1
|
Lee S, Jo M, Lee HE, Jeon YM, Kim S, Kwon Y, Woo J, Han S, Mun JY, Kim HJ. HEXA-018, a Novel Inducer of Autophagy, Rescues TDP-43 Toxicity in Neuronal Cells. Front Pharmacol 2021; 12:747975. [PMID: 34925009 PMCID: PMC8675103 DOI: 10.3389/fphar.2021.747975] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 11/05/2021] [Indexed: 11/13/2022] Open
Abstract
The autophagy-lysosomal pathway is an essential cellular mechanism that degrades aggregated proteins and damaged cellular components to maintain cellular homeostasis. Here, we identified HEXA-018, a novel compound containing a catechol derivative structure, as a novel inducer of autophagy. HEXA-018 increased the LC3-I/II ratio, which indicates activation of autophagy. Consistent with this result, HEXA-018 effectively increased the numbers of autophagosomes and autolysosomes in neuronal cells. We also found that the activation of autophagy by HEXA-018 is mediated by the AMPK-ULK1 pathway in an mTOR-independent manner. We further showed that ubiquitin proteasome system impairment- or oxidative stress-induced neurotoxicity was significantly reduced by HEXA-018 treatment. Moreover, oxidative stress-induced mitochondrial dysfunction was strongly ameliorated by HEXA-018 treatment. In addition, we investigated the efficacy of HEXA-018 in models of TDP-43 proteinopathy. HEXA-018 treatment mitigated TDP-43 toxicity in cultured neuronal cell lines and Drosophila. Our data indicate that HEXA-018 could be a new drug candidate for TDP-43-associated neurodegenerative diseases.
Collapse
Affiliation(s)
- Shinrye Lee
- Dementia Research Group, Korea Brain Research Institute (KBRI), Daegu, South Korea
| | - Myungjin Jo
- Dementia Research Group, Korea Brain Research Institute (KBRI), Daegu, South Korea
| | - Hye Eun Lee
- Neural Circuit Research Group, Korea Brain Research Institute (KBRI), Daegu, South Korea
| | - Yu-Mi Jeon
- Dementia Research Group, Korea Brain Research Institute (KBRI), Daegu, South Korea
| | - Seyeon Kim
- Dementia Research Group, Korea Brain Research Institute (KBRI), Daegu, South Korea.,Department of Brain and Cognitive Sciences, DGIST, Daegu, South Korea
| | - Younghwi Kwon
- Dementia Research Group, Korea Brain Research Institute (KBRI), Daegu, South Korea.,Department of Brain and Cognitive Sciences, DGIST, Daegu, South Korea
| | - Junghwa Woo
- Dementia Research Group, Korea Brain Research Institute (KBRI), Daegu, South Korea
| | - Shin Han
- Hexa Pharmatec, Ansan-si, South Korea
| | - Ji Young Mun
- Neural Circuit Research Group, Korea Brain Research Institute (KBRI), Daegu, South Korea
| | - Hyung-Jun Kim
- Dementia Research Group, Korea Brain Research Institute (KBRI), Daegu, South Korea
| |
Collapse
|
2
|
Lee S, Jeon YM, Jo M, Kim HJ. Overexpression of SIRT3 Suppresses Oxidative Stress-induced Neurotoxicity and Mitochondrial Dysfunction in Dopaminergic Neuronal Cells. Exp Neurobiol 2021; 30:341-355. [PMID: 34737239 PMCID: PMC8572659 DOI: 10.5607/en21021] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 10/20/2021] [Accepted: 10/21/2021] [Indexed: 12/18/2022] Open
Abstract
Sirtuin 3 (SIRT3), a well-known mitochondrial deacetylase, is involved in mitochondrial function and metabolism under various stress conditions. In this study, we found that the expression of SIRT3 was markedly increased by oxidative stress in dopaminergic neuronal cells. In addition, SIRT3 overexpression enhanced mitochondrial activity in differentiated SH-SY5Y cells. We also showed that SIRT3 overexpression attenuated rotenone- or H2O2-induced toxicity in differentiated SH-SY5Y cells (human dopaminergic cell line). We further found that knockdown of SIRT3 enhanced rotenone- or H2O2-induced toxicity in differentiated SH-SY5Y cells. Moreover, overexpression of SIRT3 mitigated cell death caused by LPS/IFN-γ stimulation in astrocytes. We also found that the rotenone treatment increases the level of SIRT3 in Drosophila brain. We observed that downregulation of sirt2 (Drosophila homologue of SIRT3) significantly accelerated the rotenone-induced toxicity in flies. Taken together, these findings suggest that the overexpression of SIRT3 mitigates oxidative stress-induced cell death and mitochondrial dysfunction in dopaminergic neurons and astrocytes.
Collapse
Affiliation(s)
- Shinrye Lee
- Dementia Research Group, Korea Brain Research Institute (KBRI), Daegu 41062, Korea
| | - Yu-Mi Jeon
- Dementia Research Group, Korea Brain Research Institute (KBRI), Daegu 41062, Korea
| | - Myungjin Jo
- Dementia Research Group, Korea Brain Research Institute (KBRI), Daegu 41062, Korea
| | - Hyung-Jun Kim
- Dementia Research Group, Korea Brain Research Institute (KBRI), Daegu 41062, Korea
| |
Collapse
|
3
|
Lee S, Kim S, Kang HY, Lim HR, Kwon Y, Jo M, Jeon YM, Kim SR, Kim K, Ha CM, Lee S, Kim HJ. The overexpression of TDP-43 in astrocytes causes neurodegeneration via a PTP1B-mediated inflammatory response. J Neuroinflammation 2020; 17:299. [PMID: 33054766 PMCID: PMC7556969 DOI: 10.1186/s12974-020-01963-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 09/23/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Cytoplasmic inclusions of transactive response DNA binding protein of 43 kDa (TDP-43) in neurons and astrocytes are a feature of some neurodegenerative diseases, such as frontotemporal lobar degeneration with TDP-43 (FTLD-TDP) and amyotrophic lateral sclerosis (ALS). However, the role of TDP-43 in astrocyte pathology remains largely unknown. METHODS To investigate whether TDP-43 overexpression in primary astrocytes could induce inflammation, we transfected primary astrocytes with plasmids encoding Gfp or TDP-43-Gfp. The inflammatory response and upregulation of PTP1B in transfected cells were examined using quantitative RT-PCR and immunoblot analysis. Neurotoxicity was analysed in a transwell coculture system of primary cortical neurons with astrocytes and cultured neurons treated with astrocyte-conditioned medium (ACM). We also examined the lifespan, performed climbing assays and analysed immunohistochemical data in pan-glial TDP-43-expressing flies in the presence or absence of a Ptp61f RNAi transgene. RESULTS PTP1B inhibition suppressed TDP-43-induced secretion of inflammatory cytokines (interleukin 1 beta (IL-1β), interleukin 6 (IL-6) and tumour necrosis factor alpha (TNF-α)) in primary astrocytes. Using a neuron-astrocyte coculture system and astrocyte-conditioned media treatment, we demonstrated that PTP1B inhibition attenuated neuronal death and mitochondrial dysfunction caused by overexpression of TDP-43 in astrocytes. In addition, neuromuscular junction (NMJ) defects, a shortened lifespan, inflammation and climbing defects caused by pan-glial overexpression of TDP-43 were significantly rescued by downregulation of ptp61f (the Drosophila homologue of PTP1B) in flies. CONCLUSIONS These results indicate that PTP1B inhibition mitigates the neuronal toxicity caused by TDP-43-induced inflammation in mammalian astrocytes and Drosophila glial cells.
