation Through Part of TPR Region of KLC1. Traffic 2012; 13:834-48. [DOI: 10.1111/j.1600-0854.2012.01350.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2011] [Revised: 03/05/2012] [Accepted: 03/08/2012] [Indexed: 12/18/2022]
Affiliation(s)
- Takanori Kawano
- Laboratory of Neuroscience; Graduate School of Pharmaceutical Sciences, Hokkaido University; Sapporo; 060-0812; Japan
| | - Masahiko Araseki
- Laboratory of Neuroscience; Graduate School of Pharmaceutical Sciences, Hokkaido University; Sapporo; 060-0812; Japan
| | - Yoichi Araki
- Laboratory of Neuroscience; Graduate School of Pharmaceutical Sciences, Hokkaido University; Sapporo; 060-0812; Japan
| | - Masataka Kinjo
- Laboratory of Molecular Cell Dynamics, Faculty of Advanced Life Science; Hokkaido University; Sapporo; 001-0021; Japan
| | - Tohru Yamamoto
- Laboratory of Neuroscience; Graduate School of Pharmaceutical Sciences, Hokkaido University; Sapporo; 060-0812; Japan
| | - Toshiharu Suzuki
- Laboratory of Neuroscience; Graduate School of Pharmaceutical Sciences, Hokkaido University; Sapporo; 060-0812; Japan
| |
Collapse
|
40
|
Vagnoni A, Perkinton MS, Gray EH, Francis PT, Noble W, Miller CCJ. Calsyntenin-1 mediates axonal transport of the amyloid precursor protein and regulates Aβ production. Hum Mol Genet 2012; 21:2845-54. [PMID: 22434822 PMCID: PMC3373235 DOI: 10.1093/hmg/dds109] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Understanding the mechanisms that control processing of the amyloid precursor protein (APP) to produce amyloid-β (Aβ) peptide represents a key area of Alzheimer's disease research. Here, we show that siRNA-mediated loss of calsyntenin-1 in cultured neurons alters APP processing to increase production of Aβ. We also show that calsyntenin-1 is reduced in Alzheimer's disease brains and that the extent of this reduction correlates with increased Aβ levels. Calsyntenin-1 is a ligand for kinesin-1 light chains and APP is transported through axons on kinesin-1 molecular motors. Defects in axonal transport are an early pathological feature in Alzheimer's disease and defective APP transport is known to increase Aβ production. We show that calsyntenin-1 and APP are co-transported through axons and that siRNA-induced loss of calsyntenin-1 markedly disrupts axonal transport of APP. Thus, perturbation to axonal transport of APP on calsyntenin-1 containing carriers induces alterations to APP processing that increase production of Aβ. Together, our findings suggest that disruption of calsyntenin-1-associated axonal transport of APP is a pathogenic mechanism in Alzheimer's disease.
Collapse
Affiliation(s)
- Alessio Vagnoni
- KCL Centre for Neurodegeneration Research, Department of Neuroscience, Institute of Psychiatry, King’s College London, De Crespigny Park, London, UK
| | | | | | | | | | | |
Collapse
|
41
|
Ringman JM, Schulman H, Becker C, Jones T, Bai Y, Immermann F, Cole G, Sokolow S, Gylys K, Geschwind DH, Cummings JL, Wan HI. Proteomic changes in cerebrospinal fluid of presymptomatic and affected persons carrying familial Alzheimer disease mutations. ACTA ACUST UNITED AC 2012; 69:96-104. [PMID: 22232349 DOI: 10.1001/archneurol.2011.642] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
OBJECTIVE To identify cerebrospinal fluid (CSF) protein changes in persons who will develop familial Alzheimer disease (FAD) due to PSEN1 and APP mutations, using unbiased proteomics. DESIGN We compared proteomic profiles of CSF from individuals with FAD who were mutation carriers (MCs) and related noncarriers (NCs). Abundant proteins were depleted and samples were analyzed using liquid chromatography-electrospray ionization-mass spectrometry on a high-resolution time-of-flight instrument. Tryptic peptides were identified by tandem mass spectrometry. Proteins differing in concentration between the MCs and NCs were identified. SETTING A tertiary dementia referral center and a proteomic biomarker discovery laboratory. PARTICIPANTS Fourteen FAD MCs (mean age, 34.2 years; 10 are asymptomatic, 12 have presenilin-1 [PSEN1 ] gene mutations, and 2 have amyloid precursor protein [APP ] gene mutations) and 5 related NCs (mean age, 37.6 years). RESULTS Fifty-six proteins were identified, represented by multiple tryptic peptides showing significant differences between MCs and NCs (46 upregulated and 10 downregulated); 40 of these proteins differed when the analysis was restricted to asymptomatic individuals. Fourteen proteins have been reported in prior proteomic studies in late-onset AD, including amyloid precursor protein, transferrin, α(1)β-glycoprotein, complement components, afamin precursor, spondin 1, plasminogen, hemopexin, and neuronal pentraxin receptor. Many other proteins were unique to our study, including calsyntenin 3, AMPA (α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid) 4 glutamate receptor, CD99 antigen, di- N-acetyl-chitobiase, and secreted phosphoprotein 1. CONCLUSIONS We found much overlap in CSF protein changes between individuals with presymptomatic and symptomatic FAD and those with late-onset AD. Our results are consistent with inflammation and synaptic loss early in FAD and suggest new presymptomatic biomarkers of potential usefulness in drug development.
Collapse
Affiliation(s)
- John M Ringman
- Mary S. Easton Center for Alzheimer's Disease Research, 10911 Weyburn Ave, Ste 200, Los Angeles, CA 90095, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Exit from the trans-Golgi network: from molecules to mechanisms. Curr Opin Cell Biol 2011; 23:443-51. [PMID: 21550789 DOI: 10.1016/j.ceb.2011.03.013] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2011] [Revised: 03/07/2011] [Accepted: 03/22/2011] [Indexed: 11/23/2022]
Abstract
The trans-Golgi network is a major sorting platform of the secretory pathway from which proteins and lipids, both newly synthesized and retrieved from endocytic compartments, are targeted to different destinations. These sorting processes occur during the formation of pleomorphic tubular-vesicular carriers. The past years have provided insights into basic mechanisms coordinating the spatial and temporal organization of machineries necessary for the segregation of membrane components into distinct microdomains, for the bending, elongation, and fission of corresponding membranes, thus revealing a complex interplay of protein-protein and protein-lipid interactions.
Collapse
|
43
|
Vagnoni A, Rodriguez L, Manser C, De Vos KJ, Miller CCJ. Phosphorylation of kinesin light chain 1 at serine 460 modulates binding and trafficking of calsyntenin-1. J Cell Sci 2011; 124:1032-42. [PMID: 21385839 DOI: 10.1242/jcs.075168] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Kinesin light chain 1 (KLC1) binds to the intracellular cytoplasmic domain of the type-1 membrane-spanning protein calsyntenin-1 (also known as alcadein-α) to mediate transport of a subset of vesicles. Here, we identify serine 460 in KLC1 (KLC1ser460) as a phosphorylation site and show that mutation of KLC1ser460 influences the binding of KLC1 to calsyntenin-1. Mutation of KLC1ser460 to an alanine residue, to preclude phosphorylation, increased the binding of calsyntenin-1, whereas mutation to an aspartate residue, to mimic permanent phosphorylation, reduced the binding. Mutation of KLC1ser460 did not affect the interaction of KLC1 with four other known binding partners: huntingtin-associated protein 1 isoform A (HAP1A), collapsin response mediator protein-2 (CRMP2), c-Jun N-terminal kinase-interacting protein-1 (JIP1) and kinase-D-interacting substrate of 220 kDa (Kidins220). KLC1ser460 is a predicted mitogen-activated protein kinase (MAPK) target site, and we show that extracellular-signal-regulated kinase (ERK) phosphorylates this residue in vitro. We also demonstrate that inhibition of ERK promotes binding of calsyntenin-1 to KLC1. Finally, we show that expression of the KLC1ser460 mutant proteins influences calsyntenin-1 distribution and transport in cultured cells. Thus, phosphorylation of KLC1ser460 represents a mechanism for selectively regulating the binding and trafficking of calsyntenin-1.
