1
|
Safreena N, Nair IC, Chandra G. Therapeutic potential of Parkin and its regulation in Parkinson's disease. Biochem Pharmacol 2024; 230:116600. [PMID: 39500382 DOI: 10.1016/j.bcp.2024.116600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 10/05/2024] [Accepted: 10/28/2024] [Indexed: 11/14/2024]
Abstract
Parkinson's disease (PD) is a debilitating neurodegenerative disorder characterized by the progressive loss of dopaminergic neurons in the midbrain substantia nigra, resulting in motor and non-motor symptoms. While the exact etiology of PD remains elusive, a growing body of evidence suggests that dysfunction in the parkin protein plays a pivotal role in the pathogenesis of the disease. Parkin is an E3 ubiquitin ligase that ubiquitinates substrate proteins to control a number of crucial cellular processes including protein catabolism, immune response, and cellular apoptosis.While autosomal recessive mutations in the PARK2 gene, which codes for parkin, are linked to an inherited form of early-onset PD, heterozygous mutations in PARK2 have also been reported in the more commonly occurring sporadic PD cases. Impairment of parkin's E3 ligase activity is believed to play a pathogenic role in both familial and sporadic forms of PD.This article provides an overview of the current understanding of the mechanistic basis of parkin's E3 ligase activity, its major physiological role in controlling cellular functions, and how these are disrupted in familial and sporadic PD. The second half of the manuscript explores the currently available and potential therapeutic strategies targeting parkin structure and/or function in order to slow down or mitigate the progressive neurodegeneration in PD.
Collapse
Affiliation(s)
- Narukkottil Safreena
- Cell Biology Laboratory, Center for Development and Aging Research, Inter University Center for Biomedical Research & Super Specialty Hospital, Mahatma Gandhi University Campus at Thalappady, Rubber Board PO, Kottayam 686009, Kerala, India
| | - Indu C Nair
- SAS SNDP Yogam College, Konni, Pathanamthitta 689691, Kerala, India
| | - Goutam Chandra
- Cell Biology Laboratory, Center for Development and Aging Research, Inter University Center for Biomedical Research & Super Specialty Hospital, Mahatma Gandhi University Campus at Thalappady, Rubber Board PO, Kottayam 686009, Kerala, India.
| |
Collapse
|
2
|
Miki R, Nomura R, Iijima Y, Kubota S, Takasugi N, Iwawaki T, Fujimura M, Uehara T. Therapeutic potential of 4-phenylbutyric acid against methylmercury-induced neuronal cell death in mice. Arch Toxicol 2024:10.1007/s00204-024-03902-3. [PMID: 39465421 DOI: 10.1007/s00204-024-03902-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 10/17/2024] [Indexed: 10/29/2024]
Abstract
Methylmercury (MeHg) is an environmental neurotoxin that induces damage to the central nervous system and is the causative agent in Minamata disease. The mechanisms underlying MeHg neurotoxicity remain largely unknown, and there is a need for effective therapeutic agents, such as those that target MeHg-induced endoplasmic reticulum (ER) stress and the unfolded protein response (UPR), which is activated as a defense mechanism. We investigated whether intraperitoneal administration of the chemical chaperone, 4-phenylbutyric acid (4-PBA), at 120 mg/kg/day can alleviate neurotoxicity in the brains of mice administered 50 ppm MeHg in drinking water for 5 weeks. 4-PBA significantly reduced MeHg-induced ER stress, neuronal apoptosis, and neurological symptoms. Furthermore, 4-PBA was effective even when administered 2 weeks after the initiation of exposure to 30 ppm MeHg in drinking water. Our results strongly indicate that ER stress and the UPR are key processes involved in MeHg toxicity, and that 4-PBA is a novel therapeutic candidate for MeHg-induced neurotoxicity.
Collapse
Affiliation(s)
- Ryohei Miki
- Department of Medicinal Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, 700‑8530, Japan
| | - Ryosuke Nomura
- Department of Medicinal Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, 700‑8530, Japan
| | - Yuta Iijima
- Department of Medicinal Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, 700‑8530, Japan
| | - Sho Kubota
- Department of Medicinal Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, 700‑8530, Japan
| | - Nobumasa Takasugi
- Department of Medicinal Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, 700‑8530, Japan
| | - Takao Iwawaki
- Division of Cell Medicine, Department of Life Science, Medical Research Institute, Kanazawa Medical University, Ishikawa, 920-0293, Japan
| | - Masatake Fujimura
- Department of International Affairs and Research, National Institute for Minamata Disease, Kumamoto, 867‑0008, Japan
| | - Takashi Uehara
- Department of Medicinal Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, 700‑8530, Japan.
