101
|
Zagaja M, Zagaja A, Szala-Rycaj J, Szewczyk A, Lemieszek MK, Raszewski G, Andres-Mach M. Influence of Umbelliferone on the Anticonvulsant and Neuroprotective Activity of Selected Antiepileptic Drugs: An In Vivo and In Vitro Study. Int J Mol Sci 2022; 23:ijms23073492. [PMID: 35408852 PMCID: PMC8999126 DOI: 10.3390/ijms23073492] [Citation(s) in RCA: 9] [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: 02/19/2022] [Revised: 03/17/2022] [Accepted: 03/21/2022] [Indexed: 02/04/2023] Open
Abstract
Umbelliferone (7-hydroxycoumarin; UMB) is a coumarin with many biological properties, including antiepileptic activity. This study evaluated the effect of UMB on the ability of classical and novel antiepileptic drugs (e.g., lacosamide (LCM), levetiracetam (LEV), phenobarbital (PB) and valproate (VPA)) to prevent seizures evoked by the 6-Hz corneal-stimulation-induced seizure model. The study also evaluated the influence of this coumarin on the neuroprotective properties of these drugs in two in vitro models of neurodegeneration, including trophic stress and excitotoxicity. The results indicate that UMB (100 mg/kg, i.p.) significantly enhanced the anticonvulsant action of PB (p < 0.01) and VPA (p < 0.05), but not that of LCM orLEV, in the 6-Hz test. Whether alone or in combination with other anticonvulsant drugs (at their ED50 values from the 6-Hz test), UMB (100 mg/kg) did not affect motor coordination; skeletal muscular strength and long-term memory, as determined in the chimney; grip strength; or passive avoidance tests, respectively. Pharmacokinetic characterization revealed that UMB had no impact on total brain concentrations of PB or VPA in mice. The in vitro study indicated that UMB has neuroprotective properties. Administration of UMB (1 µg/mL), together with antiepileptic drugs, mitigated their negative impact on neuronal viability. Under trophic stress (serum deprivation) conditions, UMB enhanced the neurotrophic abilities of all the drugs used. Moreover, this coumarin statistically enhanced the neuroprotective effects of PB (p < 0.05) and VPA (p < 0.001) in the excitotoxicity model of neurodegeneration. The obtained results clearly indicate a positive effect of UMB on the anticonvulsant and neuroprotective properties of the selected drugs.
Collapse
Affiliation(s)
- Mirosław Zagaja
- Isobolographic Analysis Laboratory, Institute of Rural Health, Jaczewskiego 2, 20-090 Lublin, Poland; (J.S.-R.); (A.S.); (M.A.-M.)
- Correspondence: (M.Z.); (A.Z.); Tel.: +48-81-718-4549 (M.Z.); +48-81-448-6850 (A.Z.)
| | - Anna Zagaja
- Chair and Department of Humanities and Social Medicine, Medical University of Lublin, Chodźki 7, 20-090 Lublin, Poland
- Correspondence: (M.Z.); (A.Z.); Tel.: +48-81-718-4549 (M.Z.); +48-81-448-6850 (A.Z.)
| | - Joanna Szala-Rycaj
- Isobolographic Analysis Laboratory, Institute of Rural Health, Jaczewskiego 2, 20-090 Lublin, Poland; (J.S.-R.); (A.S.); (M.A.-M.)
| | - Aleksandra Szewczyk
- Isobolographic Analysis Laboratory, Institute of Rural Health, Jaczewskiego 2, 20-090 Lublin, Poland; (J.S.-R.); (A.S.); (M.A.-M.)
| | - Marta Kinga Lemieszek
- Department of Medical Biology, Institute of Rural Health, Jaczewskiego 2, 20-090 Lublin, Poland;
| | - Grzegorz Raszewski
- Department of Toxicology and Food Protection, Institute of Rural Health, Jaczewskiego 2, 20-950 Lublin, Poland;
| | - Marta Andres-Mach
- Isobolographic Analysis Laboratory, Institute of Rural Health, Jaczewskiego 2, 20-090 Lublin, Poland; (J.S.-R.); (A.S.); (M.A.-M.)
| |
Collapse
|
102
|
Davies AK, Alecu JE, Ziegler M, Vasilopoulou CG, Merciai F, Jumo H, Afshar-Saber W, Sahin M, Ebrahimi-Fakhari D, Borner GHH. AP-4-mediated axonal transport controls endocannabinoid production in neurons. Nat Commun 2022; 13:1058. [PMID: 35217685 PMCID: PMC8881493 DOI: 10.1038/s41467-022-28609-w] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 01/08/2022] [Indexed: 01/20/2023] Open
Abstract
The adaptor protein complex AP-4 mediates anterograde axonal transport and is essential for axon health. AP-4-deficient patients suffer from a severe neurodevelopmental and neurodegenerative disorder. Here we identify DAGLB (diacylglycerol lipase-beta), a key enzyme for generation of the endocannabinoid 2-AG (2-arachidonoylglycerol), as a cargo of AP-4 vesicles. During normal development, DAGLB is targeted to the axon, where 2-AG signalling drives axonal growth. We show that DAGLB accumulates at the trans-Golgi network of AP-4-deficient cells, that axonal DAGLB levels are reduced in neurons from a patient with AP-4 deficiency, and that 2-AG levels are reduced in the brains of AP-4 knockout mice. Importantly, we demonstrate that neurite growth defects of AP-4-deficient neurons are rescued by inhibition of MGLL (monoacylglycerol lipase), the enzyme responsible for 2-AG hydrolysis. Our study supports a new model for AP-4 deficiency syndrome in which axon growth defects arise through spatial dysregulation of endocannabinoid signalling. Davies et al. identify a putative mechanism underlying the childhood neurological disorder AP-4 deficiency syndrome. In the absence of AP-4, an enzyme that makes 2-AG is not transported to the axon, leading to axonal growth defects, which can be rescued by inhibition of 2-AG breakdown.
Collapse
Affiliation(s)
- Alexandra K Davies
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, 82152, Germany.
| | - Julian E Alecu
- Department of Neurology, The F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Marvin Ziegler
- Department of Neurology, The F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.,Department of Functional Neuroanatomy, Institute of Anatomy and Cell Biology, Heidelberg University, INF 307, Heidelberg, 69120, Germany
| | - Catherine G Vasilopoulou
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, 82152, Germany
| | - Fabrizio Merciai
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, 82152, Germany.,Department of Pharmacy and PhD Program in Drug Discovery and Development, University of Salerno, 84084, Fisciano, SA, Italy
| | - Hellen Jumo
- Department of Neurology, The F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Wardiya Afshar-Saber
- Department of Neurology, The F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Mustafa Sahin
- Department of Neurology, The F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.,Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Darius Ebrahimi-Fakhari
- Department of Neurology, The F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Georg H H Borner
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, 82152, Germany.
| |
Collapse
|
103
|
El-Said WA, Qaisi RM, Placide V, Choi JW. A stable naked-eye colorimetric sensor for monitoring release of extracellular gamma-aminobutyric acid (GABA) neurotransmitter from SH-SY5Y cells. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2022; 267:120517. [PMID: 34739892 DOI: 10.1016/j.saa.2021.120517] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 10/04/2021] [Accepted: 10/14/2021] [Indexed: 06/13/2023]
Abstract
A novel optical γ-aminobutyric acid (GABA)-based sensor was developed on interacting thiol compounds and o-phthalaldehyde (OPA) to form thiacetal compounds. Then, the thiacetal interacts with the GABA molecule to form an isoindole compound. The effects of four thiol compounds on the stability of the resulting isoindole compound were assessed. The 2-mercaptoethanol, "one of the most used derivatizing agents," is unexpectedly the least stable; while, 16-mercaptohexadecanoic acid resulted in the most durable isoindole compound. The developed sensor showed the capability for detecting GABA within a wide concentration range spanning from 500 nmol L-1 to 100 µmol L-1. The detection limit was about 330 nmol L-1, which indicated the high sensitivity of the developed sensor compared with those previously reported. The findings illustrated the ability to detect GABA at the physiological pH (pH = 7.4) without adjusting the pH value, opening the door for real applications. Furthermore, the sensor could detect various GABA concentrations in human serum with good recovery percentages (98% to 101.4%). In addition, this assay was applied to monitor GABA release from the SH-SY5Y cell line to convert glutamate into GABA. This result indicates the capability of the proposed assay for visually monitoring the release of GABA neurotransmitters.
Collapse
Affiliation(s)
- Waleed A El-Said
- Department of Chemistry, College of Science, University of Jeddah, P.O. Box 80327, Jeddah 21589, Saudi Arabia; Department of Chemical and Biomolecular Engineering, Sogang University, 35 Baekbeom-Ro, Mapo-Gu, Seoul 121-742, Republic of Korea; Department of Chemistry, Faculty of Science, Assiut University, Assiut 71516, Egypt
| | - Ramy M Qaisi
- University of Jeddah, College of Engineering, Department of Electrical and Electronic Engineering, P.O. Box 80327, Jeddah 21589, Saudi Arabia
| | - Virginie Placide
- Department of Chemical and Biomolecular Engineering, Sogang University, 35 Baekbeom-Ro, Mapo-Gu, Seoul 121-742, Republic of Korea
| | - Jeong-Woo Choi
- Department of Chemical and Biomolecular Engineering, Sogang University, 35 Baekbeom-Ro, Mapo-Gu, Seoul 121-742, Republic of Korea.
| |
Collapse
|
104
|
Retinoic Acid-Differentiated Neuroblastoma SH-SY5Y Is an Accessible In Vitro Model to Study Native Human Acid-Sensing Ion Channels 1a (ASIC1a). BIOLOGY 2022; 11:biology11020167. [PMID: 35205034 PMCID: PMC8868828 DOI: 10.3390/biology11020167] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 01/16/2022] [Accepted: 01/17/2022] [Indexed: 12/19/2022]
Abstract
Simple Summary Human neuroblastoma SH-SY5Y is used in neurobiology for studying various neuropathophysiological processes. In this study, we differentiated neuroblastoma cells into a neuronal-like phenotype with retinoic acid and studied if functional acid-sensing, transient receptor potential vanilloid-1 and ankyrin-1 ion channels were expressed in it. We found that homomeric acid-sensing ion channels 1a were expressed predominantly and yielded large ionic currents that can be modulated with different ligands. This channel plays important roles in synaptic plasticity, neurodegeneration, and pain perception. Thus, retinoic acid-treated neuroblastoma is a suitable model system for pharmacological testing on native human acid-sensing ion channels 1a. This approach can facilitate the development of new drugs for neuroprotection and pain management. Abstract Human neuroblastoma SH-SY5Y is a prominent neurobiological tool used for studying neuropathophysiological processes. We investigated acid-sensing (ASIC) and transient receptor potential vanilloid-1 (TRPV1) and ankyrin-1 (TRPA1) ion channels present in untreated and differentiated neuroblastoma SH-SY5Y to propose a new means for their study in neuronal-like cells. Using a quantitative real-time PCR and a whole-cell patch-clamp technique, ion channel expression profiles, functionality, and the pharmacological actions of their ligands were characterized. A low-level expression of ASIC1a and ASIC2 was detected in untreated cells. The treatment with 10 μM of retinoic acid (RA) for 6 days resulted in neuronal differentiation that was accompanied by a remarkable increase in ASIC1a expression, while ASIC2 expression remained almost unaltered. In response to acid stimuli, differentiated cells showed prominent ASIC-like currents. Detailed kinetic and pharmacological characterization suggests that homomeric ASIC1a is a dominant isoform among the present ASIC channels. RA-treatment also reduced the expression of TRPV1 and TRPA1, and minor electrophysiological responses to their agonists were found in untreated cells. Neuroblastoma SH-SY5Y treated with RA can serve as a model system to study the effects of different ligands on native human ASIC1a in neuronal-like cells. This approach can improve the characterization of modulators for the development of new neuroprotective and analgesic drugs.
Collapse
|
105
|
Zhang LN, Li MJ, Shang YH, Liu YR, Han-Chang H, Lao FX. Zeaxanthin Attenuates the Vicious Circle Between Endoplasmic Reticulum Stress and Tau Phosphorylation: Involvement of GSK-3β Activation. J Alzheimers Dis 2022; 86:191-204. [PMID: 35034906 DOI: 10.3233/jad-215408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) characterized by neurofibrillary tangles caused by hyperphosphorylated tau is the most common cause of dementia. Zeaxanthin (Zea), derived from fruits and vegetables, may reduce the risk of AD. Endoplasmic reticulum stress (ERS) might cause memory impairment in AD. OBJECTIVE Here, we studied protective role of Zea on the relationship among ERS, activity of glycogen synthase kinase 3β (GSK-3β, tau phosphorylated kinase), and p-Tau (Ser 396 and Thr 231). METHODS The results were obtained in non-RA and RA group by using different treatment, such as 9-cis-retinoic acid (RA), TM (ERS inducer), Zea, 4-PBA (ERS inhibitor), and SB216763 (GSK-3β inhibitor). The methods included flow cytometry and MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] for the detections of cell cycle and cell viability and western blot as a third measure of proteins in relation to ERS and tau phosphorylation. We have collected and analyzed all the data that suggested application of drugs for the treatment in non-RA and RA group. RESULTS Zea displays its protection on TM-induced cell injury, upregulation of GRP78 expression, and change of GSK-3β activity and tau phosphorylation when 4-PBA and SB216763 interfere with the process. CONCLUSION These studies indicated that Zea is in vicious circle in ERS, GSK-3β, and tau phosphorylation, and further reflect its potential value in AD.
Collapse
Affiliation(s)
- Li-Na Zhang
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, P.R. China.,Institute of Functional Factors and Brain Science, Beijing Union University, Beijing, P.R. China.,College of Biochemical Engineering, Beijing Union University, Beijing, P.R. China
| | - Meng-Jie Li
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, P.R. China.,Institute of Functional Factors and Brain Science, Beijing Union University, Beijing, P.R. China.,College of Biochemical Engineering, Beijing Union University, Beijing, P.R. China
| | - Ying-Hui Shang
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, P.R. China.,Institute of Functional Factors and Brain Science, Beijing Union University, Beijing, P.R. China.,College of Biochemical Engineering, Beijing Union University, Beijing, P.R. China
| | - Yun-Ru Liu
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, P.R. China.,Institute of Functional Factors and Brain Science, Beijing Union University, Beijing, P.R. China.,College of Biochemical Engineering, Beijing Union University, Beijing, P.R. China
| | - Huang Han-Chang
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, P.R. China.,Institute of Functional Factors and Brain Science, Beijing Union University, Beijing, P.R. China.,College of Biochemical Engineering, Beijing Union University, Beijing, P.R. China
| | - Feng-Xue Lao
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, P.R. China.,Institute of Functional Factors and Brain Science, Beijing Union University, Beijing, P.R. China.,College of Biochemical Engineering, Beijing Union University, Beijing, P.R. China
| |
Collapse
|
106
|
In Vitro Methodologies to Study the Role of Advanced Glycation End Products (AGEs) in Neurodegeneration. Nutrients 2022; 14:nu14020363. [PMID: 35057544 PMCID: PMC8777776 DOI: 10.3390/nu14020363] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 02/07/2023] Open
Abstract
Advanced glycation end products (AGEs) can be present in food or be endogenously produced in biological systems. Their formation has been associated with chronic neurodegenerative diseases such as Alzheimer’s disease, Parkinson’s disease, multiple sclerosis, and amyotrophic lateral sclerosis. The implication of AGEs in neurodegeneration is related to their ability to bind to AGE-specific receptors and the ability of their precursors to induce the so-called “dicarbonyl stress”, resulting in cross-linking and protein damage. However, the mode of action underlying their role in neurodegeneration remains unclear. While some research has been carried out in observational clinical studies, further in vitro studies may help elucidate these underlying modes of action. This review presents and discusses in vitro methodologies used in research on the potential role of AGEs in neuroinflammation and neurodegeneration. The overview reveals the main concepts linking AGEs to neurodegeneration, the current findings, and the available and advisable in vitro models to study their role. Moreover, the major questions regarding the role of AGEs in neurodegenerative diseases and the challenges and discrepancies in the research field are discussed.