Collapse
Affiliation(s)
- Alessio Vagnoni
- MRC Centre for Neurodegeneration Research, Institute of Psychiatry, King's College London, PO Box 37, De Crespigny Park, Denmark Hill, London SE5 8AF, UK
| | | | | | | | | |
Collapse
|
44
|
Steuble M, Gerrits B, Ludwig A, Mateos JM, Diep TM, Tagaya M, Stephan A, Schätzle P, Kunz B, Streit P, Sonderegger P. Molecular characterization of a trafficking organelle: dissecting the axonal paths of calsyntenin-1 transport vesicles. Proteomics 2011; 10:3775-88. [PMID: 20925061 DOI: 10.1002/pmic.201000384] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Kinesin motors play crucial roles in the delivery of membranous cargo to its destination and thus for the establishment and maintenance of cellular polarization. Recently, calsyntenin-1 was identified as a cargo-docking protein for Kinesin-1-mediated axonal transport of tubulovesicular organelles along axons of central nervous system neurons. To further define the function of calsyntenin-1, we immunoisolated calsyntenin-1 organelles from murine brain homogenates and determined their proteome by MS. We found that calsyntenin-1 organelles are endowed with components of the endosomal trafficking machinery and contained the β-amyloid precursor protein (APP). Detailed biochemical analyses of calsyntenin-1 immunoisolates in conjunction with immunocytochemical colocalization studies with cultured hippocampal neurons, using endosomal marker proteins for distinct subcompartments of the endosomal pathways, indicated that neuronal axons contain at least two distinct, nonoverlapping calsyntenin-1-containing transport packages: one characterized as early-endosomal, APP positive, the other as recycling-endosomal, APP negative. We postulate that calsyntenin-1 acts as a general mediator of anterograde axonal transportation of endosomal vesicles. In this role, calsyntenin-1 may actively contribute to axonal growth and pathfinding in the developing as well as to the maintenance of neuronal polarity in the adult nervous system; further, it may actively contribute to the stabilization of APP during its anterograde axonal trajectory.
Collapse
Affiliation(s)
- Martin Steuble
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Perrin RJ, Craig-Schapiro R, Malone JP, Shah AR, Gilmore P, Davis AE, Roe CM, Peskind ER, Li G, Galasko DR, Clark CM, Quinn JF, Kaye JA, Morris JC, Holtzman DM, Townsend RR, Fagan AM. Identification and validation of novel cerebrospinal fluid biomarkers for staging early Alzheimer's disease. PLoS One 2011; 6:e16032. [PMID: 21264269 PMCID: PMC3020224 DOI: 10.1371/journal.pone.0016032] [Citation(s) in RCA: 128] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2010] [Accepted: 12/03/2010] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Ideally, disease modifying therapies for Alzheimer disease (AD) will be applied during the 'preclinical' stage (pathology present with cognition intact) before severe neuronal damage occurs, or upon recognizing very mild cognitive impairment. Developing and judiciously administering such therapies will require biomarker panels to identify early AD pathology, classify disease stage, monitor pathological progression, and predict cognitive decline. To discover such biomarkers, we measured AD-associated changes in the cerebrospinal fluid (CSF) proteome. METHODS AND FINDINGS CSF samples from individuals with mild AD (Clinical Dementia Rating [CDR] 1) (n = 24) and cognitively normal controls (CDR 0) (n = 24) were subjected to two-dimensional difference-in-gel electrophoresis. Within 119 differentially-abundant gel features, mass spectrometry (LC-MS/MS) identified 47 proteins. For validation, eleven proteins were re-evaluated by enzyme-linked immunosorbent assays (ELISA). Six of these assays (NrCAM, YKL-40, chromogranin A, carnosinase I, transthyretin, cystatin C) distinguished CDR 1 and CDR 0 groups and were subsequently applied (with tau, p-tau181 and Aβ42 ELISAs) to a larger independent cohort (n = 292) that included individuals with very mild dementia (CDR 0.5). Receiver-operating characteristic curve analyses using stepwise logistic regression yielded optimal biomarker combinations to distinguish CDR 0 from CDR>0 (tau, YKL-40, NrCAM) and CDR 1 from CDR<1 (tau, chromogranin A, carnosinase I) with areas under the curve of 0.90 (0.85-0.94 95% confidence interval [CI]) and 0.88 (0.81-0.94 CI), respectively. CONCLUSIONS Four novel CSF biomarkers for AD (NrCAM, YKL-40, chromogranin A, carnosinase I) can improve the diagnostic accuracy of Aβ42 and tau. Together, these six markers describe six clinicopathological stages from cognitive normalcy to mild dementia, including stages defined by increased risk of cognitive decline. Such a panel might improve clinical trial efficiency by guiding subject enrollment and monitoring disease progression. Further studies will be required to validate this panel and evaluate its potential for distinguishing AD from other dementing conditions.