| |
Collapse
|
3
|
Giannakou M, Akrani I, Tsoka A, Myrianthopoulos V, Mikros E, Vorgias C, Hatzinikolaou DG. Discovery of Novel Inhibitors against ALS-Related SOD1(A4V) Aggregation through the Screening of a Chemical Library Using Differential Scanning Fluorimetry (DSF). Pharmaceuticals (Basel) 2024; 17:1286. [PMID: 39458929 PMCID: PMC11510448 DOI: 10.3390/ph17101286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 07/26/2024] [Accepted: 08/01/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Cu/Zn Superoxide Dismutase 1 (SOD1) is a 32 kDa cytosolic dimeric metalloenzyme that neutralizes superoxide anions into oxygen and hydrogen peroxide. Mutations in SOD1 are associated with ALS, a disease causing motor neuron atrophy and subsequent mortality. These mutations exert their harmful effects through a gain of function mechanism, rather than a loss of function. Despite extensive research, the mechanism causing selective motor neuron death still remains unclear. A defining feature of ALS pathogenesis is protein misfolding and aggregation, evidenced by ubiquitinated protein inclusions containing SOD1 in affected motor neurons. This work aims to identify compounds countering SOD1(A4V) misfolding and aggregation, which could potentially aid in ALS treatment. METHODS The approach employed was in vitro screening of a library comprising 1280 pharmacologically active compounds (LOPAC®) in the context of drug repurposing. Using differential scanning fluorimetry (DSF), these compounds were tested for their impact on SOD1(A4V) thermal stability. RESULTS AND CONCLUSIONS Dimer stability was the parameter chosen as the criterion for screening, since the dissociation of the native SOD1 dimer is the step prior to its in vitro aggregation. The screening revealed one compound raising protein-ligand Tm by 6 °C, eleven inducing a higher second Tm, suggesting a stabilization effect, and fourteen reducing Tm from 10 up to 26 °C, suggesting possible interactions or non-specific binding.
Collapse
Affiliation(s)
- Maria Giannakou
- Biochemistry and Molecular Biology Unit, Department of Biology, National and Kapodistrian University of Athens, 15784 Zografou, Greece
- Enzyme and Microbial Biotechnology Unit, Department of Biology, National and Kapodistrian University of Athens, 15784 Zografou, Greece
| | - Ifigeneia Akrani
- Laboratory of Pharmaceutical Chemistry, Department of Pharmacy, National and Kapodistrian University of Athens, 15784 Zografou, Greece; (I.A.)
| | - Angeliki Tsoka
- Biochemistry and Molecular Biology Unit, Department of Biology, National and Kapodistrian University of Athens, 15784 Zografou, Greece
| | - Vassilios Myrianthopoulos
- Laboratory of Pharmaceutical Chemistry, Department of Pharmacy, National and Kapodistrian University of Athens, 15784 Zografou, Greece; (I.A.)
| | - Emmanuel Mikros
- Laboratory of Pharmaceutical Chemistry, Department of Pharmacy, National and Kapodistrian University of Athens, 15784 Zografou, Greece; (I.A.)
| | - Constantinos Vorgias
- Biochemistry and Molecular Biology Unit, Department of Biology, National and Kapodistrian University of Athens, 15784 Zografou, Greece
| | - Dimitris G. Hatzinikolaou
- Enzyme and Microbial Biotechnology Unit, Department of Biology, National and Kapodistrian University of Athens, 15784 Zografou, Greece
| |
Collapse
|
4
|
Sugiyama T, Nishitoh H. Neurodegenerative diseases associated with the disruption of proteostasis and their therapeutic strategies using chemical chaperones. J Biochem 2024; 176:179-186. [PMID: 38955196 DOI: 10.1093/jb/mvae048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/29/2024] [Accepted: 06/29/2024] [Indexed: 07/04/2024] Open
Abstract
Aberrant proteostasis is thought to be involved in the pathogenesis of neurodegenerative diseases. Some proteostasis abnormalities are ameliorated by chaperones. Chaperones are divided into three groups: molecular, pharmacological and chemical. Chemical chaperones intended to alleviate stress in organelles, such as the endoplasmic reticulum (ER), are now being administered clinically. Of the chemical chaperones, 4-phenylbutyrate (4-PBA) has been used as a research reagent, and its mechanism of action includes chaperone effects and the inhibition of histone deacetylase. Moreover, it also binds to the B-site of SEC24 and regulates COPII-mediated transport from the ER. Although its therapeutic effect may not be strong, elucidating the mechanism of action of 4-PBA may contribute to the identification of novel therapeutic targets for neurodegenerative diseases.