Collapse
|
107
|
Tsukui R, Yamamoto T, Okamura Y, Kato Y, Shibata N. Fukutin regulates tau phosphorylation and synaptic function: Novel properties of fukutin in neurons. Neuropathology 2022; 42:28-39. [PMID: 35026860 PMCID: PMC9305503 DOI: 10.1111/neup.12797] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 11/20/2021] [Accepted: 11/20/2021] [Indexed: 12/13/2022]
Abstract
Fukutin, a product of the causative gene of Fukuyama congenital muscular dystrophy (FCMD), is known to be responsible for basement membrane formation. Patients with FCMD exhibit not only muscular dystrophy but also central nervous system abnormalities, including polymicrogyria and neurofibrillary tangles (NFTs) in the cerebral cortex. The formation of NFTs cannot be explained by basement membrane disorganization. To determine the involvement of fukutin in the NFT formation, we performed molecular pathological investigations using autopsied human brains and cultured neurons of a cell line (SH-SY5Y). In human brains, NFTs, identified with an antibody against phosphorylated tau (p-tau), were observed in FCMD patients but not age-matched control subjects and were localized in cortical neurons lacking somatic immunoreactivity for glutamic acid decarboxylase (GAD), a marker of inhibitory neurons. In FCMD brains, NFTs were mainly distributed in lesions of polymicrogyria. Immunofluorescence staining revealed the colocalization of immunoreactivities for p-tau and phosphorylated glycogen synthase kinase-3β (GSK-3β), a potential tau kinase, in the somatic cytoplasm of SH-SY5Y cells; both the immunoreactivities were increased by fukutin knockdown and reduced by fukutin overexpression. Western blot analysis using SH-SY5Y cells revealed consistent results. Enzyme-linked immunosorbent assay (ELISA) confirmed the binding affinity of fukutin to tau and GSK-3β in SH-SY5Y cells. In the human brains, the density of GAD-immunoreactive neurons in the frontal cortex was significantly higher in the FCMD group than in the control group. GAD immunoreactivity on Western blots of SH-SY5Y cells was significantly increased by fukutin knockdown. On immunofluorescence staining, immunoreactivities for fukutin and GAD were colocalized in the somatic cytoplasm of the human brains and SH-SY5Y cells, whereas those for fukutin and synaptophysin were colocalized in the neuropil of the human brains and the cytoplasm of SH-SY5Y cells. ELISA confirmed the binding affinity of fukutin to GAD and synaptophysin in SH-SY5Y cells. The present results provide in vivo and in vitro evidence for novel properties of fukutin as follows: (i) there is an inverse relationship between fukutin expression and GSK-3β/tau phosphorylation in neurons; (ii) fukutin binds to GSK-3β and tau; (iii) tau phosphorylation occurs in non-GAD-immunoreactive neurons in FCMD brains; (iv) neuronal GAD expression is upregulated in the absence of fukutin; and (v) fukutin binds to GAD and synaptophysin in presynaptic vesicles of neurons.
Collapse
Affiliation(s)
- Ryota Tsukui
- Graduate School of Medicine, Tokyo Women's Medical University, Tokyo, Japan.,Division of Human Pathology & Pathological Neuroscience, Department of Pathology, Tokyo Women's Medical University, Tokyo, Japan
| | - Tomoko Yamamoto
- Division of Human Pathology & Pathological Neuroscience, Department of Pathology, Tokyo Women's Medical University, Tokyo, Japan.,Department of Surgical Pathology, Tokyo Women's Medical University, Tokyo, Japan
| | - Yukinori Okamura
- Graduate School of Medicine, Tokyo Women's Medical University, Tokyo, Japan.,Division of Human Pathology & Pathological Neuroscience, Department of Pathology, Tokyo Women's Medical University, Tokyo, Japan
| | - Yoichiro Kato
- Division of Human Pathology & Pathological Neuroscience, Department of Pathology, Tokyo Women's Medical University, Tokyo, Japan
| | - Noriyuki Shibata
- Division of Human Pathology & Pathological Neuroscience, Department of Pathology, Tokyo Women's Medical University, Tokyo, Japan.,Department of Surgical Pathology, Tokyo Women's Medical University, Tokyo, Japan
| |
Collapse
|
108
|
TREX1 Deficiency Induces ER Stress-Mediated Neuronal Cell Death by Disrupting Ca 2+ Homeostasis. Mol Neurobiol 2022; 59:1398-1418. [PMID: 34997539 PMCID: PMC8882114 DOI: 10.1007/s12035-021-02631-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 11/01/2021] [Indexed: 11/09/2022]
Abstract
TREX1 is an exonuclease that degrades extranuclear DNA species in mammalian cells. Herein, we show a novel mechanism by which TREX1 interacts with the BiP/GRP78 and TREX1 deficiency triggers ER stress through the accumulation of single-stranded DNA and activates unfolded protein response (UPR) signaling via the disruption of the TREX1-BiP/GRP78 interaction. In TREX1 knockdown cells, the activation of ER stress signaling disrupted ER Ca2+ homeostasis via the ERO1α-IP3R1-CaMKII pathway, leading to neuronal cell death. Moreover, TREX1 knockdown dysregulated the Golgi-microtubule network through Golgi fragmentation and decreased Ac-α-tubulin levels, contributing to neuronal injury. These alterations were also observed in neuronal cells harboring a TREX1 mutation (V91M) that has been identified in hereditary spastic paraplegia (HSP) patients in Korea. Notably, this mutation leads to defects in the TREX1-BiP/GRP78 interaction and mislocalization of TREX1 from the ER and possible disruption of the Golgi-microtubule network. In summary, the current study reveals TREX1 as a novel regulator of the BiP/GRP78 interaction and shows that TREX1 deficiency promotes ER stress-mediated neuronal cell death, which indicates that TREX1 may hold promise as a therapeutic target for neurodegenerative diseases such as HSP.
Collapse
|
109
|
Annuario E, Ng K, Vagnoni A. High-Resolution Imaging of Mitochondria and Mitochondrial Nucleoids in Differentiated SH-SY5Y Cells. Methods Mol Biol 2022; 2431:291-310. [PMID: 35412283 DOI: 10.1007/978-1-0716-1990-2_15] [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] [Indexed: 10/18/2022]
Abstract
Mitochondria are highly dynamic organelles which form intricate networks with complex dynamics. Mitochondrial transport and distribution are essential to ensure proper cell function, especially in cells with an extremely polarised morphology such as neurons. A layer of complexity is added when considering mitochondria have their own genome, packaged into nucleoids. Major mitochondrial morphological transitions, for example mitochondrial division, often occur in conjunction with mitochondrial DNA (mtDNA) replication and changes in the dynamic behaviour of the nucleoids. However, the relationship between mtDNA dynamics and mitochondrial motility in the processes of neurons has been largely overlooked. In this chapter, we describe a method for live imaging of mitochondria and nucleoids in differentiated SH-SY5Y cells by instant structured illumination microscopy (iSIM). We also include a detailed protocol for the differentiation of SH-SY5Y cells into cells with a pronounced neuronal-like morphology and show examples of coordinated mitochondrial and nucleoid motility in the long processes of these cells.
Collapse
Affiliation(s)
- Emily Annuario
- Department of Basic and Clinical Neurosciences, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Kristal Ng
- Department of Basic and Clinical Neurosciences, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Alessio Vagnoni
- Department of Basic and Clinical Neurosciences, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
| |
Collapse
|
110
|
Feng J, Song G, Shen Q, Chen X, Wang Q, Guo S, Zhang M. Protect Effects of Seafood-Derived Plasmalogens Against Amyloid-Beta (1-42) Induced Toxicity via Modulating the Transcripts Related to Endocytosis, Autophagy, Apoptosis, Neurotransmitter Release and Synaptic Transmission in SH-SY5Y Cells. Front Aging Neurosci 2021; 13:773713. [PMID: 34899276 PMCID: PMC8662987 DOI: 10.3389/fnagi.2021.773713] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 11/01/2021] [Indexed: 11/13/2022] Open
Abstract
To investigate the underlying mechanisms of decreased plasmalogens (Pls) levels in neurodegenerative diseases, here the effects of seafood-derived Pls on undifferentiated and differentiated human SH-SY5Y neuroblastoma cells exposed to amyloid-β1-42 was analyzed. Transcriptional profiles indicated that a total of 6,581 differentially expressed genes (DEGs) were significantly identified among different experimental groups, and KEGG analysis indicated that these DEGs were related to AD, endocytosis, synaptic vesicle cycle, autophagy and cellular apoptosis. After Pls treatment, the striking expression changes of ADORA2A, ATP6V1C2, CELF6, and SLC18A2 mRNA strongly suggest that Pls exerts a beneficial role in alleviating AD pathology partly by modulating the neurotransmitter release and synaptic transmission at the transcriptional level. Besides these, GPCRs are also broadly involved in Pls-signaling in neuronal cells. These results provide evidence for supporting the potential use of Pls as an effective therapeutic approach for AD.
Collapse
Affiliation(s)
- Junli Feng
- Zhejiang Province Joint Key Laboratory of Aquatic Products Processing, Collaborative Innovation Center of Seafood Deep Processing, Institute of Seafood, Zhejiang Gongshang University, Hangzhou, China
| | - Gongshuai Song
- Zhejiang Province Joint Key Laboratory of Aquatic Products Processing, Collaborative Innovation Center of Seafood Deep Processing, Institute of Seafood, Zhejiang Gongshang University, Hangzhou, China
| | - Qing Shen
- Zhejiang Province Joint Key Laboratory of Aquatic Products Processing, Collaborative Innovation Center of Seafood Deep Processing, Institute of Seafood, Zhejiang Gongshang University, Hangzhou, China
| | - Xi Chen
- Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Qingcheng Wang
- Department of Cardiology, Hangzhou Linping Hospital of Traditional Chinese Medicine, Hangzhou, China
| | - Shunyuan Guo
- Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Manman Zhang
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
111
|
Optimised techniques for high-throughput screening of differentiated SH-SY5Y cells and application for neurite outgrowth assays. Sci Rep 2021; 11:23935. [PMID: 34907283 PMCID: PMC8671469 DOI: 10.1038/s41598-021-03442-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 12/01/2021] [Indexed: 11/16/2022] Open
Abstract
Neuronal models are a crucial tool in neuroscientific research, helping to elucidate the molecular and cellular processes involved in disorders of the nervous system. Adapting these models to a high-throughput format enables simultaneous screening of multiple agents within a single assay. SH-SY5Y cells have been widely used as a neuronal model, yet commonly in an undifferentiated state that is not representative of mature neurons. Differentiation of the SH-SY5Y cells is a necessary step to obtain cells that express mature neuronal markers. Despite this understanding, the absence of a standardised protocol has limited the use of differentiated SH-SY5Y cells in high-throughput assay formats. Here, we describe techniques to differentiate and re-plate SH-SY5Y cells within a 96-well plate for high-throughput screening. SH-SY5Y cells seeded at an initial density of 2,500 cells/well in a 96-well plate provide sufficient space for neurites to extend, without impacting cell viability. Room temperature pre-incubation for 1 h improved the plating homogeneity within the well and the ability to analyse neurites. We then demonstrated the efficacy of our techniques by optimising it further for neurite outgrowth analysis. The presented methods achieve homogenously distributed differentiated SH-SY5Y cells, useful for researchers using these cells in high-throughput screening assays.
Collapse
|
112
|
Wang XX, Xie F, Jia CC, Yan N, Zeng YL, Wu JD, Liu ZP. Synthesis and biological evaluation of selective histone deacetylase 6 inhibitors as multifunctional agents against Alzheimer's disease. Eur J Med Chem 2021; 225:113821. [PMID: 34517222 DOI: 10.1016/j.ejmech.2021.113821] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/30/2021] [Accepted: 08/31/2021] [Indexed: 01/08/2023]
Abstract
Histone deacetylase 6 (HDAC6) is a potential target for Alzheimer's disease (AD). In this study, a series of novel phenothiazine-, memantine-, and 1,2,3,4-tetrahydro-γ-carboline-based HDAC6 inhibitors with a variety of linker moieties were designed and synthesized. As a hydrochloride salt, the phenothiazine-based hydroxamic acid W5 with a pyridyl-containing linker motif was identified as a high potent and selective HDAC6 inhibitor. It inhibited HDAC6 with an IC50 of 2.54 nM and was more than 290- to 3300-fold selective over other HDAC isoforms. In SH-SY5Y cells, W5 dose-dependently increased the acetylated α-tubulin levels and reduced the hyperphosphorylated tau proteins at Ser396. As an effective metal chelator, W5 inhibited Cu2+-induced Aβ1-42 aggregation and disaggregated Cu2+-Aβ1-42 oligomers, and showed protective effects on the SH-SY5Y cells against Aβ1-42- as well as Cu2+-Aβ1-42 induced cell damages, serving as a potential ligand to target AD metal dyshomeostasis. Moreover, W5 promoted the differentiated neuronal neurite outgrowth, increased the mRNA expression of the recognized neurogenesis markers, GAP43, N-myc, and MAP-2. Therefore, W5 might be a good lead for the development of novel HDAC6 inhibitors targeting multi-facets of AD.
Collapse
Affiliation(s)
- Xiu-Xiu Wang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China
| | - Fei Xie
- Department of Pharmacy, Qilu Hospital of Shandong University, Jinan, 250012, PR China
| | - Cong-Cong Jia
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China
| | - Ning Yan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China
| | - Yan-Li Zeng
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China
| | - Jing-De Wu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China.
| | - Zhao-Peng Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China.
| |
Collapse
|
113
|
Rathnayake AU, Abuine R, Palanisamy S, Lee JK, Byun HG. Characterization and purification of β−secretase inhibitory peptides fraction from sea cucumber (Holothuria spinifera) enzymatic hydrolysates. Process Biochem 2021. [DOI: 10.1016/j.procbio.2021.10.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
114
|
Steinberg DJ, Aqeilan RI. WWOX-Related Neurodevelopmental Disorders: Models and Future Perspectives. Cells 2021; 10:cells10113082. [PMID: 34831305 PMCID: PMC8623516 DOI: 10.3390/cells10113082] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/28/2021] [Accepted: 11/03/2021] [Indexed: 12/12/2022] Open
Abstract
The WW domain-containing oxidoreductase (WWOX) gene was originally discovered as a putative tumor suppressor spanning the common fragile site FRA16D, but as time has progressed the extent of its pleiotropic function has become apparent. At present, WWOX is a major source of interest in the context of neurological disorders, and more specifically developmental and epileptic encephalopathies (DEEs). This review article aims to introduce the many model systems used through the years to study its function and roles in neuropathies. Similarities and fundamental differences between rodent and human models are discussed. Finally, future perspectives and promising research avenues are suggested.
Collapse
|
115
|
Pauls E, Bayod S, Mateo L, Alcalde V, Juan-Blanco T, Sánchez-Soto M, Saido TC, Saito T, Berrenguer-Llergo A, Attolini CSO, Gay M, de Oliveira E, Duran-Frigola M, Aloy P. Identification and drug-induced reversion of molecular signatures of Alzheimer's disease onset and progression in App NL-G-F, App NL-F, and 3xTg-AD mouse models. Genome Med 2021; 13:168. [PMID: 34702310 PMCID: PMC8547095 DOI: 10.1186/s13073-021-00983-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 09/29/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND In spite of many years of research, our understanding of the molecular bases of Alzheimer's disease (AD) is still incomplete, and the medical treatments available mainly target the disease symptoms and are hardly effective. Indeed, the modulation of a single target (e.g., β-secretase) has proven to be insufficient to significantly alter the physiopathology of the disease, and we should therefore move from gene-centric to systemic therapeutic strategies, where AD-related changes are modulated globally. METHODS Here we present the complete characterization of three murine models of AD at different stages of the disease (i.e., onset, progression and advanced). We combined the cognitive assessment of these mice with histological analyses and full transcriptional and protein quantification profiling of the hippocampus. Additionally, we derived specific Aβ-related molecular AD signatures and looked for drugs able to globally revert them. RESULTS We found that AD models show accelerated aging and that factors specifically associated with Aβ pathology are involved. We discovered a few proteins whose abundance increases with AD progression, while the corresponding transcript levels remain stable, and showed that at least two of them (i.e., lfit3 and Syt11) co-localize with Aβ plaques in the brain. Finally, we found two NSAIDs (dexketoprofen and etodolac) and two anti-hypertensives (penbutolol and bendroflumethiazide) that overturn the cognitive impairment in AD mice while reducing Aβ plaques in the hippocampus and partially restoring the physiological levels of AD signature genes to wild-type levels. CONCLUSIONS The characterization of three AD mouse models at different disease stages provides an unprecedented view of AD pathology and how this differs from physiological aging. Moreover, our computational strategy to chemically revert AD signatures has shown that NSAID and anti-hypertensive drugs may still have an opportunity as anti-AD agents, challenging previous reports.