Collapse
Affiliation(s)
- Shinrye Lee
- Dementia Research Group, Korea Brain Research Institute (KBRI), Daegu, 41062, South Korea
| | - Seyeon Kim
- Dementia Research Group, Korea Brain Research Institute (KBRI), Daegu, 41062, South Korea
- Department of Brain & Cognitive Sciences, DGIST, Daegu, 42988, South Korea
| | - Ha-Young Kang
- Gwangju Center, Korea Basic Science Institute (KBSI), Gwangju, 61886, South Korea
| | - Hye Ryeong Lim
- Research Division and Brain Research Core Facilities, Korea Brain Research Institute (KBRI), Daegu, 41062, South Korea
| | - Younghwi Kwon
- Dementia Research Group, Korea Brain Research Institute (KBRI), Daegu, 41062, South Korea
- Department of Brain & Cognitive Sciences, DGIST, Daegu, 42988, South Korea
| | - Myungjin Jo
- Dementia Research Group, Korea Brain Research Institute (KBRI), Daegu, 41062, South Korea
| | - Yu-Mi Jeon
- Dementia Research Group, Korea Brain Research Institute (KBRI), Daegu, 41062, South Korea
| | - Sang Ryong Kim
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Institute of Life Science & Biotechnology, Kyungpook National University, Daegu, 41566, South Korea
- Brain Science and Engineering Institute, Kyungpook National University, Daegu, 41944, South Korea
| | - Kiyoung Kim
- Department of Medical Biotechnology, Soonchunhyang University, Asan, 31538, South Korea
| | - Chang Man Ha
- Research Division and Brain Research Core Facilities, Korea Brain Research Institute (KBRI), Daegu, 41062, South Korea
| | - Seongsoo Lee
- Gwangju Center, Korea Basic Science Institute (KBSI), Gwangju, 61886, South Korea.
| | - Hyung-Jun Kim
- Dementia Research Group, Korea Brain Research Institute (KBRI), Daegu, 41062, South Korea.
| |
Collapse
|
4
|
Lee S, Jeon YM, Cha SJ, Kim S, Kwon Y, Jo M, Jang YN, Lee S, Kim J, Kim SR, Lee KJ, Lee SB, Kim K, Kim HJ. PTK2/FAK regulates UPS impairment via SQSTM1/p62 phosphorylation in TARDBP/TDP-43 proteinopathies. Autophagy 2019; 16:1396-1412. [PMID: 31690171 DOI: 10.1080/15548627.2019.1686729] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
TARDBP/TDP-43 (TAR DNA binding protein) proteinopathies are a common feature in a variety of neurodegenerative disorders, including amyotrophic lateral sclerosis (ALS), frontotemporal lobar degeneration (FTLD), and Alzheimer disease (AD). However, the molecular mechanisms underlying TARDBP-induced neurotoxicity are largely unknown. In this study, we demonstrated that TARDBP proteinopathies induce impairment in the ubiquitin proteasome system (UPS), as evidenced by an accumulation of ubiquitinated proteins and a reduction in proteasome activity in neuronal cells. Through kinase inhibitor screening, we identified PTK2/FAK (PTK2 protein tyrosine kinase 2) as a suppressor of neurotoxicity induced by UPS impairment. Importantly, PTK2 inhibition significantly reduced ubiquitin aggregates and attenuated TARDBP-induced cytotoxicity in a Drosophila model of TARDBP proteinopathies. We further identified that phosphorylation of SQSTM1/p62 (sequestosome 1) at S403 (p-SQSTM1 [S403]), a key component in the autophagic degradation of poly-ubiquitinated proteins, is increased upon TARDBP overexpression and is dependent on the activation of PTK2 in neuronal cells. Moreover, expressing a non-phosphorylated form of SQSTM1 (SQSTM1S403A) significantly repressed the accumulation of insoluble poly-ubiquitinated proteins and neurotoxicity induced by TARDBP overexpression in neuronal cells. In addition, TBK1 (TANK binding kinase 1), a kinase that phosphorylates S403 of SQSTM1, was found to be involved in the PTK2-mediated phosphorylation of SQSTM1. Taken together, our data suggest that the PTK2-TBK1-SQSTM1 axis plays a critical role in the pathogenesis of TARDBP by regulating neurotoxicity induced by UPS impairment. Therefore, targeting the PTK2-TBK1-SQSTM1 axis may represent a novel therapeutic intervention for neurodegenerative diseases with TARDBP proteinopathies.Abbreviations: ALP: macroautophagy/autophagy lysosomal pathway; ALS: amyotrophic lateral sclerosis; ATXN2: ataxin 2; BafA1: bafilomycin A1; cCASP3: cleaved caspase 3; CSNK2: casein kinase 2; FTLD: frontotemporal lobar degeneration; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; OPTN: optineurin; PTK2/FAK: PTK2 protein tyrosine kinase 2; SQSTM1/p62: sequestosome 1; TARDBP/TDP-43: TAR DNA binding protein; TBK1: TANK binding kinase 1; ULK1: unc-51 like autophagy activating kinase 1; UPS: ubiquitin-proteasome system.