Collapse
Affiliation(s)
- Richard J Perrin
- Division of Neuropathology, Washington University School of Medicine, St. Louis, Missouri, United States of America.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
Axon and synapse degeneration are common components of many neurodegenerative diseases, and their rescue is essential for effective neuroprotection. The chimeric Wallerian degeneration slow protein (Wld(S)) protects axons dose dependently, but its mechanism is still elusive. We recently showed that Wld(S) acts at a non-nuclear location and is present in axons. This and other recent reports support a model in which Wld(S) protects by extranuclear redistribution of its nuclear NMNAT1 portion. However, it remains unclear whether cytoplasmic NMNAT1 acts locally in axons and synapses or at a non-nuclear site within cell bodies. The potency of axon protection by non-nuclear NMNAT1 relative to Wld(S) also needs to be established in vivo. Because the N-terminal portion of Wld(S) (N70) localized to axons, we hypothesized that it mediates the trafficking of the NMNAT1 portion. To test this, we substituted N70 with an axonal targeting peptide derived from amyloid precursor protein, and fused this to NMNAT1 with disrupted nuclear targeting. In transgenic mice, this transformed NMNAT1 from a molecule unable to inhibit Wallerian degeneration, even at high expression levels, into a protein more potent than Wld(S), able to preserve injured axons for several weeks at undetectable expression levels. Preventing NMNAT1 axonal delivery abolished its protective effect. Axonally targeted NMNAT1 localized to vesicular structures, colocalizing with extranuclear Wld(S), and was cotransported at least partially with mitochondria. We conclude that axonal targeting of NMNAT activity is both necessary and sufficient to delay Wallerian degeneration, and that promoting axonal and synaptic delivery greatly enhances the effectiveness.
Collapse
|
47
|
Fernández-Monreal M, Oung T, Hanson HH, O'Leary R, Janssen WG, Dolios G, Wang R, Phillips GR. γ-protocadherins are enriched and transported in specialized vesicles associated with the secretory pathway in neurons. Eur J Neurosci 2010; 32:921-31. [PMID: 20849527 DOI: 10.1111/j.1460-9568.2010.07386.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Gamma protocadherins (Pcdh-γs) resemble classical cadherins and have the potential to engage in cell-cell interactions with homophilic properties. Emerging evidence suggests non-conventional roles for some protocadherins in neural development. We sought to determine whether Pcdh-γ trafficking in neurons is consistent with an intracellular role for these molecules. Here we show that, in contrast to the largely surface localization of classical cadherins, endogenous Pcdh-γs are primarily intracellular in rat neurons in vivo and are equally distributed within organelles of subsynaptic dendritic and axonal compartments. A strikingly higher proportion of Pcdh-γ-containing organelles in synaptic compartments was observed at postnatal day 16. To determine the origin of Pcdh-γ-trafficking organelles, we isolated organelles with Pcdh-γ antibody-coupled magnetic beads from brain organelle suspensions. Vesicles with high levels of COPII and endoplasmic reticulum-Golgi intermediate compartment (ERGIC) components were isolated with the Pcdh-γ antibody but not with the classical cadherin antibody. In cultured hippocampal neurons, Pcdh-γ immunolabeling partially overlapped with calnexin- and COPII-positive puncta in dendrites. Mobile Pcdh-γ-GFP profiles dynamically codistributed with a DsRed construct coupled to ER retention signals by live imaging. Pcdh-γ expression correlated with accumulations of tubulovesicular and ER-like organelles in dendrites. Our results are consistent with the possibility that Pcdh-γs could have a unique function within the secretory pathway in addition to their documented surface roles.