Collapse
Affiliation(s)
- Eduardo Pauls
- Joint IRB-BSC-CRG Programme in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Catalonia, Spain
| | - Sergi Bayod
- Joint IRB-BSC-CRG Programme in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Catalonia, Spain
| | - Lídia Mateo
- Joint IRB-BSC-CRG Programme in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Catalonia, Spain
| | - Víctor Alcalde
- Joint IRB-BSC-CRG Programme in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Catalonia, Spain
| | - Teresa Juan-Blanco
- Joint IRB-BSC-CRG Programme in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Catalonia, Spain
| | - Marta Sánchez-Soto
- Joint IRB-BSC-CRG Programme in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Catalonia, Spain
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Saitama, 351-0198, Japan
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Antoni Berrenguer-Llergo
- Biostatistics and Bioinformatics Unit, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Catalonia, Spain
| | - Camille Stephan-Otto Attolini
- Biostatistics and Bioinformatics Unit, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Catalonia, Spain
| | - Marina Gay
- Proteomics Unit, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Catalonia, Spain
| | | | - Miquel Duran-Frigola
- Joint IRB-BSC-CRG Programme in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Catalonia, Spain
| | - Patrick Aloy
- Joint IRB-BSC-CRG Programme in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Catalonia, Spain.
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Catalonia, Spain.
| |
Collapse
|
116
|
Mallach A, Gobom J, Arber C, Piers TM, Hardy J, Wray S, Zetterberg H, Pocock J. Differential Stimulation of Pluripotent Stem Cell-Derived Human Microglia Leads to Exosomal Proteomic Changes Affecting Neurons. Cells 2021; 10:cells10112866. [PMID: 34831089 PMCID: PMC8616378 DOI: 10.3390/cells10112866] [Citation(s) in RCA: 2] [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: 06/02/2021] [Revised: 10/07/2021] [Accepted: 10/13/2021] [Indexed: 01/22/2023] Open
Abstract
Microglial exosomes are an emerging communication pathway, implicated in fulfilling homeostatic microglial functions and transmitting neurodegenerative signals. Gene variants of triggering receptor expressed on myeloid cells-2 (TREM2) are associated with an increased risk of developing dementia. We investigated the influence of the TREM2 Alzheimer’s disease risk variant, R47Hhet, on the microglial exosomal proteome consisting of 3019 proteins secreted from human iPS-derived microglia (iPS-Mg). Exosomal protein content changed according to how the iPS-Mg were stimulated. Thus lipopolysaccharide (LPS) induced microglial exosomes to contain more inflammatory signals, whilst stimulation with the TREM2 ligand phosphatidylserine (PS+) increased metabolic signals within the microglial exosomes. We tested the effect of these exosomes on neurons and found that the exosomal protein changes were functionally relevant and influenced downstream functions in both neurons and microglia. Exosomes from R47Hhet iPS-Mg contained disease-associated microglial (DAM) signature proteins and were less able to promote the outgrowth of neuronal processes and increase mitochondrial metabolism in neurons compared with exosomes from the common TREM2 variant iPS-Mg. Taken together, these data highlight the importance of microglial exosomes in fulfilling microglial functions. Additionally, variations in the exosomal proteome influenced by the R47Hhet TREM2 variant may underlie the increased risk of Alzheimer’s disease associated with this variant.
Collapse
Affiliation(s)
- Anna Mallach
- Department of Neuroinflammation, UCL Queen Square Institute of Neurology, University College, London WC1N 1PJ, UK; (A.M.); (T.M.P.)
| | - Johan Gobom
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, S-43180 Molndal, Sweden; (J.G.); (H.Z.)
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, S-431 80 Molndal, Sweden
| | - Charles Arber
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London WC1N 1PJ, UK; (C.A.); (J.H.); (S.W.)
| | - Thomas M. Piers
- Department of Neuroinflammation, UCL Queen Square Institute of Neurology, University College, London WC1N 1PJ, UK; (A.M.); (T.M.P.)
| | - John Hardy
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London WC1N 1PJ, UK; (C.A.); (J.H.); (S.W.)
- UK Dementia Research Institute at UCL, London WC1E 6BT, UK
| | - Selina Wray
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London WC1N 1PJ, UK; (C.A.); (J.H.); (S.W.)
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, S-43180 Molndal, Sweden; (J.G.); (H.Z.)
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, S-431 80 Molndal, Sweden
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London WC1N 1PJ, UK; (C.A.); (J.H.); (S.W.)
- UK Dementia Research Institute at UCL, London WC1E 6BT, UK
| | - Jennifer Pocock
- Department of Neuroinflammation, UCL Queen Square Institute of Neurology, University College, London WC1N 1PJ, UK; (A.M.); (T.M.P.)
- Correspondence:
| |
Collapse
|
117
|
Gagné M, Deshaies JE, Sidibé H, Benchaar Y, Arbour D, Dubinski A, Litt G, Peyrard S, Robitaille R, Sephton CF, Vande Velde C. hnRNP A1B, a Splice Variant of HNRNPA1, Is Spatially and Temporally Regulated. Front Neurosci 2021; 15:724307. [PMID: 34630013 PMCID: PMC8498194 DOI: 10.3389/fnins.2021.724307] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 08/30/2021] [Indexed: 11/28/2022] Open
Abstract
RNA binding proteins (RBPs) play a key role in cellular growth, homoeostasis and survival and are tightly regulated. A deep understanding of their spatiotemporal regulation is needed to understand their contribution to physiology and pathology. Here, we have characterized the spatiotemporal expression pattern of hnRNP A1 and its splice variant hnRNP A1B in mice. We have found that hnRNP A1B expression is more restricted to the CNS compared to hnRNP A1, and that it can form an SDS-resistant dimer in the CNS. Also, hnRNP A1B expression becomes progressively restricted to motor neurons in the ventral horn of the spinal cord, compared to hnRNP A1 which is more broadly expressed. We also demonstrate that hnRNP A1B is present in neuronal processes, while hnRNP A1 is absent. This finding supports a hypothesis that hnRNP A1B may have a cytosolic function in neurons that is not shared with hnRNP A1. Our results demonstrate that both isoforms are differentially expressed across tissues and have distinct localization profiles, suggesting that the two isoforms may have specific subcellular functions that can uniquely contribute to disease progression.
Collapse
Affiliation(s)
- Myriam Gagné
- Department of Biochemistry, Université de Montréal, Montréal, QC, Canada.,Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Jade-Emmanuelle Deshaies
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Hadjara Sidibé
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada.,Department of Neurosciences, Université de Montréal, Montréal, QC, Canada
| | - Yousri Benchaar
- Department of Psychiatry and Neuroscience, CERVO Brain Research Centre, Laval University, Quebec City, QC, Canada
| | - Danielle Arbour
- Department of Neurosciences, Université de Montréal, Montréal, QC, Canada
| | - Alicia Dubinski
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada.,Department of Neurosciences, Université de Montréal, Montréal, QC, Canada
| | - Gurleen Litt
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Sarah Peyrard
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Richard Robitaille
- Department of Neurosciences, Université de Montréal, Montréal, QC, Canada
| | - Chantelle F Sephton
- Department of Psychiatry and Neuroscience, CERVO Brain Research Centre, Laval University, Quebec City, QC, Canada
| | - Christine Vande Velde
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada.,Department of Neurosciences, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
118
|
Brum PO, Viola GD, Saibro-Girardi C, Tiefensee-Ribeiro C, Brum MO, Gasparotto J, Krolow R, Moreira JCF, Gelain DP. Hypoxia-Inducible Factor-1α (HIF-1α) Inhibition Impairs Retinoic Acid-Induced Differentiation in SH-SY5Y Neuroblastoma Cells, Leading to Reduced Neurite Length and Diminished Gene Expression Related to Cell Differentiation. Neurochem Res 2021; 47:409-421. [PMID: 34557995 PMCID: PMC8827409 DOI: 10.1007/s11064-021-03454-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 09/13/2021] [Accepted: 09/14/2021] [Indexed: 11/30/2022]
Abstract
Neuroblastoma is the most common extracranial solid tumour in childhood, originated from cells of the neural crest during the development of the Sympathetic Nervous System. Retinoids are vitamin-A derived differentiating agents utilised to avoid disease resurgence in high-risk neuroblastoma treatment. Several studies indicate that hypoxia—a common feature of the tumoural environment—is a key player in cell differentiation and proliferation. Hypoxia leads to the accumulation of the hypoxia-inducible factor-1α (HIF-1α). This work aims to investigate the effects of the selective inhibition of HIF-1α on the differentiation induced by retinoic acid in human neuroblastoma cells from the SH-SY5Y lineage to clarify its role in cell differentiation. Our results indicate that HIF-1α inhibition impairs RA-induced differentiation by reducing neuron-like phenotype and diminished immunolabeling and expression of differentiation markers.
Collapse
Affiliation(s)
- Pedro Ozorio Brum
- Departamento de Bioquímica, Centro de Estudos em Estresse Oxidativo, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil. .,Max F. Perutz Labs, University of Vienna, Dr Bohr-Gasse 9, Room 4.510, 1030, Vienna, Austria.
| | - Guilherme Danielski Viola
- Laboratório de Medicina Genômica, Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Carolina Saibro-Girardi
- Departamento de Bioquímica, Centro de Estudos em Estresse Oxidativo, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Camila Tiefensee-Ribeiro
- Departamento de Bioquímica, Centro de Estudos em Estresse Oxidativo, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | | | - Juciano Gasparotto
- Instituto de Ciências Biomédicas, Universidade Federal de Alfenas, Alfenas, MG, Brazil
| | - Rachel Krolow
- Laboratório de Programação Neurobiológica do Comportamento Alimentar, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - José Cláudio Fonseca Moreira
- Departamento de Bioquímica, Centro de Estudos em Estresse Oxidativo, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Daniel Pens Gelain
- Departamento de Bioquímica, Centro de Estudos em Estresse Oxidativo, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
119
|
Russo M, Sobh A, Zhang P, Loguinov A, Tagmount A, Vulpe CD, Liu B. Functional Pathway Identification With CRISPR/Cas9 Genome-wide Gene Disruption in Human Dopaminergic Neuronal Cells Following Chronic Treatment With Dieldrin. Toxicol Sci 2021; 176:366-381. [PMID: 32421776 DOI: 10.1093/toxsci/kfaa071] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Organochlorine pesticides, once widely used, are extremely persistent and bio-accumulative in the environment. Epidemiological studies have implicated that environmental exposure to organochlorine pesticides including dieldrin is a risk factor for the development of Parkinson's disease. However, the pertinent mechanisms of action remain poorly understood. In this study, we carried out a genome-wide (Brunello library, 19 114 genes, 76 411 sgRNAs) CRISPR/Cas9 screen in human dopaminergic SH-SY5Y neuronal cells exposed to a chronic treatment (30 days) with dieldrin to identify cellular pathways that are functionally related to the chronic cellular toxicity. Our results indicate that dieldrin toxicity was enhanced by gene disruption of specific components of the ubiquitin proteasome system as well as, surprisingly, the protein degradation pathways previously implicated in inherited forms of Parkinson's disease, centered on Parkin. In addition, disruption of regulatory components of the mTOR pathway which integrates cellular responses to both intra- and extracellular signals and is a central regulator for cell metabolism, growth, proliferation, and survival, led to increased sensitivity to dieldrin-induced cellular toxicity. This study is one of the first to apply a genome-wide CRISPR/Cas9-based functional gene disruption screening approach in an adherent neuronal cell line to globally decipher cellular mechanisms that contribute to environmental toxicant-induced neurotoxicity and provides novel insight into the dopaminergic neurotoxicity associated with chronic exposure to dieldrin.
Collapse
Affiliation(s)
- Max Russo
- Department of Pharmacodynamics, College of Pharmacy
| | - Amin Sobh
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida 32610
| | - Ping Zhang
- Department of Pharmacodynamics, College of Pharmacy
| | - Alex Loguinov
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida 32610
| | - Abderrahmane Tagmount
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida 32610
| | - Chris D Vulpe
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida 32610
| | - Bin Liu
- Department of Pharmacodynamics, College of Pharmacy
| |
Collapse
|
120
|
Dusan M, Jastrow C, Alyce MM, Yingkai W, Shashikanth M, Andelain E, Christine BM, Stuart BM, Oliver BG, Michael MZ, Nicolas VH, Damien KJ, Rainer HV. Differentiation of the 50B11 dorsal ganglion cells into NGF and GDNF responsive nociceptor subtypes. Mol Pain 2021; 16:1744806920970368. [PMID: 33307981 PMCID: PMC7745567 DOI: 10.1177/1744806920970368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The embryonic rat dorsal root ganglion (DRG) neuron-derived 50B11 cell line is a promising sensory neuron model expressing markers characteristic of NGF and GDNF-dependent C-fibre nociceptors. Whether these cells have the capacity to develop into distinct nociceptive subtypes based on NGF- or GDNF-dependence has not been investigated. Here we show that by augmenting forskolin (FSK) and growth factor supplementation with NGF or GDNF, 50B11 cultures can be driven to acquire differential functional responses to common nociceptive agonists capsaicin and ATP respectively. In addition, to previous studies, we also demonstrate that a differentiated neuronal phenotype can be maintained for up to 7 days. Western blot analysis of nociceptive marker proteins further demonstrates that the 50B11 cells partially recapitulate the functional phenotypes of classical NGF-dependent (peptidergic) and GDNF-dependent (non-peptidergic) neuronal subtypes described in DRGs. Further, 50B11 cells differentiated with NGF/FSK, but not GDNF/FSK, show sensitization to acute prostaglandin E2 treatment. Finally, RNA-Seq analysis confirms that differentiation with NGF/FSK or GDNF/FSK produces two 50B11 cell subtypes with distinct transcriptome expression profiles. Gene ontology comparison of the two subtypes of differentiated 50B11 cells to rodent DRG neurons studies shows significant overlap in matching or partially matching categories. This transcriptomic analysis will aid future suitability assessment of the 50B11 cells as a high-throughput nociceptor model for a broad range of experimental applications. In conclusion, this study shows that the 50B11 cell line is capable of partially recapitulating features of two distinct types of embryonic NGF and GDNF-dependent nociceptor-like cells.