Collapse
Affiliation(s)
- Shinrye Lee
- Dementia Research Group, Korea Brain Research Institute (KBRI) , Daegu, South Korea
| | - Yu-Mi Jeon
- Dementia Research Group, Korea Brain Research Institute (KBRI) , Daegu, South Korea
| | - Sun Joo Cha
- Soonchunhyang Institute of Medi-bio Science, Soonchunhyang University , Cheonan, South Korea
| | - Seyeon Kim
- Dementia Research Group, Korea Brain Research Institute (KBRI) , Daegu, South Korea.,Department of Brain & Cognitive Sciences, DGIST , Daegu, South Korea
| | - Younghwi Kwon
- Dementia Research Group, Korea Brain Research Institute (KBRI) , Daegu, South Korea.,Department of Brain & Cognitive Sciences, DGIST , Daegu, South Korea
| | - Myungjin Jo
- Dementia Research Group, Korea Brain Research Institute (KBRI) , Daegu, South Korea
| | - You-Na Jang
- Neural circuits Research Group, Korea Brain Research Institute (KBRI) , Daegu, South Korea
| | - Seongsoo Lee
- Gwangju Center, Korea Basic Science Institute (KBSI) , Gwangju, South Korea
| | - Jaekwang Kim
- Dementia Research Group, Korea Brain Research Institute (KBRI) , Daegu, South Korea
| | - Sang Ryong Kim
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Institute of Life Science & Biotechnology, Kyungpook National University , Daegu, South Korea.,Brain Science and Engineering Institute, Kyungpook National University , Daegu, South Korea
| | - Kea Joo Lee
- Neural circuits Research Group, Korea Brain Research Institute (KBRI) , Daegu, South Korea
| | - Sung Bae Lee
- Department of Brain & Cognitive Sciences, DGIST , Daegu, South Korea
| | - Kiyoung Kim
- Soonchunhyang Institute of Medi-bio Science, Soonchunhyang University , Cheonan, South Korea.,Department of Medical Biotechnology, Soonchunhyang University , Asan, South Korea
| | - Hyung-Jun Kim
- Dementia Research Group, Korea Brain Research Institute (KBRI) , Daegu, South Korea
| |
Collapse
|
5
|
Jo M, Lee S, Kim K, Lee S, Kim SR, Kim HJ. Inhibition of MEK5 suppresses TDP-43 toxicity via the mTOR-independent activation of the autophagy-lysosome pathway. Biochem Biophys Res Commun 2019; 513:925-932. [PMID: 31005259 DOI: 10.1016/j.bbrc.2019.04.088] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 04/12/2019] [Indexed: 12/19/2022]
Abstract
The most prominent hallmarks of many neurodegenerative diseases are the accumulation of misfolded protein aggregates and the death of certain neuronal populations. Autophagy is the major intracellular mechanism that degrades protein aggregates and damaged cellular components. Many studies have reported that the dysfunction of autophagy is associated with several neurodegenerative diseases, such as Alzheimer's disease, amyotrophic lateral sclerosis (ALS), and Parkinson's disease. Here, we identified a novel mechanism of autophagy regulation. Inhibition of MEK5 reduced the level of p62 and increased the ratio of LC3-II to LC3-I, which is a marker for the activation of the autophagy-lysosome pathway (ALP). One of the most well-known regulators of the ALP is mTOR, and previous studies have reported that the major substrate of MEK5 is ERK5. However, we found that MEK5 modulates the autophagy-lysosome pathway in an mTOR- and ERK5-independent manner. Moreover, MEK5 inhibition alleviated the mislocalization of TDP-43 (an ALS-associated protein) and cell death in TDP-43-GFP-expressing neuronal cells. Taken together, these findings suggest that MEK5 is a novel autophagy modulator and that this kinase could be a therapeutic target for neurodegenerative diseases such as amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Myungjin Jo
- Korea Brain Research Institute, Department of Neural Development and Disease, 61, Cheomdan-ro, Dong-gu, Daegu, 41062, Republic of Korea
| | - Shinrye Lee
- Korea Brain Research Institute, Department of Neural Development and Disease, 61, Cheomdan-ro, Dong-gu, Daegu, 41062, Republic of Korea
| | - Kiyoung Kim
- Soonchunhyang Institute of Medi-bio Science, Soonchunhyang University, Cheonan, 31151, South Korea; Department of Medical Biotechnology, Soonchunhyang University, Asan, 31538, South Korea
| | - Seongsoo Lee
- Gwangju Center, Korea Basic Science Institute (KBSI), Gwangju, 61186, South Korea
| | - Sang Ryong Kim
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Institute of Life Science & Biotechnology, Kyungpook National University, Daegu, 41566, South Korea; Brain Science and Engineering Institute, Kyungpook National University, Daegu, 41944, South Korea
| | - Hyung-Jun Kim
- Korea Brain Research Institute, Department of Neural Development and Disease, 61, Cheomdan-ro, Dong-gu, Daegu, 41062, Republic of Korea.
| |
Collapse
|
6
|
Jeon YM, Lee S, Kim S, Kwon Y, Kim K, Chung CG, Lee S, Lee SB, Kim HJ. Neuroprotective Effects of Protein Tyrosine Phosphatase 1B Inhibition against ER Stress-Induced Toxicity. Mol Cells 2017; 40:280-290. [PMID: 28359145 PMCID: PMC5424274 DOI: 10.14348/molcells.2017.2320] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 03/21/2017] [Accepted: 03/22/2017] [Indexed: 12/15/2022] Open
Abstract
Several lines of evidence suggest that endoplasmic reticulum (ER) stress plays a critical role in the pathogenesis of many neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. Protein tyrosine phosphatase 1B (PTP1B) is known to regulate the ER stress signaling pathway, but its role in neuronal systems in terms of ER stress remains largely unknown. Here, we showed that rotenone-induced toxicity in human neuroblastoma cell lines and mouse primary cortical neurons was ameliorated by PTP1B inhibition. Moreover, the increase in the level of ER stress markers (eIF2α phosphorylation and PERK phosphorylation) induced by rotenone treatment was obviously suppressed by concomitant PTP1B inhibition. However, the rotenone-induced production of reactive oxygen species (ROS) was not affected by PTP1B inhibition, suggesting that the neuroprotective effect of the PTP1B inhibitor is not associated with ROS production. Moreover, we found that MG132-induced toxicity involving proteasome inhibition was also ameliorated by PTP1B inhibition in a human neuroblastoma cell line and mouse primary cortical neurons. Consistently, downregulation of the PTP1B homologue gene in Drosophila mitigated rotenone- and MG132-induced toxicity. Taken together, these findings indicate that PTP1B inhibition may represent a novel therapeutic approach for ER stress-mediated neurodegenerative diseases.