Collapse
|
48
|
Chen G, Yang Y, Fröhlich O, Klein JD, Sands JM. Suppression subtractive hybridization analysis of low-protein diet- and vitamin D-induced gene expression from rat kidney inner medullary base. Physiol Genomics 2010; 41:203-11. [PMID: 20197420 DOI: 10.1152/physiolgenomics.00129.2009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Protein restriction and hypercalcemia result in a urinary concentrating defect in rats and humans. Previous tubular perfusion studies show that there is an increased active urea transport activity in the initial inner medullary (IM) collecting duct in low-protein diet (LPD) and vitamin D (Vit D) animal models. To investigate the possible mechanisms that cause the urinary concentrating defect and to clone the new active urea transporter, we employed a modified two-tester suppression subtractive hybridization (ttSSH) approach and examined gene expression induced by LPD and Vit D in kidney IM base. Approximately 600 clones from the subtracted library were randomly selected; 150 clones were further confirmed to be the true positive genes by slot blot hybridization with subtracted probes from LPD and Vit D and sent for DNA sequencing. We identified 10 channel/transporter genes that were upregulated in IM base in LPD and Vit D animal models; 8 were confirmed by real-time PCR. These genes include aquaporin 2 (AQP2), two-pore calcium channel protein 2, brain-specific organic cation transporter, Na(+)- and H(+)-coupled glutamine transporter, and solute carrier family 25. Nine genes are totally new, and twelve are uncharacterized hypothetical proteins. Among them, four genes were shown to be new transmembrane proteins as judged by Kyte-Doolittle hydrophobic plot analysis. ttSSH provides a useful method to identify new genes from two conditioned populations.
Collapse
|
49
|
Hata S, Fujishige S, Araki Y, Kato N, Araseki M, Nishimura M, Hartmann D, Saftig P, Fahrenholz F, Taniguchi M, Urakami K, Akatsu H, Martins RN, Yamamoto K, Maeda M, Yamamoto T, Nakaya T, Gandy S, Suzuki T. Alcadein cleavages by amyloid beta-precursor protein (APP) alpha- and gamma-secretases generate small peptides, p3-Alcs, indicating Alzheimer disease-related gamma-secretase dysfunction. J Biol Chem 2009; 284:36024-36033. [PMID: 19864413 PMCID: PMC2794718 DOI: 10.1074/jbc.m109.057497] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2009] [Revised: 10/24/2009] [Indexed: 02/03/2023] Open
Abstract
Alcadeins (Alcs) constitute a family of neuronal type I membrane proteins, designated Alc(alpha), Alc(beta), and Alc(gamma). The Alcs express in neurons dominantly and largely colocalize with the Alzheimer amyloid precursor protein (APP) in the brain. Alcs and APP show an identical function as a cargo receptor of kinesin-1. Moreover, proteolytic processing of Alc proteins appears highly similar to that of APP. We found that APP alpha-secretases ADAM 10 and ADAM 17 primarily cleave Alc proteins and trigger the subsequent secondary intramembranous cleavage of Alc C-terminal fragments by a presenilin-dependent gamma-secretase complex, thereby generating "APP p3-like" and non-aggregative Alc peptides (p3-Alcs). We determined the complete amino acid sequence of p3-Alc(alpha), p3-Alc(beta), and p3-Alc(gamma), whose major species comprise 35, 37, and 31 amino acids, respectively, in human cerebrospinal fluid. We demonstrate here that variant p3-Alc C termini are modulated by FAD-linked presenilin 1 mutations increasing minor beta-amyloid species Abeta42, and these mutations alter the level of minor p3-Alc species. However, the magnitudes of C-terminal alteration of p3-Alc(alpha), p3-Alc(beta), and p3-Alc(gamma) were not equivalent, suggesting that one type of gamma-secretase dysfunction does not appear in the phenotype equivalently in the cleavage of type I membrane proteins. Because these C-terminal alterations are detectable in human cerebrospinal fluid, the use of a substrate panel, including Alcs and APP, may be effective to detect gamma-secretase dysfunction in the prepathogenic state of Alzheimer disease subjects.