Collapse
Affiliation(s)
- Matusica Dusan
- Anatomy and Histology, Flinders Health & Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Canlas Jastrow
- Anatomy and Histology, Flinders Health & Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Martin M Alyce
- Human Physiology, Flinders Health & Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Wei Yingkai
- Flinders Centre for Innovation in Cancer, College of Medicine and Public Health, Flinders University and Medical Centre, Bedford Park, Adelaide, South Australia, Australia
| | - Marri Shashikanth
- Visceral Pain Research Group, College of Medicine and Public Health, South Australian Health and Medical Research Institute, Flinders University, Adelaide, South Australia, Australia
| | - Erickson Andelain
- Flow Cytometry Facility, Department of Molecular Medicine and Genetics, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Barry M Christine
- Anatomy and Histology, Flinders Health & Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Brierley M Stuart
- Flow Cytometry Facility, Department of Molecular Medicine and Genetics, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Best G Oliver
- Flow Cytometry Facility, Department of Molecular Medicine and Genetics, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Michael Z Michael
- Visceral Pain Research Group, College of Medicine and Public Health, South Australian Health and Medical Research Institute, Flinders University, Adelaide, South Australia, Australia
| | - Voelcker H Nicolas
- Flinders Centre for Innovation in Cancer, College of Medicine and Public Health, Flinders University and Medical Centre, Bedford Park, Adelaide, South Australia, Australia
| | - Keating J Damien
- Human Physiology, Flinders Health & Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Haberberger V Rainer
- Anatomy and Histology, Flinders Health & Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| |
Collapse
|
121
|
Strother L, Miles GB, Holiday AR, Cheng Y, Doherty GH. Long-term culture of SH-SY5Y neuroblastoma cells in the absence of neurotrophins: A novel model of neuronal ageing. J Neurosci Methods 2021; 362:109301. [PMID: 34343572 PMCID: PMC8434422 DOI: 10.1016/j.jneumeth.2021.109301] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 07/07/2021] [Accepted: 07/29/2021] [Indexed: 01/06/2023]
Abstract
Background Studying human ageing is of increasing importance due to the worldwide ageing population. However, it faces the challenge of lengthy experiments to produce an ageing phenotype. Often, to recreate the hallmarks of ageing requires complex empirical conditions that can confound data interpretation. Indeed, many studies use whole organisms with relatively short life spans, which may have little, or limited, relevance to human ageing. There has been extensive use of cell lines to study ageing in human somatic cells, but the modelling of human neuronal ageing is somewhat more complex in vitro. New Method We cultured the well-characterised SH-SY5Y human neural cell line to produce high purity cultures of cells differentiated to express a neuronal phenotype, and designed a protocol to maintain these cells in culture until they accumulated biomarkers of cellular ageing. Results Our data validate a novel and simple technique for the efficient differentiation and long-term maintenance of SH-SY5Y cells, expressing markers of neuronal differentiation and demonstrating electrical activity in culture. Over time in vitro, these cells progressively accumulate markers of ageing such as enhanced production of reactive oxygen species and accumulation of oxidative damage. Comparison to Existing Methods In comparison to existing techniques to model neuronal ageing our method is cost effective, requiring no specialist equipment or growth factors. Conclusions We demonstrate that SH-SY5Y cells, grown under these culture conditions, represent a simple model of neuronal ageing that is amenable to cell biological, biochemical and electrophysiological investigation. Ageing study is often hindered by the need for complex and lengthy experiments. SH-SY5Y cells underwent neuronal differentiation and were cultured until they were of an aged phenotype. These cells were electrically active and acquired oxidative damage. This is a novel technique to model neuronal ageing in vitro.
Collapse
Affiliation(s)
- Lisa Strother
- Bute Building, School of Psychology and Neuroscience, University of St Andrews, West Burn Lane, St Andrews, Fife KY16 9TS, UK
| | - Gareth B Miles
- Bute Building, School of Psychology and Neuroscience, University of St Andrews, West Burn Lane, St Andrews, Fife KY16 9TS, UK
| | - Alison R Holiday
- Bute Building, School of Psychology and Neuroscience, University of St Andrews, West Burn Lane, St Andrews, Fife KY16 9TS, UK
| | - Ying Cheng
- Bute Building, School of Psychology and Neuroscience, University of St Andrews, West Burn Lane, St Andrews, Fife KY16 9TS, UK
| | - Gayle H Doherty
- Bute Building, School of Psychology and Neuroscience, University of St Andrews, West Burn Lane, St Andrews, Fife KY16 9TS, UK.
| |
Collapse
|
122
|
Costa CP, Cunha S, Moreira JN, Silva R, Gil-Martins E, Silva V, Azevedo L, Peixoto AF, Sousa Lobo JM, Silva AC. Quality by design (QbD) optimization of diazepam-loaded nanostructured lipid carriers (NLC) for nose-to-brain delivery: Toxicological effect of surface charge on human neuronal cells. Int J Pharm 2021; 607:120933. [PMID: 34324988 DOI: 10.1016/j.ijpharm.2021.120933] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/10/2021] [Accepted: 07/21/2021] [Indexed: 12/24/2022]
Abstract
Diazepam is commonly used in the management of epileptic seizures, although it has limitations that can be overcome by using formulations that are easier to administer and capable of directing the drug to the brain. In this field, it has been reported that the use of nanostructured lipid carriers (NLC) via intranasal (or via nose-to-brain) promotes the targeting of drugs to the brain, improving the effectiveness of therapy. The aim of this work was to optimize two diazepam-loaded NLC formulations for nose-to-brain delivery, one with positive surface charge and one with negative surface charge. The quality by design (QbD) approach was used to design the experiments, where the quality target product profile (QTPP), the risk assessment and the critical quality attributes (CQAs) were defined to ensure safety, efficacy and quality of the final formulations. The experiments started with the optimization of critical material attributes (CMAs), related to the ratios of lipids and emulsifiers, followed by the selection of critical process parameters (CPPs), related to the production methods of the diazepam-loaded NLC formulation (ultrasound technique and high-pressure homogenization - HPH). Afterwards, the positive surface charge of the diazepam-loaded NLC was optimized. Finally, the biocompatibility with human neuronal cells of the formulation with a negative surface charge and of the formulation with a positive surface charge was evaluated. The results of the optimization of the CMAs showed that the ratios of lipids and emulsifiers more adequate were 6.7:2.9 and 4.2:0.3 (% w,w), respectively. Regarding the CPPs, HPH was considered the most suitable production method, resulting in an optimized diazepam-loaded NLC formulation (F1C15) with negative surface charge, showing particle size of 69.59 ± 0.22 nm, polydispersity index (PDI) of 0.19 ± 0.00, zeta potential (ZP) of -23.50 ± 0.24 mV and encapsulation efficiency (EE) of 96.60 ± 0.03 %. The optimized diazepam-loaded NLC formulation (F2A8) with positive surface charge had particle size of 124.40 ± 0.84 nm, PDI of 0.17 ± 0.01, ZP of 32.60 ± 1.13 mV and EE of 95.76 ± 0.24 %. In addition, the incorporation of diazepam in NLC resulted in a sustained release of the drug. No significant changes in particle size, PDI, ZP and EE were observed for the formulation F1C15, after 3 months of storage, whereas for formulation F2A8, particle size increased significantly. Biocompatibility studies showed that the formulation F2A8 was more cytotoxic than the formulation F1C15. Thereby, we conclude that the formulation F1C15 is more suitable for targeting the brain, when compared with the formulation F2A8. From the results of these studies, it can be confirmed that the QbD approach is an adequate and central tool to optimize NLC formulations.
Collapse
Affiliation(s)
- C P Costa
- UCIBIO/REQUIMTE, MEDTECH, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - S Cunha
- UCIBIO/REQUIMTE, MEDTECH, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - J N Moreira
- CNC - Center for Neuroscience and Cell Biology, Center for Innovative Biomedicine and Biotechnology (CIBB), Faculty of Medicine (Pólo I), University of Coimbra, 3004-531 Coimbra, Portugal; UC - University of Coimbra, CIBB, Faculty of Pharmacy, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - R Silva
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, Porto University, Porto, Portugal
| | - E Gil-Martins
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, Porto University, Porto, Portugal
| | - V Silva
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, Porto University, Porto, Portugal
| | - L Azevedo
- UCIBIO/REQUIMTE, MEDTECH, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - A F Peixoto
- LAQV/REQUIMTE, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal
| | - J M Sousa Lobo
- UCIBIO/REQUIMTE, MEDTECH, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - A C Silva
- UCIBIO/REQUIMTE, MEDTECH, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; FP-ENAS (UFP Energy, Environment and Health Research Unit), CEBIMED (Biomedical Research Centre), Faculty of Health Sciences, University Fernando Pessoa, 4249-004 Porto, Portugal.
| |
Collapse
|
123
|
Ban M, Shimoda R, Chen J. Investigation of nanoplastic cytotoxicity using SH-SY5Y human neuroblastoma cells and polystyrene nanoparticles. Toxicol In Vitro 2021; 76:105225. [PMID: 34293433 DOI: 10.1016/j.tiv.2021.105225] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 07/09/2021] [Accepted: 07/15/2021] [Indexed: 11/15/2022]
Abstract
Nanoplastics have spread widely throughout not only the oceans but also the atmosphere, and recently created great concern about human health relevant to ingestion and accumulation of the nanoparticles by aquatic organisms in the human food-chain. However, how the nanoplastics have an affect on actual human body remains largely unknown, and in particular, little knowledge about nanoplastic exposure to the nervous system in human has been obtained in vitro and still less vivo. Here, we evaluated how much concentration of nanoplastics had a direct impact on cells in the nervous system as the fundamental information. Specifically, the cytotoxicity was investigated by exposure of polystyrene nanoparticles (PS) to cultured neural cells, human neuroblastoma cells, SH-SY5Y. Our results demonstrated that the PS exposure induced the cytotoxicity in the cells promoted differentiation into neuronal phenotype, and the adverse effect was comparable to or exceed that of acrylamide, a well-recognized potent neurotoxin. Also, the cells under PS exposure exhibited shrinkage of neurite outgrowth, morphology alteration and swelling of the nuclei, and spilling of intracellular components. Moreover, our findings indicate that the concentration of nanoplastics caused the cytotoxicity on neuronal cells is likely to be much higher than those predicted from the marine environment.
Collapse
Affiliation(s)
- Masahito Ban
- Dept. of Applied Chemistry, Nippon Institute of Technology, 4-1, Gakuendai, Miyashiro, Minami-saitama, Saitama 345-8501, Japan; Environmental Symbiotic System Major, Nippon Institute of Technology, 4-1, Gakuendai, Miyashiro, Minami-saitama, Saitama 345-8501, Japan.
| | - Ryouta Shimoda
- Environmental Symbiotic System Major, Nippon Institute of Technology, 4-1, Gakuendai, Miyashiro, Minami-saitama, Saitama 345-8501, Japan
| | - Jing Chen
- Environmental Symbiotic System Major, Nippon Institute of Technology, 4-1, Gakuendai, Miyashiro, Minami-saitama, Saitama 345-8501, Japan
| |
Collapse
|
124
|
Oikawa N, Fabiano M, Müller UC, Walter J. Carboxy-terminal fragment of amyloid precursor protein mediates lipid droplet accumulation upon γ-secretase inhibition. Biochem Biophys Res Commun 2021; 570:137-142. [PMID: 34280617 DOI: 10.1016/j.bbrc.2021.07.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 07/07/2021] [Indexed: 11/20/2022]
Abstract
γ-Secretase is a protease catalysing the proteolysis of type-I membrane proteins usually after precedent ectodomain shedding of the respective protein substrates. Since proteolysis of membrane proteins is involved in fundamental cellular signaling pathways, dysfunction of γ-secretase can have significant impact on cellular metabolism and differentiation. Here, we examined the role of γ-secretase in cellular lipid metabolism using neuronally differentiated human SH-SY5Y cells. The pharmacological inhibition of γ-secretase induced lipid droplet (LD) accumulation. The LD accumulation was significantly attenuated by preventing the accumulation of C-terminal fragment of the amyloid precursor protein (APP-CTF), which is a direct substrate of γ-secretase. Additionally, LD accumulation upon γ-secretase inhibition was not induced in APP-knock out (APP-KO) mouse embryonic fibroblasts (MEFs), suggesting significant involvement of APP-CTF accumulation in LD accumulation upon γ-secretase inhibition. On the other hand, γ-secretase inhibition-dependent cholesterol accumulation was not attenuated by inhibition of APP-CTF accumulation in the differentiated SH-SY5Y cells nor in APP-KO MEFs. These results suggest that γ-secretase inhibition can induce accumulation of LD and cholesterol differentially via APP-CTF accumulation.
Collapse
Affiliation(s)
- Naoto Oikawa
- Department of Neurology, University Hospital Bonn, 53127, Bonn, Germany.
| | - Marietta Fabiano
- Department of Neurology, University Hospital Bonn, 53127, Bonn, Germany
| | - Ulrike C Müller
- Institute for Pharmacy and Molecular Biotechnology, University of Heidelberg, 69120, Heidelberg, Germany
| | - Jochen Walter
- Department of Neurology, University Hospital Bonn, 53127, Bonn, Germany.
| |
Collapse
|
125
|
Sánchez-Alegría K, Bastián-Eugenio CE, Vaca L, Arias C. Palmitic acid induces insulin resistance by a mechanism associated with energy metabolism and calcium entry in neuronal cells. FASEB J 2021; 35:e21712. [PMID: 34110637 DOI: 10.1096/fj.202100243r] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/27/2021] [Accepted: 05/17/2021] [Indexed: 01/05/2023]
Abstract
Palmitic acid (PA) is a saturated fatty acid whose high consumption has been largely associated with the development of different metabolic alterations, such as insulin resistance, metabolic syndrome, and type 2 diabetes. Particularly in the brain, insulin signaling disruption has been linked to cognitive decline and is considered a risk factor for Alzheimer's disease. Cumulative evidence has demonstrated the participation of PA in the molecular cascade underlying cellular insulin resistance in peripheral tissues, but its role in the development of neuronal insulin resistance and the mechanisms involved are not fully understood. It has generally been accepted that the brain does not utilize fatty acids as a primary energy source, but recent evidence shows that neurons possess the machinery for fatty acid β-oxidation. However, it is still unclear under what conditions neurons use fatty acids as energy substrates and the implications of their oxidative metabolism in modifying insulin-stimulated effects. In the present work, we have found that neurons differentiated from human neuroblastoma MSN exposed to high but nontoxic concentrations of PA generate ATP through mitochondrial metabolism, which is associated with an increase in the cytosolic Ca2+ and diminished insulin signaling in neurons. These findings reveal a novel mechanism by which saturated fatty acids produce Ca2+ entry and insulin resistance that may play a causal role in increasing neuronal vulnerability associated with metabolic diseases.
Collapse
Affiliation(s)
- Karina Sánchez-Alegría
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Carlos Ernesto Bastián-Eugenio
- Departamento de Genética Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Luis Vaca
- Departamento de Genética Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Clorinda Arias
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| |
Collapse
|
126
|
Ma C, Zhang W, Wang W, Shen J, Cai K, Liu M, Cao M. SKP-SCs transplantation alleviates 6-OHDA-induced dopaminergic neuronal injury by modulating autophagy. Cell Death Dis 2021; 12:674. [PMID: 34226513 PMCID: PMC8257782 DOI: 10.1038/s41419-021-03967-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 06/15/2021] [Accepted: 06/18/2021] [Indexed: 02/06/2023]
Abstract
Parkinson's disease is a common neurodegenerative disease. Cell transplantation is a promising therapeutic option for improving the survival and function of dopaminergic neurons, but the mechanisms underlying the interaction between the transplanted cells and the recipient neurons remain to be studied. In this study, we investigated the effects of skin precursor cell-derived Schwann cells (SKP-SCs) directly cocultured with 6-OHDA-injured dopaminergic neurons in vitro and of SKP-SCs transplanted into the brains of 6-OHDA-induced PD mice in vivo. In vitro and in vivo studies revealed that SKP-SCs could reduce the damage to dopaminergic neurons by enhancing self-autophagy and modulating neuronal autophagy. Thus, the present study provides the first evidence that cell transplantation mitigates 6-OHDA-induced damage to dopaminergic neurons by enhancing self-autophagy, suggesting that earlier transplantation of Schwann cells might help alleviate the loss of dopaminergic neurons.
Collapse
Affiliation(s)
- Chengxiao Ma
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, China
| | - Wen Zhang
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, China
| | - Wengcong Wang
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, China
| | - Jiabing Shen
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, China
| | - Kefu Cai
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, China
| | - Mei Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.
| | - Maohong Cao
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, China.
| |
Collapse
|
127
|
Hudák A, Jósvay K, Domonkos I, Letoha A, Szilák L, Letoha T. The Interplay of Apoes with Syndecans in Influencing Key Cellular Events of Amyloid Pathology. Int J Mol Sci 2021; 22:ijms22137070. [PMID: 34209175 PMCID: PMC8268055 DOI: 10.3390/ijms22137070] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/20/2021] [Accepted: 06/28/2021] [Indexed: 01/06/2023] Open
Abstract
Apolipoprotein E (ApoE) isoforms exert intricate effects on cellular physiology beyond lipid transport and metabolism. ApoEs influence the onset of Alzheimer’s disease (AD) in an isoform-dependent manner: ApoE4 increases AD risk, while ApoE2 decreases it. Previously we demonstrated that syndecans, a transmembrane proteoglycan family with increased expression in AD, trigger the aggregation and modulate the cellular uptake of amyloid beta (Aβ). Utilizing our previously established syndecan-overexpressing cellular assays, we now explore how the interplay of ApoEs with syndecans contributes to key events, namely uptake and aggregation, in Aβ pathology. The interaction of ApoEs with syndecans indicates isoform-specific characteristics arising beyond the frequently studied ApoE–heparan sulfate interactions. Syndecans, and among them the neuronal syndecan-3, increased the cellular uptake of ApoEs, especially ApoE2 and ApoE3, while ApoEs exerted opposing effects on syndecan-3-mediated Aβ uptake and aggregation. ApoE2 increased the cellular internalization of monomeric Aβ, hence preventing its extracellular aggregation, while ApoE4 decreased it, thus helping the buildup of extracellular plaques. The contrary effects of ApoE2 and ApoE4 remained once Aβ aggregated: while ApoE2 reduced the uptake of Aβ aggregates, ApoE4 facilitated it. Fibrillation studies also revealed ApoE4′s tendency to form fibrillar aggregates. Our results uncover yet unknown details of ApoE cellular biology and deepen our molecular understanding of the ApoE-dependent mechanism of Aβ pathology.