Collapse
Affiliation(s)
- Yu-Mi Jeon
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), Daegu 41068,
Korea
| | - Shinrye Lee
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), Daegu 41068,
Korea
| | - Seyeon Kim
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), Daegu 41068,
Korea
| | - Younghwi Kwon
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), Daegu 41068,
Korea
| | - Kiyoung Kim
- Department of Medical Biotechnology, Soonchunhyang University, Asan 31538,
Korea
| | - Chang Geon Chung
- Department of Brain & Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988,
Korea
| | - Seongsoo Lee
- Gwangju Center, Korea Basic Science Institute (KBSI), Gwangju 61186,
Korea
| | - Sung Bae Lee
- Department of Brain & Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988,
Korea
| | - Hyung-Jun Kim
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), Daegu 41068,
Korea
| |
Collapse
|
7
|
Jeon S, Jha MK, Ock J, Seo J, Jin M, Cho H, Lee WH, Suk K. Role of lipocalin-2-chemokine axis in the development of neuropathic pain following peripheral nerve injury. J Biol Chem 2013; 288:24116-27. [PMID: 23836894 DOI: 10.1074/jbc.m113.454140] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Lipocalin 2 (LCN2), which is also known as 24p3 and neutrophil gelatinase-associated lipocalin (NGAL), binds small, hydrophobic ligands and interacts with cell surface receptor 24p3R to regulate diverse cellular processes. In the present study, we examined the role of LCN2 in the pathogenesis of neuropathic pain using a mouse model of spared nerve injury (SNI). Lcn2 mRNA levels were significantly increased in the dorsal horn of the spinal cord after SNI, and LCN2 protein was mainly localized in neurons of the dorsal and ventral horns. LCN2 receptor 24p3R was expressed in spinal neurons and microglia after SNI. Lcn2-deficient mice exhibited significantly less mechanical pain hypersensitivity during the early phase after SNI, and an intrathecal injection of recombinant LCN2 protein elicited mechanical pain hypersensitivity in naive animals. Lcn2 deficiency, however, did not affect acute nociceptive pain. Lcn2-deficient mice showed significantly less microglial activation and proalgesic chemokine (CCL2 and CXCL1) production in the spinal cord after SNI than wild-type mice, and recombinant LCN2 protein induced the expression of these chemokines in cultured neurons. Furthermore, the expression of LCN2 and its receptor was detected in neutrophils and macrophages in the sciatic nerve following SNI, suggesting the potential role of peripheral LCN2 in neuropathic pain. Taken together, our results indicate that LCN2 plays a critical role in the development of pain hypersensitivity following peripheral nerve injury and suggest that LCN2 mediates neuropathic pain by inducing chemokine expression and subsequent microglial activation.
Collapse
Affiliation(s)
- Sangmin Jeon
- Department of Pharmacology, Brain Science and Engineering Institute, Kyungpook National University School of Medicine, Daegu 700-422, Korea
| | | | | | | | | | | | | | | |
Collapse
|
8
|
Chami L, Buggia-Prévot V, Duplan E, Delprete D, Chami M, Peyron JF, Checler F. Nuclear factor-κB regulates βAPP and β- and γ-secretases differently at physiological and supraphysiological Aβ concentrations. J Biol Chem 2012; 287:24573-84. [PMID: 22654105 PMCID: PMC3397882 DOI: 10.1074/jbc.m111.333054] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Revised: 05/14/2012] [Indexed: 11/06/2022] Open
Abstract
Anatomical lesions in Alzheimer disease-affected brains mainly consist of senile plaques, inflammation stigmata, and oxidative stress. The nuclear factor-κB (NF-κB) is a stress-activated transcription factor that is activated around senile plaques. We have assessed whether NF-κB could be differentially regulated at physiological or supraphysiological levels of amyloid β (Aβ) peptides. Under these experimental conditions, we delineated the putative NF-κB-dependent modulation of all cellular participants in Aβ production, namely its precursor βAPP (β-amyloid precursor protein) and the β- and γ-secretases, the two enzymatic machines involved in Aβ genesis. Under physiological conditions, NF-κB lowers the transcriptional activity of the promoters of βAPP, β-secretase (β-site APP-cleaving enzyme 1, BACE1), and of the four protein components (Aph-1, Pen-2, nicastrin, presenilin-1, or presenilin-2) of the γ-secretase in HEK293 cells. This was accompanied by a reduction of both protein levels and enzymatic activities, thereby ultimately yielding lower amounts of Aβ and AICD (APP intracellular domain). In stably transfected Swedish βAPP-expressing HEK293 cells triggering supraphysiological concentrations of Aβ peptides, NF-κB activates the transcription of βAPP, BACE1, and some of the γ-secretase members and increases protein expression and enzymatic activities, resulting in enhanced Aβ production. Our pharmacological approach using distinct NF-κB kinase modulators indicates that both NF-κB canonical and alternative pathways are involved in the control of Aβ production. Overall, our data demonstrate that under physiological conditions, NF-κB triggers a repressive effect on Aβ production that contributes to maintaining its homeostasis, while NF-κB participates in a degenerative cycle where Aβ would feed its own production under pathological conditions.
Collapse
Affiliation(s)
- Linda Chami
- From the Équipe Labellisée “Fondation pour la Recherche Médicale” and “Excellence Laboratory Distalz”, Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275 CNRS/UNSA, 660 Route des Lucioles, 06560, Valbonne, France and
| | - Virginie Buggia-Prévot
- From the Équipe Labellisée “Fondation pour la Recherche Médicale” and “Excellence Laboratory Distalz”, Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275 CNRS/UNSA, 660 Route des Lucioles, 06560, Valbonne, France and
| | - Eric Duplan
- From the Équipe Labellisée “Fondation pour la Recherche Médicale” and “Excellence Laboratory Distalz”, Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275 CNRS/UNSA, 660 Route des Lucioles, 06560, Valbonne, France and
| | - Dolores Delprete
- From the Équipe Labellisée “Fondation pour la Recherche Médicale” and “Excellence Laboratory Distalz”, Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275 CNRS/UNSA, 660 Route des Lucioles, 06560, Valbonne, France and
| | - Mounia Chami
- From the Équipe Labellisée “Fondation pour la Recherche Médicale” and “Excellence Laboratory Distalz”, Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275 CNRS/UNSA, 660 Route des Lucioles, 06560, Valbonne, France and
| | - Jean-François Peyron
- the Centre Méditerranéen de Médecine Moléculaire, UMR INSERM U895/UNS, 151 route Saint Antoine de Ginestière, BP 2 3194, 06204 Nice Cedex 3, France
| | - Frédéric Checler
- From the Équipe Labellisée “Fondation pour la Recherche Médicale” and “Excellence Laboratory Distalz”, Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275 CNRS/UNSA, 660 Route des Lucioles, 06560, Valbonne, France and
| |
Collapse
|
9
|
Lee S, Jang E, Kim JH, Kim JH, Lee WH, Suk K. Lipocalin-type prostaglandin D2 synthase protein regulates glial cell migration and morphology through myristoylated alanine-rich C-kinase substrate: prostaglandin D2-independent effects. J Biol Chem 2012; 287:9414-28. [PMID: 22275363 DOI: 10.1074/jbc.m111.330662] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Prostaglandin D synthase (PGDS) is responsible for the conversion of PGH(2) to PGD(2). Two distinct types of PGDS have been identified: hematopoietic-type PGDS (H-PGDS) and lipocalin-type PGDS (L-PGDS). L-PGDS acts as both a PGD(2)-synthesizing enzyme and as an extracellular transporter of various lipophilic small molecules. Although L-PGDS is one of the most abundant proteins in the cerebrospinal fluid, little is known about the function of L-PGDS in the central nervous system (CNS). To better understand the role of L-PGDS in the CNS, effects of L-PGDS on the migration and morphology of glial cells were investigated. The L-PGDS protein accelerated the migration of cultured glial cells. Expression of the L-pgds gene was detected in glial cells and neurons. L-PGDS protein also induced morphological changes in glia similar to the characteristic phenotypic changes in reactive gliosis. L-PGDS-induced cell migration was associated with augmented formation of actin filaments and focal adhesion, which was accompanied by activation of AKT, RhoA, and JNK pathways. L-PGDS protein injected into the mouse brain promoted migration and accumulation of astrocytes in vivo. Furthermore, the cell migration-promoting effect of L-PGDS on glial cells was independent of the PGD(2) products. The L-PGDS protein interacted with myristoylated alanine-rich protein kinase C substrate (MARCKS) to promote cell migration. These results demonstrate the critical role of L-PGDS as a secreted lipocalin in the regulation of glial cell migration and morphology. The results also indicate that L-PGDS may participate in reactive gliosis in an autocrine or paracrine manner, and may have pathological implications in neuroinflammatory diseases.