Collapse
Affiliation(s)
- Saori Hata
- Department of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Sayaka Fujishige
- Department of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Yoichi Araki
- Department of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Naoko Kato
- Department of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Masahiko Araseki
- Department of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Masaki Nishimura
- Neurology Unit, Molecular Neuroscience Research Center, Shiga University of Medical Science, Otsu 520-2192, Japan
| | - Dieter Hartmann
- Department of Anatomy, University of Bonn, 53115 Bonn, Germany
| | - Paul Saftig
- Institute of Biochemistry, University of Kiel, Kiel, Germany
| | - Falk Fahrenholz
- Institute of Biochemistry, Johannes Gutenberg-University, Mainz, Germany
| | - Miyako Taniguchi
- Department of Biological Regulation, School of Health Science, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan
| | - Katsuya Urakami
- Department of Biological Regulation, School of Health Science, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan
| | - Hiroyasu Akatsu
- Choju Medical Institute, Fukushimura Hospital, Toyohashi 441-8124, Japan
| | - Ralph N Martins
- Centre of Excellence for Alzheimer's Disease Research and Care and the Sir James McCusker Alzheimer's Disease Research Unit, School of Exercise, Biomedical and Health Sciences, Edith Cowan University, Joondalup, 6027 Western Australia, Australia
| | - Kazuo Yamamoto
- Department of Integrated Bioscience, Graduate School of Frontier Sciences, University of Tokyo, Kashiwa 277-8562, Japan
| | - Masahiro Maeda
- Immuno-Biological Laboratories Co., Ltd., Fujioka 375-0005, Japan
| | - Tohru Yamamoto
- Department of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Tadashi Nakaya
- Department of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Sam Gandy
- Department of Neurology and Psychiatry, Alzheimer's Disease Research Center, Mount Sinai School of Medicine, New York, New York 10029; James J. Peters Veterans Administration Medical Center, Bronx, New York 10468.
| | - Toshiharu Suzuki
- Department of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan.
| |
Collapse
|
50
|
Mitchell JC, Ariff BB, Yates DM, Lau KF, Perkinton MS, Rogelj B, Stephenson JD, Miller CCJ, McLoughlin DM. X11beta rescues memory and long-term potentiation deficits in Alzheimer's disease APPswe Tg2576 mice. Hum Mol Genet 2009; 18:4492-500. [PMID: 19744962 DOI: 10.1093/hmg/ddp408] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Increased production and deposition of amyloid beta-protein (Abeta) are believed to be key pathogenic events in Alzheimer's disease. As such, routes for lowering cerebral Abeta levels represent potential therapeutic targets for Alzheimer's disease. X11beta is a neuronal adaptor protein that binds to the intracellular domain of the amyloid precursor protein (APP). Overexpression of X11beta inhibits Abeta production in a number of experimental systems. However, whether these changes to APP processing and Abeta production induced by X11beta overexpression also induce beneficial effects to memory and synaptic plasticity are not known. We report here that X11beta-mediated reduction in cerebral Abeta is associated with normalization of both cognition and in vivo long-term potentiation in aged APPswe Tg2576 transgenic mice that model the amyloid pathology of Alzheimer's disease. Overexpression of X11beta itself has no detectable adverse effects upon mouse behaviour. These findings support the notion that modulation of X11beta function represents a therapeutic target for Abeta-mediated neuronal dysfunction in Alzheimer's disease.
Collapse
Affiliation(s)
- Jacqueline C Mitchell
- MRC Centre for Neurodegeneration Research, King's College London, Institute of Psychiatry, London SE5 8AF, UK
| | | | | | | | | | | | | | | | | |
Collapse
|