Collapse
Affiliation(s)
- Anett Hudák
- Pharmacoidea Ltd., H-6726 Szeged, Hungary; (A.H.); (L.S.)
| | - Katalin Jósvay
- Institute of Biochemistry, Biological Research Centre, H-6726 Szeged, Hungary;
| | - Ildikó Domonkos
- Institute of Plant Biology, Biological Research Centre, H-6726 Szeged, Hungary;
| | - Annamária Letoha
- Department of Medicine, Albert Szent-Györgyi Clinical Center, Faculty of Medicine, University of Szeged, H-6725 Szeged, Hungary;
| | - László Szilák
- Pharmacoidea Ltd., H-6726 Szeged, Hungary; (A.H.); (L.S.)
| | - Tamás Letoha
- Pharmacoidea Ltd., H-6726 Szeged, Hungary; (A.H.); (L.S.)
- Correspondence: ; Tel.: +36-(30)-2577393
| |
Collapse
|
128
|
Chin MY, Patwardhan AR, Ang KH, Wang AL, Alquezar C, Welch M, Nguyen PT, Grabe M, Molofsky AV, Arkin MR, Kao AW. Genetically Encoded, pH-Sensitive mTFP1 Biosensor for Probing Lysosomal pH. ACS Sens 2021; 6:2168-2180. [PMID: 34102054 PMCID: PMC8240087 DOI: 10.1021/acssensors.0c02318] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 04/08/2021] [Indexed: 12/15/2022]
Abstract
Lysosomes are important sites for macromolecular degradation, defined by an acidic lumenal pH of ∼4.5. To better understand lysosomal pH, we designed a novel, genetically encoded, fluorescent protein (FP)-based pH biosensor called Fluorescence Indicator REporting pH in Lysosomes (FIRE-pHLy). This biosensor was targeted to lysosomes with lysosomal-associated membrane protein 1 (LAMP1) and reported lumenal pH between 3.5 and 6.0 with monomeric teal fluorescent protein 1 (mTFP1), a bright cyan pH-sensitive FP variant with a pKa of 4.3. Ratiometric quantification was enabled with cytosolically oriented mCherry using high-content quantitative imaging. We expressed FIRE-pHLy in several cellular models and quantified the alkalinizing response to bafilomycin A1, a specific V-ATPase inhibitor. In summary, we have engineered FIRE-pHLy, a specific, robust, and versatile lysosomal pH biosensor, that has broad applications for investigating pH dynamics in aging- and lysosome-related diseases, as well as in lysosome-based drug discovery.
Collapse
Affiliation(s)
- Marcus Y Chin
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, California 94158, United States
- Small Molecule Discovery Center, Department of Pharmaceutical Chemistry, University of California, San Francisco, California 94143, United States
| | - Anand R Patwardhan
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, California 94158, United States
| | - Kean-Hooi Ang
- Small Molecule Discovery Center, Department of Pharmaceutical Chemistry, University of California, San Francisco, California 94143, United States
| | - Austin L Wang
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, California 94158, United States
| | - Carolina Alquezar
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, California 94158, United States
| | - Mackenzie Welch
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, California 94158, United States
| | - Phi T Nguyen
- Weill Institute for Neurosciences, Department of Psychiatry, University of California, San Francisco, California 94158, United States
| | - Michael Grabe
- Cardiovascular Research Institute, Department of Pharmaceutical Chemistry, University of California, San Francisco, California 94158, United States
| | - Anna V Molofsky
- Weill Institute for Neurosciences, Department of Psychiatry, University of California, San Francisco, California 94158, United States
| | - Michelle R Arkin
- Small Molecule Discovery Center, Department of Pharmaceutical Chemistry, University of California, San Francisco, California 94143, United States
| | - Aimee W Kao
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, California 94158, United States
| |
Collapse
|
129
|
Chung YS, Ahmed PK, Othman I, Shaikh MF. Orthosiphon stamineus Proteins Alleviate Hydrogen Peroxide Stress in SH-SY5Y Cells. Life (Basel) 2021; 11:life11060585. [PMID: 34202937 PMCID: PMC8235403 DOI: 10.3390/life11060585] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/16/2021] [Accepted: 06/17/2021] [Indexed: 12/12/2022] Open
Abstract
The neuroprotective potential of Orthosiphon stamineus leaf proteins (OSLPs) has never been evaluated in SH-SY5Y cells challenged by hydrogen peroxide (H2O2). This work thus aims to elucidate OSLP neuroprotective potential in alleviating H2O2 stress. OSLPs at varying concentrations were evaluated for cytotoxicity (24 and 48 h) and neuroprotective potential in H2O2-induced SH-SY5Y cells (24 h). The protective mechanism of H2O2-induced SH-SY5Y cells was also explored via mass-spectrometry-based label-free quantitative proteomics (LFQ) and bioinformatics. OSLPs (25, 50, 125, 250, 500, and 1000 µg/mL; 24 and 48 h) were found to be safe. Pre-treatments with OSLP doses (250, 500, and 1000 µg/mL, 24 h) significantly increased the survival of SH-SY5Y cells in a concentration-dependent manner and improved cell architecture—pyramidal-shaped cells, reduced clumping and shrinkage, with apparent neurite formations. OSLP pre-treatment (1000 µg/mL, 24 h) lowered the expressions of two major heat shock proteins, HSPA8 (heat shock protein family A (Hsp70) member 8) and HSP90AA1 (heat shock protein 90), which promote cellular stress signaling under stress conditions. OSLP is, therefore, suggested to be anti-inflammatory by modulating the “signaling of interleukin-4 and interleukin-13” pathway as the predominant mechanism in addition to regulating the “attenuation phase” and “HSP90 chaperone cycle for steroid hormone receptors” pathways to counteract heat shock protein (HSP)-induced damage under stress conditions.
Collapse
Affiliation(s)
- Yin-Sir Chung
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Malaysia; (Y.-S.C.); (I.O.)
| | - Pervaiz Khalid Ahmed
- School of Business, Monash University Malaysia, Bandar Sunway 47500, Malaysia;
- Global Asia in the 21st Century (GA21), Monash University Malaysia, Bandar Sunway 47500, Malaysia
| | - Iekhsan Othman
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Malaysia; (Y.-S.C.); (I.O.)
- Liquid Chromatography-Mass Spectrometry (LCMS) Platform, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Malaysia
| | - Mohd. Farooq Shaikh
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Malaysia; (Y.-S.C.); (I.O.)
- Global Asia in the 21st Century (GA21), Monash University Malaysia, Bandar Sunway 47500, Malaysia
- Correspondence:
| |
Collapse
|
130
|
Selected Kefir Water from Malaysia Attenuates Hydrogen Peroxide-Induced Oxidative Stress by Upregulating Endogenous Antioxidant Levels in SH-SY5Y Neuroblastoma Cells. Antioxidants (Basel) 2021; 10:antiox10060940. [PMID: 34200854 PMCID: PMC8230435 DOI: 10.3390/antiox10060940] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/03/2021] [Accepted: 06/08/2021] [Indexed: 01/27/2023] Open
Abstract
Kefir, a fermented probiotic drink was tested for its potential anti-oxidative, anti-apoptotic, and neuroprotective effects to attenuate cellular oxidative stress on human SH-SY5Y neuroblastoma cells. Here, the antioxidant potentials of the six different kefir water samples were analysed by total phenolic content (TPC), total flavonoid content (TFC), ferric reducing antioxidant power (FRAP), and 2,2'-diphenyl-1-picrylhydrazyl radical (DPPH) assays, whereas the anti-apoptotic activity on hydrogen peroxide (H2O2) induced SH-SY5Y cells was examined using MTT, AO/PI double staining, and PI/Annexin V-FITC assays. The surface and internal morphological features of SH-SY5Y cells were studied using scanning and transmission electron microscopy. The results indicate that Kefir B showed the higher TPC (1.96 ± 0.54 µg GAE/µL), TFC (1.09 ± 0.02 µg CAT eq/µL), FRAP (19.68 ± 0.11 mM FRAP eq/50 µL), and DPPH (0.45 ± 0.06 mg/mL) activities compared to the other kefir samples. The MTT and PI/Annexin V-FITC assays showed that Kefir B pre-treatment at 10 mg/mL for 48 h resulted in greater cytoprotection (97.04%), and a significantly lower percentage of necrotic cells (7.79%), respectively. The Kefir B pre-treatment also resulted in greater protection to cytoplasmic and cytoskeleton inclusion, along with the conservation of the surface morphological features and the overall integrity of SH-SY5Y cells. Our findings indicate that the anti-oxidative, anti-apoptosis, and neuroprotective effects of kefir were mediated via the upregulation of SOD and catalase, as well as the modulation of apoptotic genes (Tp73, Bax, and Bcl-2).
Collapse
|
131
|
Vu L, Ghosh A, Tran C, Tebung WA, Sidibé H, Garcia-Mansfield K, David-Dirgo V, Sharma R, Pirrotte P, Bowser R, Vande Velde C. Defining the Caprin-1 Interactome in Unstressed and Stressed Conditions. J Proteome Res 2021; 20:3165-3178. [PMID: 33939924 PMCID: PMC9083243 DOI: 10.1021/acs.jproteome.1c00016] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cytoplasmic stress granules (SGs) are dynamic foci containing translationally arrested mRNA and RNA-binding proteins (RBPs) that form in response to a variety of cellular stressors. It has been debated that SGs may evolve into cytoplasmic inclusions observed in many neurodegenerative diseases. Recent studies have examined the SG proteome by interrogating the interactome of G3BP1. However, it is widely accepted that multiple baits are required to capture the full SG proteome. To gain further insight into the SG proteome, we employed immunoprecipitation coupled with mass spectrometry of endogenous Caprin-1, an RBP implicated in mRNP granules. Overall, we identified 1543 proteins that interact with Caprin-1. Interactors under stressed conditions were primarily annotated to the ribosome, spliceosome, and RNA transport pathways. We validated four Caprin-1 interactors that localized to arsenite-induced SGs: ANKHD1, TALIN-1, GEMIN5, and SNRNP200. We also validated these stress-induced interactions in SH-SY5Y cells and further determined that SNRNP200 also associated with osmotic- and thermal-induced SGs. Finally, we identified SNRNP200 in cytoplasmic aggregates in amyotrophic lateral sclerosis (ALS) spinal cord and motor cortex. Collectively, our findings provide the first description of the Caprin-1 protein interactome, identify novel cytoplasmic SG components, and reveal a SG protein in cytoplasmic aggregates in ALS patient neurons. Proteomic data collected in this study are available via ProteomeXchange with identifier PXD023271.
Collapse
Affiliation(s)
- Lucas Vu
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Asmita Ghosh
- Department of Neurosciences, Université de Montréal, Montreal, QC, Canada
- CHUM Research Center, Montréal, QC, Canada
| | - Chelsea Tran
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, AZ, USA
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Walters Aji Tebung
- Department of Neurosciences, Université de Montréal, Montreal, QC, Canada
- CHUM Research Center, Montréal, QC, Canada
| | - Hadjara Sidibé
- Department of Neurosciences, Université de Montréal, Montreal, QC, Canada
- CHUM Research Center, Montréal, QC, Canada
| | - Krystine Garcia-Mansfield
- Collaborative Center for Translational Mass Spectrometry, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Victoria David-Dirgo
- Collaborative Center for Translational Mass Spectrometry, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Ritin Sharma
- Collaborative Center for Translational Mass Spectrometry, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Patrick Pirrotte
- Collaborative Center for Translational Mass Spectrometry, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Robert Bowser
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Christine Vande Velde
- Department of Neurosciences, Université de Montréal, Montreal, QC, Canada
- CHUM Research Center, Montréal, QC, Canada
| |
Collapse
|
132
|
ERK1/2 kinases and dopamine D2 receptors participate in the anticonvulsant effects of a new derivative of benzoylpyridine oxime and valproic acid. Eur J Pharmacol 2021; 903:174150. [PMID: 33961874 DOI: 10.1016/j.ejphar.2021.174150] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 04/26/2021] [Accepted: 04/30/2021] [Indexed: 01/11/2023]
Abstract
Inhibition of the activity of extracellular signal-regulated kinases (ERK1/2) induced by the activation of the dopamine D2 receptor signalling cascade may be a promising pharmacological target. The aim of this work was to study the involvement of ERK1/2 and dopamine D2 receptor in the mechanism of the anticonvulsant action of valproic acid (VA) and a new benzoylpyridine oxime derivative (GIZH-298), which showed antiepileptic activity in different models of epilepsy. We showed that subchronic exposure to maximal electroshock seizures (MES) for 5 days reduced the density of dopamine D2 receptors in the striatum of mice. GIZH-298 counteracted the decrease in the number of dopamine D2 receptors associated with MES and increased the number of ligand binding sites of dopamine D2 receptors in mice without MES. The affinity of dopamine D2 receptors to the ligand was not changed by GIZH-298. MES caused an increase in ERK1/2 and synapsin I phosphorylation in the striatum while GIZH-298, similar to VA, reduced the levels of both phospho-ERK1/2 and phosphosynapsin I after MES, which correlated with the decrease in the intensity of seizure in mice. In addition, GIZH-298 suppressed ERK1/2 phosphorylation in SH-SY5Y human neuroblastoma cells at therapeutic concentrations, while VA inhibited ERK1/2 phosphorylation in vivo but not in vitro. The data obtained expand the understanding of the mechanisms of action of VA and GIZH-298, which involve regulating the activity of ERK1/2 kinases, probably by modulating dopamine D2 receptors in limbic structures, as well as (in the case of GIZH-298) directly inhibiting of the ERK1/2 cascade.
Collapse
|
133
|
Krstic A, Konietzny A, Halasz M, Cain P, Oppermann U, Kolch W, Duffy DJ. A Chemo-Genomic Approach Identifies Diverse Epigenetic Therapeutic Vulnerabilities in MYCN-Amplified Neuroblastoma. Front Cell Dev Biol 2021; 9:612518. [PMID: 33968920 PMCID: PMC8097097 DOI: 10.3389/fcell.2021.612518] [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: 09/30/2020] [Accepted: 03/25/2021] [Indexed: 11/13/2022] Open
Abstract
Although a rare disease, neuroblastoma accounts for the highest proportion of childhood cancer deaths. There is a lack of recurrent somatic mutations in neuroblastoma embryonal tumours, suggesting a possible role for epigenetic alterations in driving this cancer. While an increasing number of reports suggest an association of MYCN with epigenetic machinery, the mechanisms of these interactions are poorly understood in the neuroblastoma setting. Utilising chemo-genomic approaches we revealed global MYCN-epigenetic interactions and identified numerous epigenetic proteins as MYCN targets. The epigenetic regulators HDAC2, CBX8 and CBP (CREBBP) were all MYCN target genes and also putative MYCN interactors. MYCN-related epigenetic genes included SMARCs, HDACs, SMYDs, BRDs and CREBBP. Expression levels of the majority of MYCN-related epigenetic genes showed predictive ability for neuroblastoma patient outcome. Furthermore, a compound library screen targeting epigenetic proteins revealed broad susceptibility of neuroblastoma cells to all classes of epigenetic regulators, belonging to families of bromodomains, HDACs, HATs, histone methyltransferases, DNA methyltransferases and lysin demethylases. Ninety-six percent of the compounds reduced MYCN-amplified neuroblastoma cell viability. We show that the C646 (CBP-bromodomain targeting compound) exhibits switch-like temporal and dose response behaviour and is effective at reducing neuroblastoma viability. Responsiveness correlates with MYCN expression, with MYCN-amplified cells being more susceptible to C646 treatment. Thus, exploiting the broad vulnerability of neuroblastoma cells to epigenetic targeting compounds represents an exciting strategy in neuroblastoma treatment, particularly for high-risk MYCN-amplified tumours.