Collapse
Affiliation(s)
- Shinrye Lee
- Department of Pharmacology, Brain Science & Engineering Institute, CMRI, Kyungpook National University School of Medicine, Kyungpook National University, Daegu, Korea
| | | | | | | | | | | |
Collapse
|
10
|
Kim H, Lee S, Park HC, Lee WH, Lee MS, Suk K. Modulation of glial and neuronal migration by lipocalin-2 in zebrafish. Immune Netw 2011; 11:342-7. [PMID: 22346773 PMCID: PMC3275702 DOI: 10.4110/in.2011.11.6.342] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Revised: 10/14/2011] [Accepted: 10/18/2011] [Indexed: 12/01/2022] Open
Abstract
Background Glial cells are involved in immune and inflammatory responses in the central nervous system (CNS). Glial cells such as microglia and astrocytes also provide structural and functional support for neurons. Migration and morphological changes of CNS cells are associated with their physiological as well as pathological functions. The secreted protein lipocalin-2 (LCN2) has been previously implicated in regulation of diverse cellular processes of glia and neurons, including cell migration and morphology. Methods Here, we employed a zebrafish model to analyze the role of LCN2 in CNS cell migration and morphology in vivo. In the first part of this study, we examined the indirect effect of LCN2 on cell migration and morphology of microglia, astrocytes, and neurons cultured in vitro. Results Conditioned media collected from LCN2-treated astrocytes augmented migration of glia and neurons in the Boyden chamber assay. The conditioned media also increased the number of neuronal processes. Next, in order to further understand the role of LCN2 in the CNS in vivo, LCN2 was ectopically expressed in the zebrafish spinal cord. Expression of exogenous LCN2 modulated neuronal cell migration in the spinal cord of zebrafish embryos, supporting the role of LCN2 as a cell migration regulator in the CNS. Conclusion Thus, LCN2 proteins secreted under diverse conditions may play an important role in CNS immune and inflammatory responses by controlling cell migration and morphology.
Collapse
Affiliation(s)
- Ho Kim
- Department of Medical Science, Korea University Ansan Hospital, Ansan 425-707, Korea
| | | | | | | | | | | |
Collapse
|
11
|
Lee S, Lee WH, Lee MS, Mori K, Suk K. Regulation by lipocalin-2 of neuronal cell death, migration, and morphology. J Neurosci Res 2011; 90:540-50. [PMID: 22038922 DOI: 10.1002/jnr.22779] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2011] [Revised: 06/21/2011] [Accepted: 07/27/2011] [Indexed: 11/11/2022]
Abstract
A secreted protein, lipocalin-2 (LCN2), has been previously shown to regulate a variety of cellular phenotypes such as cell death, migration, and morphology. The role of LCN2, however, appears to be different depending on the cellular context. Here, we investigated how LCN2 influences neuronal phenotypes by using primary cortical neuronal cell cultures and neuroblastoma cell lines as a model. When exposed to LCN2 protein, neurons and neuroblastoma cells were sensitized to cell death evoked by nitric oxide, oxidative stress, and tumor necrosis factor-α (TNF-α). A forced expression of lcn2 in glia enhanced neuronal cell death in cocultures of glia and neurons, indicating that both exogenous protein addition and endogenous expression of lcn2 give rise to similar results. Iron and BCL2-interacting mediator of cell death (BIM) protein were involved in LCN2-induced cell death sensitization, based on the studies using iron donor, chelator, siderophore, and short hairpin RNA (shRNA)-mediated knockdown of bim expression. Furthermore, cell migration assay and immunofluorescence microscopic observation revealed that LCN2 accelerated neuronal motility and process extension, suggesting multiple roles for LCN2 in the regulation of neuronal cell death, migration, and morphology.
Collapse
Affiliation(s)
- Shinrye Lee
- Department of Pharmacology, Brain Science and Engineering Institute, CMRI, Kyungpook National University School of Medicine, Daegu, Korea
| | | | | | | | | |
Collapse
|
12
|
Lee S, Kim JH, Kim JH, Seo JW, Han HS, Lee WH, Mori K, Nakao K, Barasch J, Suk K. Lipocalin-2 Is a chemokine inducer in the central nervous system: role of chemokine ligand 10 (CXCL10) in lipocalin-2-induced cell migration. J Biol Chem 2011; 286:43855-43870. [PMID: 22030398 DOI: 10.1074/jbc.m111.299248] [Citation(s) in RCA: 135] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The secreted protein lipocalin-2 (LCN2) has been implicated in diverse cellular processes, including cell morphology and migration. Little is known, however, about the role of LCN2 in the CNS. Here, we show that LCN2 promotes cell migration through up-regulation of chemokines in brain. Studies using cultured glial cells, microvascular endothelial cells, and neuronal cells suggest that LCN2 may act as a chemokine inducer on the multiple cell types in the CNS. In particular, up-regulation of CXCL10 by JAK2/STAT3 and IKK/NF-κB pathways in astrocytes played a pivotal role in LCN2-induced cell migration. The cell migration-promoting activity of LCN2 in the CNS was verified in vivo using mouse models. The expression of LCN2 was notably increased in brain following LPS injection or focal injury. Mice lacking LCN2 showed the impaired migration of astrocytes to injury sites with a reduced CXCL10 expression in the neuroinflammation or injury models. Thus, the LCN2 proteins, secreted under inflammatory conditions, may amplify neuroinflammation by inducing CNS cells to secrete chemokines such as CXCL10, which recruit additional inflammatory cells.