Collapse
Affiliation(s)
- Aleksandar Krstic
- Systems Biology Ireland and Precision Oncology Ireland, School of Medicine, University College Dublin, Dublin, Ireland
| | - Anja Konietzny
- Systems Biology Ireland and Precision Oncology Ireland, School of Medicine, University College Dublin, Dublin, Ireland.,Centre for Molecular Neurobiology Hamburg (ZMNH), Emmy-Noether Group "Neuronal Protein Transport", University Medical Centre Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Melinda Halasz
- Systems Biology Ireland and Precision Oncology Ireland, School of Medicine, University College Dublin, Dublin, Ireland
| | - Peter Cain
- Botnar Research Centre, NIHR Oxford Biomedical Research Unit, Institute of Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom.,Centre for Medicines Discovery, University of Oxford, Oxford, United Kingdom
| | - Udo Oppermann
- Botnar Research Centre, NIHR Oxford Biomedical Research Unit, Institute of Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom.,Centre for Medicines Discovery, University of Oxford, Oxford, United Kingdom
| | - Walter Kolch
- Systems Biology Ireland and Precision Oncology Ireland, School of Medicine, University College Dublin, Dublin, Ireland.,Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin, Ireland
| | - David J Duffy
- Systems Biology Ireland and Precision Oncology Ireland, School of Medicine, University College Dublin, Dublin, Ireland.,The Whitney Laboratory for Marine Bioscience and Sea Turtle Hospital, University of Florida, St. Augustine, FL, United States.,Department of Biology, University of Florida, Gainesville, FL, United States
| |
Collapse
|
134
|
Bell M, Zempel H. SH-SY5Y-derived neurons: a human neuronal model system for investigating TAU sorting and neuronal subtype-specific TAU vulnerability. Rev Neurosci 2021; 33:1-15. [PMID: 33866701 DOI: 10.1515/revneuro-2020-0152] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 03/06/2021] [Indexed: 11/15/2022]
Abstract
The microtubule-associated protein (MAP) TAU is mainly sorted into the axon of healthy brain neurons. Somatodendritic missorting of TAU is a pathological hallmark of many neurodegenerative diseases, including Alzheimer's disease (AD). Cause, consequence and (patho)physiological mechanisms of TAU sorting and missorting are understudied, in part also because of the lack of readily available human neuronal model systems. The human neuroblastoma cell line SH-SY5Y is widely used for studying TAU physiology and TAU-related pathology in AD and related tauopathies. SH-SY5Y cells can be differentiated into neuron-like cells (SH-SY5Y-derived neurons) using various substances. This review evaluates whether SH-SY5Y-derived neurons are a suitable model for (i) investigating intracellular TAU sorting in general, and (ii) with respect to neuron subtype-specific TAU vulnerability. (I) SH-SY5Y-derived neurons show pronounced axodendritic polarity, high levels of axonally localized TAU protein, expression of all six human brain isoforms and TAU phosphorylation similar to the human brain. As SH-SY5Y cells are highly proliferative and readily accessible for genetic engineering, stable transgene integration and leading-edge genome editing are feasible. (II) SH-SY5Y-derived neurons display features of subcortical neurons early affected in many tauopathies. This allows analyzing brain region-specific differences in TAU physiology, also in the context of differential vulnerability to TAU pathology. However, several limitations should be considered when using SH-SY5Y-derived neurons, e.g., the lack of clearly defined neuronal subtypes, or the difficulty of mimicking age-related tauopathy risk factors in vitro. In brief, this review discusses the suitability of SH-SY5Y-derived neurons for investigating TAU (mis)sorting mechanisms and neuron-specific TAU vulnerability in disease paradigms.
Collapse
Affiliation(s)
- Michael Bell
- Institute of Human Genetics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 34, 50931Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Robert-Koch-Str. 21, 50931Cologne, Germany
| | - Hans Zempel
- Institute of Human Genetics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 34, 50931Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Robert-Koch-Str. 21, 50931Cologne, Germany
| |
Collapse
|
135
|
Naia L, Pinho CM, Dentoni G, Liu J, Leal NS, Ferreira DMS, Schreiner B, Filadi R, Fão L, Connolly NMC, Forsell P, Nordvall G, Shimozawa M, Greotti E, Basso E, Theurey P, Gioran A, Joselin A, Arsenian-Henriksson M, Nilsson P, Rego AC, Ruas JL, Park D, Bano D, Pizzo P, Prehn JHM, Ankarcrona M. Neuronal cell-based high-throughput screen for enhancers of mitochondrial function reveals luteolin as a modulator of mitochondria-endoplasmic reticulum coupling. BMC Biol 2021; 19:57. [PMID: 33761951 PMCID: PMC7989211 DOI: 10.1186/s12915-021-00979-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 02/11/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Mitochondrial dysfunction is a common feature of aging, neurodegeneration, and metabolic diseases. Hence, mitotherapeutics may be valuable disease modifiers for a large number of conditions. In this study, we have set up a large-scale screening platform for mitochondrial-based modulators with promising therapeutic potential. RESULTS Using differentiated human neuroblastoma cells, we screened 1200 FDA-approved compounds and identified 61 molecules that significantly increased cellular ATP without any cytotoxic effect. Following dose response curve-dependent selection, we identified the flavonoid luteolin as a primary hit. Further validation in neuronal models indicated that luteolin increased mitochondrial respiration in primary neurons, despite not affecting mitochondrial mass, structure, or mitochondria-derived reactive oxygen species. However, we found that luteolin increased contacts between mitochondria and endoplasmic reticulum (ER), contributing to increased mitochondrial calcium (Ca2+) and Ca2+-dependent pyruvate dehydrogenase activity. This signaling pathway likely contributed to the observed effect of luteolin on enhanced mitochondrial complexes I and II activities. Importantly, we observed that increased mitochondrial functions were dependent on the activity of ER Ca2+-releasing channels inositol 1,4,5-trisphosphate receptors (IP3Rs) both in neurons and in isolated synaptosomes. Additionally, luteolin treatment improved mitochondrial and locomotory activities in primary neurons and Caenorhabditis elegans expressing an expanded polyglutamine tract of the huntingtin protein. CONCLUSION We provide a new screening platform for drug discovery validated in vitro and ex vivo. In addition, we describe a novel mechanism through which luteolin modulates mitochondrial activity in neuronal models with potential therapeutic validity for treatment of a variety of human diseases.
Collapse
Affiliation(s)
- Luana Naia
- Center for Alzheimer Research, Division of Neurogeriatrics, Department of Neurobiology Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Catarina M Pinho
- Center for Alzheimer Research, Division of Neurogeriatrics, Department of Neurobiology Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Giacomo Dentoni
- Center for Alzheimer Research, Division of Neurogeriatrics, Department of Neurobiology Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Jianping Liu
- Department of Medicine-Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Nuno Santos Leal
- Center for Alzheimer Research, Division of Neurogeriatrics, Department of Neurobiology Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Duarte M S Ferreira
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Bernadette Schreiner
- Center for Alzheimer Research, Division of Neurogeriatrics, Department of Neurobiology Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Riccardo Filadi
- Department of Biomedical Sciences, University of Padua, Padua, Italy
- Neuroscience Institute, National Research Council (CNR), 35131, Padua, Italy
| | - Lígia Fão
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Niamh M C Connolly
- Royal College of Surgeons in Ireland, Department of Physiology & Medical Physics Department, Dublin, Ireland
| | | | | | - Makoto Shimozawa
- Center for Alzheimer Research, Division of Neurogeriatrics, Department of Neurobiology Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Elisa Greotti
- Department of Biomedical Sciences, University of Padua, Padua, Italy
- Neuroscience Institute, National Research Council (CNR), 35131, Padua, Italy
| | - Emy Basso
- Department of Biomedical Sciences, University of Padua, Padua, Italy
- Neuroscience Institute, National Research Council (CNR), 35131, Padua, Italy
| | - Pierre Theurey
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Anna Gioran
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Alvin Joselin
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | | | - Per Nilsson
- Center for Alzheimer Research, Division of Neurogeriatrics, Department of Neurobiology Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - A Cristina Rego
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Faculty of Medicine, Institute of Biochemistry, University of Coimbra, Coimbra, Portugal
| | - Jorge L Ruas
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - David Park
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Daniele Bano
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Paola Pizzo
- Department of Biomedical Sciences, University of Padua, Padua, Italy
- Neuroscience Institute, National Research Council (CNR), 35131, Padua, Italy
| | - Jochen H M Prehn
- Royal College of Surgeons in Ireland, Department of Physiology & Medical Physics Department, Dublin, Ireland
| | - Maria Ankarcrona
- Center for Alzheimer Research, Division of Neurogeriatrics, Department of Neurobiology Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
136
|
Thomson AC, Schuhmann T, de Graaf TA, Sack AT, Rutten BPF, Kenis G. The Effects of Serum Removal on Gene Expression and Morphological Plasticity Markers in Differentiated SH-SY5Y Cells. Cell Mol Neurobiol 2021; 42:1829-1839. [PMID: 33656634 PMCID: PMC9239930 DOI: 10.1007/s10571-021-01062-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 02/12/2021] [Indexed: 11/06/2022]
Abstract
Despite the widespread use of the SH-SY5Y human neuroblastoma cell line in modeling human neurons in vitro, protocols for growth, differentiation and experimentation differ considerably across the literature. Many studies fully differentiate SH-SY5Y cells before experimentation, to investigate plasticity measures in a mature, human neuronal-like cell model. Prior to experimentation, serum is often removed from cell culture media, to arrest the cell growth cycle and synchronize cells. However, the exact effect of this serum removal before experimentation on mature, differentiated SH-SY5Y cells has not yet been described. In studies using differentiated SH-SY5Y cells, any effect of serum removal on plasticity markers may influence results. The aim of the current study was to systematically characterize, in differentiated, neuronal-like SH-SY5Y cells, the potentially confounding effects of complete serum removal in terms of morphological and gene expression markers of plasticity. We measured changes in commonly used morphological markers and in genes related to neuroplasticity and synaptogenesis, particularly in the BDNF-TrkB signaling pathway. We found that complete serum removal from already differentiated SH-SY5Y cells increases neurite length, neurite branching, and the proportion of cells with a primary neurite, as well as proportion of βIII-Tubulin and MAP2 expressing cells. Gene expression results also indicate increased expression of PSD95 and NTRK2 expression 24 h after serum removal. We conclude that serum deprivation in differentiated SH-SY5Y cells affects morphology and gene expression and can potentially confound plasticity-related outcome measures, having significant implications for experimental design in studies using differentiated SH-SY5Y cells as a model of human neurons.
Collapse
Affiliation(s)
- Alix C Thomson
- Section Brain Stimulation and Cognition, Department of Cognitive Neuroscience, Faculty of Psychology and Neuroscience, Maastricht University, Oxfordlaan 55, Maastricht, The Netherlands. .,Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Brain+Nerve Centre, Maastricht University Medical Centre+ (MUMC+), Maastricht, The Netherlands. .,Maastricht Brain Imaging Centre (MBIC), Maastricht University, Maastricht, The Netherlands. .,Centre for Integrative Neuroscience, Faculty of Psychology and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands.
| | - Teresa Schuhmann
- Section Brain Stimulation and Cognition, Department of Cognitive Neuroscience, Faculty of Psychology and Neuroscience, Maastricht University, Oxfordlaan 55, Maastricht, The Netherlands.,Maastricht Brain Imaging Centre (MBIC), Maastricht University, Maastricht, The Netherlands.,Centre for Integrative Neuroscience, Faculty of Psychology and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Tom A de Graaf
- Section Brain Stimulation and Cognition, Department of Cognitive Neuroscience, Faculty of Psychology and Neuroscience, Maastricht University, Oxfordlaan 55, Maastricht, The Netherlands.,Maastricht Brain Imaging Centre (MBIC), Maastricht University, Maastricht, The Netherlands.,Centre for Integrative Neuroscience, Faculty of Psychology and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Alexander T Sack
- Section Brain Stimulation and Cognition, Department of Cognitive Neuroscience, Faculty of Psychology and Neuroscience, Maastricht University, Oxfordlaan 55, Maastricht, The Netherlands.,Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Brain+Nerve Centre, Maastricht University Medical Centre+ (MUMC+), Maastricht, The Netherlands.,Maastricht Brain Imaging Centre (MBIC), Maastricht University, Maastricht, The Netherlands.,Centre for Integrative Neuroscience, Faculty of Psychology and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Bart P F Rutten
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Brain+Nerve Centre, Maastricht University Medical Centre+ (MUMC+), Maastricht, The Netherlands.,Centre for Integrative Neuroscience, Faculty of Psychology and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Gunter Kenis
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Brain+Nerve Centre, Maastricht University Medical Centre+ (MUMC+), Maastricht, The Netherlands.,Centre for Integrative Neuroscience, Faculty of Psychology and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
137
|
Mangold CA, Rathbun MM, Renner DW, Kuny CV, Szpara ML. Viral infection of human neurons triggers strain-specific differences in host neuronal and viral transcriptomes. PLoS Pathog 2021; 17:e1009441. [PMID: 33750985 PMCID: PMC8016332 DOI: 10.1371/journal.ppat.1009441] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 04/01/2021] [Accepted: 03/01/2021] [Indexed: 12/11/2022] Open
Abstract
Infection with herpes simplex virus 1 (HSV-1) occurs in over half the global population, causing recurrent orofacial and/or genital lesions. Individual strains of HSV-1 demonstrate differences in neurovirulence in vivo, suggesting that viral genetic differences may impact phenotype. Here differentiated SH-SY5Y human neuronal cells were infected with one of three HSV-1 strains known to differ in neurovirulence in vivo. Host and viral RNA were sequenced simultaneously, revealing strain-specific differences in both viral and host transcription in infected neurons. Neuronal morphology and immunofluorescence data highlight the pathological changes in neuronal cytoarchitecture induced by HSV-1 infection, which may reflect host transcriptional changes in pathways associated with adherens junctions, integrin signaling, and others. Comparison of viral protein levels in neurons and epithelial cells demonstrated that a number of differences were neuron-specific, suggesting that strain-to-strain variations in host and virus transcription are cell type-dependent. Together, these data demonstrate the importance of studying virus strain- and cell-type-specific factors that may contribute to neurovirulence in vivo, and highlight the specificity of HSV-1-host interactions.
Collapse
Affiliation(s)
- Colleen A. Mangold
- Departments of Biology, Biochemistry and Molecular Biology, Center for Infectious Disease Dynamics, and the Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, Pennsylvania, United States of America
- Department of Entomology, College of Agricultural Sciences, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Molly M. Rathbun
- Departments of Biology, Biochemistry and Molecular Biology, Center for Infectious Disease Dynamics, and the Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Daniel W. Renner
- Departments of Biology, Biochemistry and Molecular Biology, Center for Infectious Disease Dynamics, and the Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Chad V. Kuny
- Departments of Biology, Biochemistry and Molecular Biology, Center for Infectious Disease Dynamics, and the Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Moriah L. Szpara
- Departments of Biology, Biochemistry and Molecular Biology, Center for Infectious Disease Dynamics, and the Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, Pennsylvania, United States of America
| |
Collapse
|
138
|
Head SA, Hernandez-Alias X, Yang JS, Ciampi L, Beltran-Sastre V, Torres-Méndez A, Irimia M, Schaefer MH, Serrano L. Silencing of SRRM4 suppresses microexon inclusion and promotes tumor growth across cancers. PLoS Biol 2021; 19:e3001138. [PMID: 33621242 PMCID: PMC7935315 DOI: 10.1371/journal.pbio.3001138] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 03/05/2021] [Accepted: 02/04/2021] [Indexed: 01/14/2023] Open
Abstract
RNA splicing is widely dysregulated in cancer, frequently due to altered expression or activity of splicing factors (SFs). Microexons are extremely small exons (3–27 nucleotides long) that are highly evolutionarily conserved and play critical roles in promoting neuronal differentiation and development. Inclusion of microexons in mRNA transcripts is mediated by the SF Serine/Arginine Repetitive Matrix 4 (SRRM4), whose expression is largely restricted to neural tissues. However, microexons have been largely overlooked in prior analyses of splicing in cancer, as their small size necessitates specialized computational approaches for their detection. Here, we demonstrate that despite having low expression in normal nonneural tissues, SRRM4 is further silenced in tumors, resulting in the suppression of normal microexon inclusion. Remarkably, SRRM4 is the most consistently silenced SF across all tumor types analyzed, implying a general advantage of microexon down-regulation in cancer independent of its tissue of origin. We show that this silencing is favorable for tumor growth, as decreased SRRM4 expression in tumors is correlated with an increase in mitotic gene expression, and up-regulation of SRRM4 in cancer cell lines dose-dependently inhibits proliferation in vitro and in a mouse xenograft model. Further, this proliferation inhibition is accompanied by induction of neural-like expression and splicing patterns in cancer cells, suggesting that SRRM4 expression shifts the cell state away from proliferation and toward differentiation. We therefore conclude that SRRM4 acts as a proliferation brake, and tumors gain a selective advantage by cutting off this brake. Using data from The Cancer Genome Atlas, this study shows that the splicing factor SRRM4 and its program of differentiation-promoting microexons are downregulated across tumor types with remarkable consistency, providing tumors with a proliferative advantage.
Collapse
Affiliation(s)
- Sarah A. Head
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
- * E-mail: (SAH); (MHS); (LS)
| | - Xavier Hernandez-Alias
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Jae-Seong Yang
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Centre de Recerca en Agrigenòmica, Consortium CSIC-IRTA-UAB-UB, Cerdanyola del Vallès, Barcelona, Spain
| | - Ludovica Ciampi
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Violeta Beltran-Sastre
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Antonio Torres-Méndez
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Manuel Irimia
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- ICREA, Barcelona, Spain
| | - Martin H. Schaefer
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
- IEO European Institute of Oncology IRCCS, Department of Experimental Oncology, Milan, Italy
- * E-mail: (SAH); (MHS); (LS)
| | - Luis Serrano
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- ICREA, Barcelona, Spain
- * E-mail: (SAH); (MHS); (LS)
| |
Collapse
|
139
|
Generation of APOE knock-down SK-N-SH human neuroblastoma cells using CRISPR/Cas9: a novel cellular model relevant to Alzheimer's disease research. Biosci Rep 2021; 41:227846. [PMID: 33600562 PMCID: PMC7897917 DOI: 10.1042/bsr20204243] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 02/08/2021] [Accepted: 02/10/2021] [Indexed: 12/26/2022] Open
Abstract
APOE ε4 is the major genetic risk factor for Alzheimer’s disease (AD). A precise role for apolipoprotein E (apoE) in the pathogenesis of the disease remains unclear in part due to its expression in multiple cell types of the brain. APOE is highly expressed in astrocytes and microglia, however its expression can also be induced in neurons under various conditions. The neuron-like cell line SK-N-SH is a useful model in the study of the cellular and molecular effects of apoE as it can be differentiated with retinoic acid to express and secrete high levels of apoE and it also shows the same apoE fragmentation patterns observed in the human brain. We previously found that apoE is cleaved into a 25-kDa fragment by high temperature-requirement serine protease A1 (HtrA1) in SK-N-SH cells. To further understand the endogenous functions of apoE, we used CRISPR/Cas9 to generate SK-N-SH cell lines with APOE expression knocked-down (KD). APOE KD cells showed lower APOE and HTRA1 expression than parental SK-N-SH cells but no overt differences in neuritogenesis or cell proliferation compared with the CRISPR/Cas9 control cells. This research shows that the loss of apoE and HtrA1 has a negligible effect on neuritogenesis and cell survival in SK-N-SH neuron-like cells.
Collapse
|
140
|
Impact of DJ-1 and Helix 8 on the Proteome and Degradome of Neuron-Like Cells. Cells 2021; 10:cells10020404. [PMID: 33669258 PMCID: PMC7920061 DOI: 10.3390/cells10020404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 02/08/2021] [Accepted: 02/10/2021] [Indexed: 12/04/2022] Open
Abstract
DJ-1 is an abundant and ubiquitous component of cellular proteomes. DJ-1 supposedly exerts a wide variety of molecular functions, ranging from enzymatic activities as a deglycase, protease, and esterase to chaperone functions. However, a consensus perspective on its molecular function in the cellular context has not yet been reached. Structurally, the C-terminal helix 8 of DJ-1 has been proposed to constitute a propeptide whose proteolytic removal transforms a DJ-1 zymogen to an active hydrolase with potential proteolytic activity. To better understand the cell-contextual functionality of DJ-1 and the role of helix 8, we employed post-mitotically differentiated, neuron-like SH-SY5Y neuroblastoma cells with stable over-expression of full length DJ-1 or DJ-1 lacking helix 8 (ΔH8), either with a native catalytically active site (C106) or an inactive site (C106A active site mutation). Global proteome comparison of cells over-expressing DJ-1 ΔH8 with native or mutated active site cysteine indicated a strong impact on mitochondrial biology. N-terminomic profiling however did not highlight direct protease substrate candidates for DJ-1 ΔH8, but linked DJ-1 to elevated levels of activated lysosomal proteases, albeit presumably in an indirect manner. Finally, we show that DJ-1 ΔH8 loses the deglycation activity of full length DJ-1. Our study further establishes DJ-1 as deglycation enzyme. Helix 8 is essential for the deglycation activity but dispensable for the impact on lysosomal and mitochondrial biology; further illustrating the pleiotropic nature of DJ-1.
Collapse
|
141
|
Jacob RS, Eichmann C, Dema A, Mercadante D, Selenko P. α-Synuclein plasma membrane localization correlates with cellular phosphatidylinositol polyphosphate levels. eLife 2021; 10:61951. [PMID: 33587036 PMCID: PMC7929559 DOI: 10.7554/elife.61951] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 02/12/2021] [Indexed: 12/11/2022] Open
Abstract
The Parkinson's disease protein α-synuclein (αSyn) promotes membrane fusion and fission by interacting with various negatively charged phospholipids. Despite postulated roles in endocytosis and exocytosis, plasma membrane (PM) interactions of αSyn are poorly understood. Here, we show that phosphatidylinositol 4,5-bisphosphate (PIP2) and phosphatidylinositol 3,4,5-trisphosphate (PIP3), two highly acidic components of inner PM leaflets, mediate PM localization of endogenous pools of αSyn in A2780, HeLa, SK-MEL-2, and differentiated and undifferentiated neuronal SH-SY5Y cells. We demonstrate that αSyn binds to reconstituted PIP2 membranes in a helical conformation in vitro and that PIP2 synthesizing kinases and hydrolyzing phosphatases reversibly redistribute αSyn in cells. We further delineate that αSyn-PM targeting follows phosphoinositide-3 kinase (PI3K)-dependent changes of cellular PIP2 and PIP3 levels, which collectively suggests that phosphatidylinositol polyphosphates contribute to αSyn's function(s) at the plasma membrane.
Collapse
Affiliation(s)
- Reeba Susan Jacob
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Cédric Eichmann
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Alessandro Dema
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Davide Mercadante
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Philipp Selenko
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
142
|
Pavlidis N, Kofinas A, Papanikolaou MG, Miras HN, Drouza C, Kalampounias AG, Kabanos TA, Konstandi M, Leondaritis G. Synthesis, characterization and pharmacological evaluation of quinoline derivatives and their complexes with copper(ΙΙ) in in vitro cell models of Alzheimer's disease. J Inorg Biochem 2021; 217:111393. [PMID: 33610031 DOI: 10.1016/j.jinorgbio.2021.111393] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/30/2021] [Accepted: 02/05/2021] [Indexed: 12/17/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder of the central nervous system. The main pathophysiological mechanisms involve cholinergic neurotransmission, beta-amyloid (Αβ) and Tau proteins, several metal ions and oxidative stress, among others. Current drugs offer only relief of symptoms and not a cure of AD. Accumulating evidence suggests that multifunctional compounds, targeting multiple pathophysiological mechanisms, may have a great potential for the treatment of AD. In this study, we report on the synthesis and physicochemical characterization of four quinoline-based metal chelators and their respective copper(II) complexes. Most compounds were non-toxic at concentrations ≤5 μM. In neuroprotection studies employing undifferentiated and differentiated SH-SY5Y cells, the metal chelator N2,N6-di(quinolin-8-yl)pyridine-2,6-dicarboxamide (H2dqpyca) appeared to exert significant neuroprotection against both, Aβ peptide- and H2O2-induced toxicities. The copper(II) complex [CuII(H2bqch)Cl2].3H2O (H2bqch = N,N'-Bis(8-quinolyl)cyclohexane-1,2-diamine) also protected against H2O2-induced toxicity, with a half-maximal effective concentration of 80 nM. Molecular docking simulations, using the crystal structure of the acetylcholinesterase (AChE)-rivastigmine complex as a template, indicated a strong interaction of the metal chelator H2dqpyca, followed by H2bqch, with both the peripheral anionic site and the catalytic active site of AChE. In conclusion, the sufficient neuroprotection provided by the metal chelator H2dqpyca and the copper(II) complex [CuII(H2bqch)Cl2].3H2O along with the evidence for interaction between H2dqpyca and AChE, indicate that these compounds have the potential and should be further investigated in the framework of preclinical studies employing animal models of AD as candidate multifunctional lead compounds for the treatment of the disease.
Collapse
Affiliation(s)
- Nikolaos Pavlidis
- Department of Pharmacology, Faculty of Medicine, University of Ioannina, Ioannina 45110, Greece; Section of Inorganic and Analytical Chemistry, Department of Chemistry, University of Ioannina, Ioannina 45110, Greece.
| | - Aristeidis Kofinas
- Department of Pharmacology, Faculty of Medicine, University of Ioannina, Ioannina 45110, Greece.
| | - Michael G Papanikolaou
- Section of Inorganic and Analytical Chemistry, Department of Chemistry, University of Ioannina, Ioannina 45110, Greece.
| | - Haralampos N Miras
- West CHEM, School of Chemistry, University of Glasgow, Glasgow G12 8QQ, UK.
| | - Chryssoula Drouza
- Department of Agricultural Sciences, Biotechnology and Food Science, Cyprus University of Technology, Limassol 3036, Cyprus.
| | - Angelos G Kalampounias
- Physical Chemistry Laboratory, Department of Chemistry, University of Ioannina, Ioannina 45110, Greece; Institute of Materials Science and Computing, University Research Center of Ioannina (URCI), Ioannina 45110, Greece.
| | - Themistoklis A Kabanos
- Section of Inorganic and Analytical Chemistry, Department of Chemistry, University of Ioannina, Ioannina 45110, Greece.
| | - Maria Konstandi
- Department of Pharmacology, Faculty of Medicine, University of Ioannina, Ioannina 45110, Greece.
| | - George Leondaritis
- Department of Pharmacology, Faculty of Medicine, University of Ioannina, Ioannina 45110, Greece.
| |
Collapse
|
143
|
Mallach A, Gobom J, Zetterberg H, Hardy J, Piers TM, Wray S, Pocock JM. The influence of the R47H triggering receptor expressed on myeloid cells 2 variant on microglial exosome profiles. Brain Commun 2021; 3:fcab009. [PMID: 34704019 PMCID: PMC8244649 DOI: 10.1093/braincomms/fcab009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 12/03/2020] [Accepted: 12/04/2020] [Indexed: 01/08/2023] Open
Abstract
Variants in the triggering receptor expressed on myeloid cells 2 gene are linked with an increased risk of dementia, in particular the R47Hhet triggering receptor expressed on myeloid cells 2 variant is linked to late-onset Alzheimer's disease. Using human induced pluripotent stem cells-derived microglia, we assessed whether variations in the dynamics of exosome secretion, including their components, from these cells might underlie some of this risk. We found exosome size was not altered between common variant controls and R47Hhet variants, but the amount and constitution of exosomes secreted were different. Exosome quantities were rescued by incubation with an ATP donor or with lipids via a phosphatidylserine triggering receptor expressed on myeloid cells 2 ligand. Following a lipopolysaccharide or phagocytic cell stimulus, exosomes from common variant and R47Hhet microglia were found to contain cytokines, chemokines, APOE and triggering receptor expressed on myeloid cells 2. Differences were observed in the expression of CCL22, IL-1β and triggering receptor expressed on myeloid cells 2 between common variant and R47Hhet derived exosomes. Furthermore unlike common variant-derived exosomes, R47Hhet exosomes contained additional proteins linked to negative regulation of transcription and metabolic processes. Subsequent addition of exosomes to stressed neurones showed R47Hhet-derived exosomes to be less protective. These data have ramifications for the responses of microglia in Alzheimer's disease and may point to further targets for therapeutic intervention.
Collapse
Affiliation(s)
- Anna Mallach
- Department of Neuroinflammation, Queen Square Institute of Neurology, University College London, London WC1N 1PJ, UK
| | - Johan Gobom
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, S-431 80, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, S-431 80, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, S-431 80, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, S-431 80, Mölndal, Sweden
- Department of Neurodegenerative Disease, University College London Queen Square Institute of Neurology, London WC1N 1PJ, UK
- Department of Neurodegenerative Disease, University College London Queen Square Institute of Neurology, London WC1N 3BG, UK
- UK Dementia Research Institute University College London, London WC1E 6BT, UK
| | - John Hardy
- Department of Neurodegenerative Disease, University College London Queen Square Institute of Neurology, London WC1N 1PJ, UK
- Department of Neurodegenerative Disease, University College London Queen Square Institute of Neurology, London WC1N 3BG, UK
- UK Dementia Research Institute University College London, London WC1E 6BT, UK
- Reta Lila Weston Institute, UCL Queen Square Institute of Neurology, London WC1N 1PJ, UK
- NIHR University College London Hospitals Biomedical Research Centre, London, UK
- Institute for Advanced Study, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Thomas M Piers
- Department of Neuroinflammation, Queen Square Institute of Neurology, University College London, London WC1N 1PJ, UK
| | - Selina Wray
- Department of Neurodegenerative Disease, University College London Queen Square Institute of Neurology, London WC1N 1PJ, UK
- Department of Neurodegenerative Disease, University College London Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Jennifer M Pocock
- Department of Neuroinflammation, Queen Square Institute of Neurology, University College London, London WC1N 1PJ, UK
| |
Collapse
|
144
|
Amat-ur-Rasool H, Ahmed M, Hasnain S, Carter WG. Anti-Cholinesterase Combination Drug Therapy as a Potential Treatment for Alzheimer's Disease. Brain Sci 2021; 11:184. [PMID: 33540879 PMCID: PMC7913148 DOI: 10.3390/brainsci11020184] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/27/2021] [Accepted: 01/30/2021] [Indexed: 01/29/2023] Open
Abstract
Alzheimer's disease (AD) is a burgeoning social and healthcare problem. Cholinesterase inhibitors (ChEIs) are employed for symptomatic treatment of AD, but often elicit adverse drug reactions (ADRs). Herein, the potency of the ChEIs, donepezil, tacrine, berberine, and galantamine to inhibit human or Torpedo californica acetylcholinesterase (tcAChE) proteins were evaluated. The efficacy of dual-drug combinations to inhibit human AChE directly and within differentiated neurons was also quantified. ChEI potency was in the order: donepezil > tacrine > berberine > galantamine for both AChEs. Dual-drug combinations of berberine and tacrine (BerTac), berberine and galantamine (BerGal), and tacrine and donepezil (TacDon) all produced synergistic outcomes for AChE inhibition. Donepezil and berberine (DonBer) and tacrine and galantamine (TacGal) elicited antagonistic responses. Donepezil and galantamine (DonGal) was synergistic for human AChE but antagonistic for tcAChE. After application of dual-drug combinations to neuronal cells, BerTac, BerGal, DonGal, and DonBer all showed synergistic inhibition of AChE, TacDon additive, and TacGal antagonistic effects. BerGal produced the most potent synergism and reduced total drug dose by 72%. Individual ChEIs or dual-drug combinations were relatively non-toxic to neuronal cells, and only reduced cell viability at concentrations two-three orders of magnitude greater than that required to inhibit AChE. In summary, dual-drug combinations of ChEIs potentially represent a novel means of AD patient treatment, with reduced and more cost-effective drug dosing, and lowered likelihood of ADRs.