Collapse
Affiliation(s)
- Shinrye Lee
- Department of Pharmacology, Kyungpook National University School of Medicine, Daegu 700-422, Korea
| | - Jong-Heon Kim
- Department of Pharmacology, Kyungpook National University School of Medicine, Daegu 700-422, Korea
| | - Jae-Hong Kim
- Department of Pharmacology, Kyungpook National University School of Medicine, Daegu 700-422, Korea
| | - Jung-Wan Seo
- Department of Pharmacology, Kyungpook National University School of Medicine, Daegu 700-422, Korea
| | - Hyung-Soo Han
- Department of Physiology, Brain Science & Engineering Institute, Cell and Matrix Research Institute, Kyungpook National University School of Medicine, Daegu 700-422, Korea
| | - Won-Ha Lee
- Departments of School of Life Sciences and Biotechnology, Kyungpook National University School of Medicine, Daegu 700-422, Korea
| | - Kiyoshi Mori
- Department of Medicine and Clinical Science, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Kazuwa Nakao
- Department of Medicine and Clinical Science, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Jonathan Barasch
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, New York 10027
| | - Kyoungho Suk
- Department of Pharmacology, Kyungpook National University School of Medicine, Daegu 700-422, Korea.
| |
Collapse
|
13
|
Ock J, Han HS, Hong SH, Lee SY, Han YM, Kwon BM, Suk K. Obovatol attenuates microglia-mediated neuroinflammation by modulating redox regulation. Br J Pharmacol 2010; 159:1646-62. [PMID: 20397299 DOI: 10.1111/j.1476-5381.2010.00659.x] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND AND PURPOSE Obovatol isolated from the medicinal herb Magnolia obovata exhibits a variety of biological activities. Here, the effect of obovatol and its mechanism of action on microglial activation, neuroinflammation and neurodegeneration were investigated. EXPERIMENTAL APPROACH In microglial BV-2 cells stimulated with lipopolysaccharide (LPS), we measured nitric oxide (NO) and cytokine production, and activation of intracellular signalling pathways by reverse transcription-polymerase chain reaction and Western blots. Cell death was assayed in co-cultures of activated microglia (with bacterial LPS) and neurons and in LPS- induced neuroinflammation in mice in vivo. KEY RESULTS Obovatol inhibited microglial NO production with an IC50 value of 10 mM. Obovatol also inhibited microglial expression of proinflammatory cytokines and inducible nitric-oxide synthase, which was accompanied by the inhibition of multiple signalling pathways such as nuclear factor kappa B, signal transducers and activators of transcription 1, and mitogen-activated protein kinases. In addition, obovatol protected cultured neurons from microglial toxicity and inhibited neuroinflammation in mice in vivo. One molecular target of obovatol in microglia was peroxiredoxin 2 (Prx2), identified by affinity chromatography and mass spectrometry. Obovatol enhanced the reactive oxygen species (ROS)-scavenging activity of Prx2 in vitro, thereby suppressing proinflammatory signalling pathways of microglia where ROS plays an important role. CONCLUSIONS AND IMPLICATIONS Obovatol is not only a useful chemical tool that can be used to investigate microglial signalling, but also a promising drug candidate against neuroinflammatory diseases. Furthermore, our results indicate that Prx2 is a novel drug target that can be exploited for the therapeutic modulation of neuroinflammatory signalling.
Collapse
Affiliation(s)
- Jiyeon Ock
- Department of Pharmacology, Brain Science and Engineering Institute, CMRI, Kyungpook National University, Daegu, Korea
| | | | | | | | | | | | | |
Collapse
|
14
|
Niikura T, Tajima H, Kita Y. Neuronal cell death in Alzheimer's disease and a neuroprotective factor, humanin. Curr Neuropharmacol 2010; 4:139-47. [PMID: 18615127 DOI: 10.2174/157015906776359577] [Citation(s) in RCA: 126] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2005] [Revised: 11/09/2005] [Accepted: 12/08/2005] [Indexed: 11/22/2022] Open
Abstract
Brain atrophy caused by neuronal loss is a prominent pathological feature of Alzheimer's disease (AD). Amyloid beta (Abeta), the major component of senile plaques, is considered to play a central role in neuronal cell death. In addition to removal of the toxic Abeta, direct suppression of neuronal loss is an essential part of AD treatment; however, no such neuroprotective therapies have been developed. Excess amount of Abeta evokes multiple cytotoxic mechanisms, involving increase of the intracellular Ca(2+) level, oxidative stress, and receptor-mediated activation of cell-death cascades. Such diversity in cytotoxic mechanisms induced by Abeta clearly indicates a complex nature of the AD-related neuronal cell death. We have identified a 24-residue peptide, Humanin (HN), which suppresses in vitro neuronal cell death caused by all AD-related insults, including Abeta, so far tested. The anti-AD effect of HN has been further confirmed in vivo using mice with Abeta-induced amnesia. Altogether, such potent neuroprotective activity of HN against AD-relevant cytotoxicity both in vitro and in vivo suggests the potential clinical applications of HN in novel AD therapies aimed at controlling neuronal death.
Collapse
Affiliation(s)
- Takako Niikura
- Department of Pharmacology, KEIO University School of Medicine, 35 Shinanomachi, Tokyo 160-8582, Japan.
| | | | | |
Collapse
|
15
|
Ock J, Kim S, Yi KY, Kim NJ, Han HS, Cho JY, Suk K. A novel anti-neuroinflammatory pyridylimidazole compound KR-31360. Biochem Pharmacol 2010; 79:596-609. [DOI: 10.1016/j.bcp.2009.09.026] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2009] [Revised: 09/23/2009] [Accepted: 09/23/2009] [Indexed: 02/06/2023]
|
16
|
Niikura T. Humanin: a potential peptide for neuroprotective therapy against Alzheimer's disease. Expert Opin Drug Discov 2007; 2:1273-82. [DOI: 10.1517/17460441.2.9.1273] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
17
|
Watts HR, Vince V, Walsh DT, Bresciani LG, Gentleman SM, Jen LS, Anderson PJB. Alterations in presenilin 1 processing by amyloid-beta peptide in the rat retina. Exp Brain Res 2007; 181:69-77. [PMID: 17333007 DOI: 10.1007/s00221-007-0904-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2006] [Accepted: 02/07/2007] [Indexed: 01/03/2023]
Abstract
Accumulating evidence indicates that mutations in the presenilin 1 (PS1) gene are responsible for most cases of familial Alzheimer's disease (AD). Although its biological functions are not yet fully understood, it appears that PS1 plays a role in the processing and trafficking of the amyloid precursor protein (APP). However, little is known about factors that are involved in regulating the metabolism of PS1 especially in relation to AD pathology. In this study, we have examined the effect of optic nerve crush, intravitreal injection of the inflammatory agent lipopolysaccharide (LPS) or injection of amyloid beta(1-42) (A beta(1-42)) on the expression and processing of PS1 in the rat retina. We found that 48 h after injection of A beta(1-42) there was a dramatic alteration in the banding pattern of PS1 on Western blots, as indicated by marked changes in the levels of expression of some of its C- and N-terminal fragments in retinal homogenates. These results suggest an A beta(1-42)-induced potentiation of a non-specific stress-related but inflammation-independent alteration of processing of PS1 in this in vivo model.