Collapse
Affiliation(s)
- Hafsa Amat-ur-Rasool
- Royal Derby Hospital Centre, School of Medicine, University of Nottingham, Derby DE22 3DT, UK;
- Department of Microbiology and Molecular Genetics, University of the Punjab, Lahore 54590, Pakistan; (M.A.); (S.H.)
| | - Mehboob Ahmed
- Department of Microbiology and Molecular Genetics, University of the Punjab, Lahore 54590, Pakistan; (M.A.); (S.H.)
| | - Shahida Hasnain
- Department of Microbiology and Molecular Genetics, University of the Punjab, Lahore 54590, Pakistan; (M.A.); (S.H.)
| | - Wayne G. Carter
- Royal Derby Hospital Centre, School of Medicine, University of Nottingham, Derby DE22 3DT, UK;
| |
Collapse
|
145
|
Ke M, Chong CM, Zhu Q, Zhang K, Cai CZ, Lu JH, Qin D, Su H. Comprehensive Perspectives on Experimental Models for Parkinson's Disease. Aging Dis 2021; 12:223-246. [PMID: 33532138 PMCID: PMC7801282 DOI: 10.14336/ad.2020.0331] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 03/31/2020] [Indexed: 11/19/2022] Open
Abstract
Parkinson’s disease (PD) ranks second among the most common neurodegenerative diseases, characterized by progressive and selective loss of dopaminergic neurons. Various cross-species preclinical models, including cellular models and animal models, have been established through the decades to study the etiology and mechanism of the disease from cell lines to nonhuman primates. These models are aimed at developing effective therapeutic strategies for the disease. None of the current models can replicate all major pathological and clinical phenotypes of PD. Selection of the model for PD largely relies on our interest of study. In this review, we systemically summarized experimental PD models, including cellular and animal models used in preclinical studies, to understand the pathogenesis of PD. This review is intended to provide current knowledge about the application of these different PD models, with focus on their strengths and limitations with respect to their contributions to the assessment of the molecular pathobiology of PD and identification of the therapeutic strategies for the disease.
Collapse
Affiliation(s)
- Minjing Ke
- 1State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Cheong-Meng Chong
- 1State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Qi Zhu
- 1State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Ke Zhang
- 1State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Cui-Zan Cai
- 1State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Jia-Hong Lu
- 1State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Dajiang Qin
- 2Guangzhou Regenerative Medicine and Health Guangdong Laboratory, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,3South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Huanxing Su
- 1State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| |
Collapse
|
146
|
Turkez H, Cacciatore I, Marinelli L, Fornasari E, Aslan ME, Cadirci K, Kahraman CY, Caglar O, Tatar A, Di Biase G, Hacimuftuoglu A, Di Stefano A, Mardinoglu A. Glycyl-L-Prolyl-L-Glutamate Pseudotripeptides for Treatment of Alzheimer's Disease. Biomolecules 2021; 11:biom11010126. [PMID: 33478054 PMCID: PMC7835747 DOI: 10.3390/biom11010126] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/13/2021] [Accepted: 01/15/2021] [Indexed: 12/15/2022] Open
Abstract
So far, there is no effective disease-modifying therapies for Alzheimer’s Disease (AD) in clinical practice. In this context, glycine-L-proline-L-glutamate (GPE) and its analogs may open the way for developing a novel molecule for treating neurodegenerative disorders, including AD. In turn, this study was aimed to investigate the neuroprotective potentials exerted by three novel GPE peptidomimetics (GPE1, GPE2, and GPE3) using an in vitro AD model. Anti-Alzheimer potentials were determined using a wide array of techniques, such as measurements of mitochondrial viability (MTT) and lactate dehydrogenase (LDH) release assays, determination of acetylcholinesterase (AChE), α-secretase and β-secretase activities, comparisons of total antioxidant capacity (TAC) and total oxidative status (TOS) levels, flow cytometric and microscopic detection of apoptotic and necrotic neuronal death, and investigating gene expression responses via PCR arrays involving 64 critical genes related to 10 different pathways. Our analysis showed that GPE peptidomimetics modulate oxidative stress, ACh depletion, α-secretase inactivation, apoptotic, and necrotic cell death. In vitro results suggested that treatments with novel GPE analogs might be promising therapeutic agents for treatment and/or or prevention of AD.
Collapse
Affiliation(s)
- Hasan Turkez
- Department of Medical Biology, Faculty of Medicine, Atatürk University, 25240 Erzurum, Turkey
- Correspondence: (H.T.); (A.M.)
| | - Ivana Cacciatore
- Department of Pharmacy, Univerisity “G. d’Annunzio” of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti Scalo, Chieti, Italy; (I.C.); (L.M.); (E.F.); (G.D.B.); (A.D.S.)
| | - Lisa Marinelli
- Department of Pharmacy, Univerisity “G. d’Annunzio” of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti Scalo, Chieti, Italy; (I.C.); (L.M.); (E.F.); (G.D.B.); (A.D.S.)
| | - Erika Fornasari
- Department of Pharmacy, Univerisity “G. d’Annunzio” of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti Scalo, Chieti, Italy; (I.C.); (L.M.); (E.F.); (G.D.B.); (A.D.S.)
| | - Mehmet Enes Aslan
- Department of Molecular Biology and Genetics, Faculty of Science, Erzurum Technical University, 25200 Erzurum, Turkey; (M.E.A.); (O.C.)
| | - Kenan Cadirci
- Department of Internal Medicine, Erzurum Regional Training and Research Hospital, Health Sciences University, 25200 Erzurum, Turkey;
| | - Cigdem Yuce Kahraman
- Department of Medical Genetics, Faculty of Medicine, Atatürk University, 25240 Erzurum, Turkey; (C.Y.K.); (A.T.)
| | - Ozge Caglar
- Department of Molecular Biology and Genetics, Faculty of Science, Erzurum Technical University, 25200 Erzurum, Turkey; (M.E.A.); (O.C.)
| | - Abdulgani Tatar
- Department of Medical Genetics, Faculty of Medicine, Atatürk University, 25240 Erzurum, Turkey; (C.Y.K.); (A.T.)
| | - Giuseppe Di Biase
- Department of Pharmacy, Univerisity “G. d’Annunzio” of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti Scalo, Chieti, Italy; (I.C.); (L.M.); (E.F.); (G.D.B.); (A.D.S.)
| | - Ahmet Hacimuftuoglu
- Department of Medical Pharmacology, Faculty of Medicine, Atatürk University, 25240 Erzurum, Turkey;
| | - Antonio Di Stefano
- Department of Pharmacy, Univerisity “G. d’Annunzio” of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti Scalo, Chieti, Italy; (I.C.); (L.M.); (E.F.); (G.D.B.); (A.D.S.)
| | - Adil Mardinoglu
- Science for Life Laboratory, KTH—Royal Institute of Technology, 24075 Stockholm, Sweden
- Centre for Host-Microbiome Interactions, Dental Institute, King’s College London, London SE1 9RT, UK
- Correspondence: (H.T.); (A.M.)
| |
Collapse
|
147
|
Şahin M, Öncü G, Yılmaz MA, Özkan D, Saybaşılı H. Transformation of SH-SY5Y cell line into neuron-like cells: Investigation of electrophysiological and biomechanical changes. Neurosci Lett 2021; 745:135628. [PMID: 33440235 DOI: 10.1016/j.neulet.2021.135628] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 12/14/2020] [Accepted: 01/04/2021] [Indexed: 02/07/2023]
Abstract
SH-SY5Y human neuroblastoma cells are commonly used as neuronal models. Here, we examined different aspects of SH-SY5Y cell differentiation. Various differentiation protocols have been proposed previously, including treatments with retinoic acid, brain-derived neurotrophic factor (BDNF), cholesterol and oestradiol. We examined undifferentiated SH-SY5Y cells (UNDIFF); cells differentiated by the treatment with retinoic acid (RA); retinoic acid + BDNF (RB); and retinoic acid + BDNF + cholesterol + oestradiol (RBCE). We performed whole-cell patch-clamp recordings from these cells and nanomechanically characterised them by using atomic force microscopy (AFM). Our results indicated that Na+ currents become most pronounced in the differentiated RB cells, whereas UNDIFF SH-SY5Y cells had significantly larger K+ currents, which is a characteristic feature of cancer cells. AFM observations of these two groups showed that Young's moduli of SH-SY5Y cells increased threefold with differentiation. Furthermore, we showed a direct relationship between Na+ channel activity and elasticity in these cells. We conclude that SH-SY5Y human neuroblastoma cells should be used as a neuronal model only when they are differentiated by the treatment with retinoic acid and BDNF.
Collapse
Affiliation(s)
- Meryem Şahin
- Institute of Biomedical Engineering, Boğaziçi University, Istanbul, Turkey
| | - Gül Öncü
- Institute of Biomedical Engineering, Boğaziçi University, Istanbul, Turkey
| | - Mustafa Alper Yılmaz
- Department of Mechanical Engineering, National Defense University, Naval Academy, Istanbul, Turkey
| | - Doğuş Özkan
- Department of Mechanical Engineering, National Defense University, Naval Academy, Istanbul, Turkey
| | - Hale Saybaşılı
- Institute of Biomedical Engineering, Boğaziçi University, Istanbul, Turkey.
| |
Collapse
|
148
|
Yeman KB, Isik S. Down regulation of DNA topoisomerase IIβ exerts neurodegeneration like effect through Rho GTPases in cellular model of Parkinson's disease by Down regulating tyrosine hydroxylase. Neurol Res 2021; 43:464-473. [PMID: 33402057 DOI: 10.1080/01616412.2020.1867949] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Initiating the transcriptional activation of neuronal genes, DNA topoisomerase IIβ (topo IIβ) has a crucial role in neural differentiation and brain development. Inhibition of topo IIβ activity causes shorter axons and deteriorated neuronal connections common in neurodegenerative diseases. We previously reported that topo IIβ silencing could give rise to neurodegeneration through dysregulation of Rho GTPases and may contribute to pathogenesis of neurodegenerative diseases. Although there are several studies available proposing a link between Parkinson's Disease (PD) and Rho GTPases, there have been no reports analyzing the topo IIβ-dependent association of PD and Rho GTPases. Here, for the first time, we identified that topo IIβ has a regulatory role on Rho GTPases contributing to PD-like pathology. We analyzed the association between topo IIβ and PD by comparing topo IIβ expression levels of Retinoic Acid (RA) and Brain-derived neutrophic factor (BDNF) induced and MPP+-intoxicated SH-SY5Y cells used as an in vitro PD model. While both mRNA and protein levels of topo IIβ increase in neural differentiated cells, a significant decrease is detected in the PD model. Additionally, silencing of topo IIβ by specific siRNAs caused phenotypic alterations like deteriorated neural connections and transcriptional regulations such as upregulation of RhoA and downregulation of Cdc42, Rac1, and tyrosine hydroxylase gene expressions. Our results suggest that topo IIβ downregulation may cause neurodegeneration through dysregulation of Rho-GTPases leading to PD-like pathology.
Collapse
Affiliation(s)
- Kiyak Bercem Yeman
- Department of Molecular Medicine, Institute of Health Sciences, University of Health Sciences, Istanbul, Turkey
| | - Sevim Isik
- Department of Molecular Biology and Genetics, Faculty of Engineering and Natural Sciences, Uskudar University, Istanbul, Turkey
| |
Collapse
|
149
|
Paik S, Somvanshi RK, Oliveira HA, Zou S, Kumar U. Somatostatin Ameliorates β-Amyloid-Induced Cytotoxicity via the Regulation of CRMP2 Phosphorylation and Calcium Homeostasis in SH-SY5Y Cells. Biomedicines 2021; 9:biomedicines9010027. [PMID: 33401710 PMCID: PMC7823260 DOI: 10.3390/biomedicines9010027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 12/24/2020] [Accepted: 12/25/2020] [Indexed: 11/17/2022] Open
Abstract
Somatostatin is involved in the regulation of multiple signaling pathways and affords neuroprotection in response to neurotoxins. In the present study, we investigated the role of Somatostatin-14 (SST) in cell viability and the regulation of phosphorylation of Collapsin Response Mediator Protein 2 (CRMP2) (Ser522) via the blockade of Ca2+ accumulation, along with the inhibition of cyclin-dependent kinase 5 (CDK5) and Calpain activation in differentiated SH-SY5Y cells. Cell Viability and Caspase 3/7 assays suggest that the presence of SST ameliorates mitochondrial stability and cell survival pathways while augmenting pro-apoptotic pathways activated by Aβ. SST inhibits the phosphorylation of CRMP2 at Ser522 site, which is primarily activated by CDK5. Furthermore, SST effectively regulates Ca2+ influx in the presence of Aβ, directly affecting the activity of calpain in differentiated SH-SY5Y cells. We also demonstrated that SSTR2 mediates the protective effects of SST. In conclusion, our results highlight the regulatory role of SST in intracellular Ca2+ homeostasis. The neuroprotective role of SST via axonal regeneration and synaptic integrity is corroborated by regulating changes in CRMP2; however, SST-mediated changes in the blockade of Ca2+ influx, calpain expression, and toxicity did not correlate with CDK5 expression and p35/25 accumulation. To summarize, our findings suggest two independent mechanisms by which SST mediates neuroprotection and confirms the therapeutic implications of SST in AD as well as in other neurodegenerative diseases where the effective regulation of calcium homeostasis is required for a better prognosis.
Collapse
Affiliation(s)
| | | | | | | | - Ujendra Kumar
- Correspondence: ; Tel.: +1-604-827-3660; Fax: +1-604-822-3035
| |
Collapse
|
150
|
Riegerová P, Brejcha J, Bezděková D, Chum T, Mašínová E, Čermáková N, Ovsepian SV, Cebecauer M, Štefl M. Expression and Localization of AβPP in SH-SY5Y Cells Depends on Differentiation State. J Alzheimers Dis 2021; 82:485-491. [PMID: 34057078 PMCID: PMC8385523 DOI: 10.3233/jad-201409] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/21/2021] [Indexed: 11/27/2022]
Abstract
Neuroblastoma cell line SH-SY5Y, due to its capacity to differentiate into neurons, easy handling, and low cost, is a common experimental model to study molecular events leading to Alzheimer's disease (AD). However, it is prevalently used in its undifferentiated state, which does not resemble neurons affected by the disease. Here, we show that the expression and localization of amyloid-β protein precursor (AβPP), one of the key molecules involved in AD pathogenesis, is dramatically altered in SH-SY5Y cells fully differentiated by combined treatment with retinoic acid and BDNF. We show that insufficient differentiation of SH-SY5Y cells results in AβPP mislocalization.
Collapse
Affiliation(s)
- Petra Riegerová
- Department of Biophysical Chemistry, J. Heyrovský Institute of Physical Chemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | - Jindřich Brejcha
- Department of Biophysical Chemistry, J. Heyrovský Institute of Physical Chemistry of the Czech Academy of Sciences, Prague, Czech Republic
- Department of Philosophy and History of Science, Faculty of Science, Charles University, Prague, Czech Republic
| | - Dagmar Bezděková
- Department of Experimental Neurobiology, National Institute of Mental Health, Klecany, Czech Republic
- Department of Psychiatry and Medical Psychology, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Tomáš Chum
- Department of Biophysical Chemistry, J. Heyrovský Institute of Physical Chemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | - Eva Mašínová
- Department of Biophysical Chemistry, J. Heyrovský Institute of Physical Chemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | - Nikola Čermáková
- Department of Biophysical Chemistry, J. Heyrovský Institute of Physical Chemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | - Saak V. Ovsepian
- Department of Experimental Neurobiology, National Institute of Mental Health, Klecany, Czech Republic
- Department of Psychiatry and Medical Psychology, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Marek Cebecauer
- Department of Biophysical Chemistry, J. Heyrovský Institute of Physical Chemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | - Martin Štefl
- Department of Biophysical Chemistry, J. Heyrovský Institute of Physical Chemistry of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|