Collapse
Affiliation(s)
- Helena R Watts
- Department of Cellular and Molecular Neuroscience, Division of Neuroscience and Mental Health, Faculty of Medicine, Imperial College London, Charing Cross Campus, Fulham Palace Road, London, W6 8RF, UK
| | | | | | | | | | | | | |
Collapse
|
18
|
Alves da Costa C, Sunyach C, Pardossi-Piquard R, Sévalle J, Vincent B, Boyer N, Kawarai T, Girardot N, St. George-Hyslop P, Checler F. Presenilin-dependent gamma-secretase-mediated control of p53-associated cell death in Alzheimer's disease. J Neurosci 2006; 26:6377-85. [PMID: 16763046 PMCID: PMC6675197 DOI: 10.1523/jneurosci.0651-06.2006] [Citation(s) in RCA: 132] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Presenilins (PSs) are part of the gamma-secretase complex that produces the amyloid beta-peptide (Abeta) from its precursor [beta-amyloid precursor protein (betaAPP)]. Mutations in PS that cause familial Alzheimer's disease (FAD) increase Abeta production and trigger p53-dependent cell death. We demonstrate that PS deficiency, catalytically inactive PS mutants, gamma-secretase inhibitors, and betaAPP or amyloid precursor protein-like protein 2 (APLP2) depletion all reduce the expression and activity of p53 and lower the transactivation of its promoter and mRNA expression. p53 expression also is diminished in the brains of PS- or betaAPP-deficient mice. The gamma- and epsilon-secretase-derived amyloid intracellular C-terminal domain (AICD) fragments (AICDC59 and AICDC50, respectively) of betaAPP trigger p53-dependent cell death and increase p53 activity and mRNA. Finally, PS1 mutations enhance p53 activity in human embryonic kidney 293 cells and p53 expression in FAD-affected brains. Thus our study shows that AICDs control p53 at a transcriptional level, in vitro and in vivo, and that FAD mutations increase p53 expression and activity in cells and human brains.
Collapse
|
19
|
Abe Y, Hashimoto Y, Tomita Y, Terashita K, Aiso S, Tajima H, Niikura T, Matsuoka M, Nishimoto I. Cytotoxic mechanisms by M239V presenilin 2, a little-analyzed Alzheimer's disease-causative mutant. J Neurosci Res 2004; 77:583-95. [PMID: 15264228 DOI: 10.1002/jnr.20163] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Although neurotoxic functions are well characterized in familial Alzheimer's disease (FAD)-linked N141I mutant of presenilin (PS)2, little has been known about M239V-PS2, another established FAD-causative mutant. We found that expression of M239V-PS2 caused neuronal cytotoxicity. M239V-PS2 exerted three forms of cytotoxicity: one was sensitive to both an antioxidant glutathione-ethyl-ester (GEE) and a caspase inhibitor Ac-DEVD-CHO (DEVD); the second was sensitive to GEE but resistant to DEVD; and the third was resistant to both. The GEE/DEVD-sensitive cytotoxicity by M239V-PS2 was likely through NADPH oxidase and the GEE-sensitive/DEVD-resistant cytotoxicity through xanthine oxidase (XO). Both mechanisms by M239V-PS2 were suppressed by pertussis toxin (PTX) and were mediated by Galpha(o), but not by Galpha(i). Although Abeta1-43 itself induced no cytotoxicity, Abeta1-43 potentiated all three components of M239V-PS2 cytotoxicity. As these cytotoxic mechanisms by M239V-PS2 are fully shared with N141I-PS2, they are most likely implicated in the pathomechanism of FAD by PS2 mutations. Notably, cytotoxicity by M239V-PS2 could be inhibited by the combination of two clinically usable inhibitors of superoxide-generating enzymes, apocynin and oxypurinol.
Collapse
Affiliation(s)
- Yoichiro Abe
- Department of Pharmacology, KEIO University School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Takahashi N, Kariya S, Hirano M, Ueno S. Two novel spliced presenilin 2 transcripts in human lymphocyte with oxidant stress and brain. Mol Cell Biochem 2004; 252:279-83. [PMID: 14577603 DOI: 10.1023/a:1025537021543] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
We identified two novel spliced human presenilin 2 (PS2) transcripts. The first, PS2deltaEx3-7, lacked part ofexon 3, all of exons 4, 5, and 6, and part of exon 7, resulting in an in-frame shift and inclusion of the natural start codon and proteolytic region. This transcript was detected in cerebral cortex and peripheral lymphocytes. The second transcript, PS2deltaEx4, lacked exon 4, resulting in a frame shift and inclusion of the natural start codon, and was transcribed in peripheral lymphocytes and heart but not in brain. Quantitative RT-PCR analysis revealed that the PS2deltaEx3-7 significantly increased in lymphocytes treated with H2O2, suggesting that this transcript is a novel genetic marker that can be used to study the pathogenesis of Alzheimer's disease.
Collapse
|
21
|
Tabira T. Alzheimer's disease: Mechanisms and development of therapeutic strategies. Geriatr Gerontol Int 2003. [DOI: 10.1111/j.1444-1586.2003.00082.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
22
|
Gamliel A, Teicher C, Hartmann T, Beyreuther K, Stein R. Overexpression of wild-type presenilin 2 or its familial Alzheimer's disease-associated mutant does not induce or increase susceptibility to apoptosis in different cell lines. Neuroscience 2003; 117:19-28. [PMID: 12605888 DOI: 10.1016/s0306-4522(02)00830-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Programmed cell death, or apoptosis, has been implicated in Alzheimer's disease. Mutations in the presenilin (PS) genes, PS1 and PS2, are a major cause of early-onset familial Alzheimer's disease (FAD). Previous studies have suggested that the PS play a role in apoptosis. However, the mechanisms whereby presenilins affect apoptosis and the relationship of FAD-associated presenilin mutants to the apoptotic effect have not been elucidated. In the present study, in an attempt to further explore the effect of PS2 on apoptosis we examined whether overexpression of wild-type or mutant PS2 can directly induce apoptosis or increase cell susceptibility to apoptosis in various cell lines, such as N2a, CHO, and HEK 293T. Wild-type or mutant PS2 was transiently transfected into these cell lines and the viability of the transfected cells was evaluated by their morphology, DNA fragmentation and condensation, appearance of sub-G(1/0) cells, and caspase activation. We also examined the susceptibility of the PS2-transfected cells to apoptosis induced by the apoptotic inducers staurosporine and H(2)O(2). Our results showed that overexpression of either wild type or mutant PS2 in these cell lines did not directly induce apoptosis or increase the susceptibility to apoptosis induced by staurosporine or H(2)O(2). Taken together, these results suggest that overexpression of PS2 does not cause pro-apoptotic effects, at least not in the cellular systems and conditions employed in this study, and therefore it seems unlikely that apoptosis plays a prominent role in the neuropathological effects of PS2 in Alzheimer's disease.
Collapse
Affiliation(s)
- A Gamliel
- Department of Neurobiochemistry, George S. Wise Faculty of Life Sciences, Tel-Aviv University, Israel
| | | | | | | | | |
Collapse
|
23
|
Niikura T, Hashimoto Y, Tajima H, Nishimoto I. Death and survival of neuronal cells exposed to Alzheimer's insults. J Neurosci Res 2002; 70:380-91. [PMID: 12391601 DOI: 10.1002/jnr.10354] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Neuronal cell death is the central abnormality occurring in brains suffering from Alzheimer's disease (AD). The notion that AD is a disease caused by loss of neurons points toward suppression of neuronal death as the most important therapeutic target. Nevertheless, the mechanisms for neuronal death in AD are still relatively unclear. Three known mutant genes cause familial AD (FAD): amyloid precursor protein, presenilin 1, and presenilin 2. Detailed analysis of cytotoxic mechanisms of the FAD-linked mutant genes reveals that they cause neuronal cell death at physiologically low expression levels. Unexpectedly, cytotoxic mechanisms vary depending on the type of mutations and genes, suggesting that various mechanisms for neuronal cell death are involved in AD patients. In support of this, activity-dependent neurotrophic factor, basic fibroblast growth factor, and insulin-like growth factor-I can completely protect neurons from beta-amyloid (A beta) cytotoxicity but exhibit incomplete or little effect on cytotoxicity by FAD mutant genes. By contrast, Humanin, a newly identified 24-residue peptide, suppresses neuronal cell death by various FAD mutants and A beta, whereas this factor has no effect on cytotoxicity from AD-irrelevant insults. Studies investigating death and survival of neuronal cells exposed to AD insults will open a new horizon in developing therapy aimed at neuroprotection.
Collapse
Affiliation(s)
- Takako Niikura
- Department of Pharmacology and Neurosciences, Keio University School of Medicine, Shinanomachi, Tokyo, Japan
| | | | | | | |
Collapse
|
24
|
Hashimoto Y, Niikura T, Ito Y, Kita Y, Terashita K, Nishimoto I. Neurotoxic mechanisms by Alzheimer's disease-linked N141I mutant presenilin 2. J Pharmacol Exp Ther 2002; 300:736-45. [PMID: 11861776 DOI: 10.1124/jpet.300.3.736] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Although it has been established that oxidative stress mediates cytotoxicity by familial Alzheimer's disease (FAD)-linked mutants of presenilin (PS)1 and that pertussis toxin inhibits cytotoxicity by FAD-linked N141I-PS2, it has not been determined whether oxidative stress is involved in cytotoxicity by N141I-PS2 or which pertussis toxin-sensitive proteins mediate the cytotoxicity. Here we report that low expression of N141I-PS2 caused neuronal cell death, whereas low expression of wild-type PS2 did not. Cytotoxicities by low and high expression of N141I-PS2 occurred through dissimilar mechanisms: the former cytotoxicity was blocked by a cell-permeable caspase inhibitor, and the latter was not. Since both mechanisms were sensitive to a cell-permeable antioxidant, we examined potential sources of reactive oxygen species in each mechanism, and found that the caspase inhibitor-sensitive neurotoxicity by N141I-PS2 was likely through NADPH oxidase and the caspase inhibitor-resistant neurotoxicity by N141I-PS2 through xanthine oxidase. Pertussis toxin greatly suppressed both toxic mechanisms by N141I-PS2, and only Galpha(o), a neuron-enriched pertussis toxin-sensitive G protein, was involved in both mechanisms. We therefore conclude that N141I-PS2 is capable of triggering multiple neurotoxic mechanisms, which can be inhibited by the combination of clinically usable inhibitors of NADPH oxidase and xanthine oxidase. This study thus provides a novel insight into the therapeutic intervention of PS2 mutant-associated FAD.
Collapse
Affiliation(s)
- Yuichi Hashimoto
- Department of Pharmacology and Neurosciences, KEIO University School of Medicine, Shinanomachi, Shinjuku-ku, Tokyo Japan
| | | | | | | | | | | |
Collapse
|
25
|
Abstract
Ageing is accompanied by a general decline of physiological function, especially at later stages, and significant increases in the incidence of cancer and other degenerative diseases. It has recently been hypothesized that alterations in apoptosis may contribute to these age-associated changes. However, whether there is a role for apoptosis in the ageing process and how ageing may modify the regulatory machinery of apoptosis remains obscure. Although the literature addressing these issues is scarce, research in this area is gaining momentum. Molecules involved in apoptosis signaling in mammals have been found to regulate ageing in organisms such as Caenorhabditis elegans and Drosophila melanogaster. Caloric restriction studies in a wide variety of organisms, ranging from yeast to mammals, suggest the conserved nature of the ageing regulatory systems. It seems very likely that signals that regulate ageing will impact apoptosis and the extent of apoptosis may then impact ageing. However, to date, there has been no direct evidence supporting the existence of such cross-communication between ageing and apoptosis in mammalian system. Here we review progress in the field.
Collapse
Affiliation(s)
- Yingpei Zhang
- Department of Cellular and Structural Biology, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | | |
Collapse
|
26
|
Araki W, Yuasa K, Takeda S, Takeda K, Shirotani K, Takahashi K, Tabira T. Pro-apoptotic effect of presenilin 2 (PS2) overexpression is associated with down-regulation of Bcl-2 in cultured neurons. J Neurochem 2001; 79:1161-8. [PMID: 11752057 DOI: 10.1046/j.1471-4159.2001.00638.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Presenilin 2 (PS2) is a polytopic membrane protein that is mutated in some cases of familial Alzheimer's disease (AD). The normal functions of PS2 and its pathogenic role in AD remain unclear. We investigated the biological role of this protein in neurons, using adenovirus-mediated transduction of the PS2 gene into rat primary cortical neurons. Immunocytochemical analyses demonstrated increased PS2 immunoreactivity in most neurons infected with recombinant adenoviruses expressing PS2. Neurons infected with wild-type or mutant (N141I) PS2-expressing adenoviruses showed a significant increase in basal cell death, compared with those infected with control beta-galactosidase-expressing adenovirus. Moreover, PS2 overexpression markedly increased neuronal susceptibility to staurosporine-induced apoptosis. Mutant PS2 was more effective in enhancing apoptosis than its wild-type counterpart. Staurosporine-induced death was significantly inhibited by a specific caspase 3 inhibitor. Western analyses revealed that Bcl-2 protein expression was specifically down-regulated in neurons overexpressing PS2, which temporally corresponded to the accumulation of C- and N-terminal fragments of PS2. Additionally, expression of mutant, but not wild-type PS2, increased the production of beta-amyloid protein (Abeta) 42. These data collectively suggest that the pro-apoptotic effect of PS2 is mediated by down-regulation of Bcl-2. PS2 mutations may increase the susceptibility of neurons to apoptotic stimuli by perturbing the regulation of cell death.
Collapse
Affiliation(s)
- W Araki
- Division of Demyelinating Disease and Aging, National Institute of Neuroscience, NCNP, Kodaira, Tokyo, Japan.
| | | | | | | | | | | | | |
Collapse
|