Collapse
Affiliation(s)
- Takashi Sugiyama
- Laboratory of Biochemistry and Molecular Biology, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan
- Department of Neurology, Faculty of Medicine, University of Miyazaki Hospital, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan
- Division of Respirology, Rheumatology, Infectious Diseases, and Neurology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan
| | - Hideki Nishitoh
- Laboratory of Biochemistry and Molecular Biology, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan
- Frontier Science Research Center, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan
| |
Collapse
|
5
|
Shah N, Kasture AS, Fischer FP, Sitte HH, Hummel T, Sucic S. A transporter's doom or destiny: SLC6A1 in health and disease, novel molecular targets and emerging therapeutic prospects. Front Mol Neurosci 2024; 17:1466694. [PMID: 39268250 PMCID: PMC11390516 DOI: 10.3389/fnmol.2024.1466694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 08/19/2024] [Indexed: 09/15/2024] Open
Abstract
As the first member of the solute carrier 6 (SLC6) protein family, the γ-aminobutyric acid (GABA) transporter 1 (GAT1, SLC6A1), plays a pivotal role in the uptake of GABA from the synaptic cleft into neurons and astrocytes. This process facilitates the subsequent storage of GABA in presynaptic vesicles. The human SLC6A1 gene is highly susceptible to missense mutations, leading to severe clinical outcomes, such as epilepsy, in the afflicted patients. The molecular mechanisms of SLC6A1-associated disorders are discerned to some degree; many SLC6A1 mutations are now known to impair protein folding, and consequently fail to reach the plasma membrane. Inherently, once inside the endoplasmic reticulum (ER), GAT1 abides by a complex cascade of events that enable efficient intracellular trafficking. This involves association with specialized molecular chaperones responsible for steering the protein folding process, oligomerization, sorting through the Golgi apparatus, and ultimately delivery to the cell surface. The entire process is subject to stringent quality control mechanisms at multiple checkpoints. While the majority of the existing loss-of-function SLC6A1 variants interfere with folding and membrane targeting, certain mutants retain abundant surface expression. In either scenario, suppressed GAT1 activity disrupts GABAergic neurotransmission, preceding the disease manifestation in individuals harboring these mutations. The nervous system is enthralling and calls for systematic, groundbreaking research efforts to dissect the precise molecular factors associated with the onset of complex neurological disorders, and uncover additional non-canonical therapeutic targets. Recent research has given hope for some of the misfolded SLC6A1 variants, which can be salvaged by small molecules, i.e., chemical and pharmacological chaperones, acting on multiple upstream targets in the secretory pathway. We here highlight the significance of pharmacochaperoning as a therapeutic strategy for the treatment of SLC6A1-related disorders.
Collapse
Affiliation(s)
- Nikita Shah
- Institute of Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Ameya S Kasture
- Institute of Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Florian P Fischer
- Department of Epileptology and Neurology, RWTH Aachen University, Aachen, Germany
| | - Harald H Sitte
- Institute of Pharmacology, Medical University of Vienna, Vienna, Austria
- Hourani Center for Applied Scientific Research, Al-Ahliyya Amman University, Amman, Jordan
- Center for Addiction Research and Science-AddRess, Medical University of Vienna, Vienna, Austria
| | - Thomas Hummel
- Department of Neuroscience and Developmental Biology, University of Vienna, Vienna, Austria
| | - Sonja Sucic
- Institute of Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
6
|
Kim HS, Lee D, Shen S. Endoplasmic reticular stress as an emerging therapeutic target for chronic pain: a narrative review. Br J Anaesth 2024; 132:707-724. [PMID: 38378384 PMCID: PMC10925894 DOI: 10.1016/j.bja.2024.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 12/11/2023] [Accepted: 01/05/2024] [Indexed: 02/22/2024] Open
Abstract
Chronic pain is a severely debilitating condition with enormous socioeconomic costs. Current treatment regimens with nonsteroidal anti-inflammatory drugs (NSAIDs), steroids, or opioids have been largely unsatisfactory with uncertain benefits or severe long-term side effects. This is mainly because chronic pain has a multifactorial aetiology. Although conventional pain medications can alleviate pain by keeping several dysfunctional pathways under control, they can mask other underlying pathological causes, ultimately worsening nerve pathologies and pain outcome. Recent preclinical studies have shown that endoplasmic reticulum (ER) stress could be a central hub for triggering multiple molecular cascades involved in the development of chronic pain. Several ER stress inhibitors and unfolded protein response modulators, which have been tested in randomised clinical trials or apprpoved by the US Food and Drug Administration for other chronic diseases, significantly alleviated hyperalgesia in multiple preclinical pain models. Although the role of ER stress in neurodegenerative disorders, metabolic disorders, and cancer has been well established, research on ER stress and chronic pain is still in its infancy. Here, we critically analyse preclinical studies and explore how ER stress can mechanistically act as a central node to drive development and progression of chronic pain. We also discuss therapeutic prospects, benefits, and pitfalls of using ER stress inhibitors and unfolded protein response modulators for managing intractable chronic pain. In the future, targeting ER stress to impact multiple molecular networks might be an attractive therapeutic strategy against chronic pain refractory to steroids, NSAIDs, or opioids. This novel therapeutic strategy could provide solutions for the opioid crisis and public health challenge.
Collapse
Affiliation(s)
- Harper S Kim
- Medical Scientist Training Program, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Donghwan Lee
- Department of Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Shiqian Shen
- Department of Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
7
|
Patel S, Pangarkar A, Mahajan S, Majumdar A. Therapeutic potential of endoplasmic reticulum stress inhibitors in the treatment of diabetic peripheral neuropathy. Metab Brain Dis 2023; 38:1841-1856. [PMID: 37289403 DOI: 10.1007/s11011-023-01239-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 05/19/2023] [Indexed: 06/09/2023]
Abstract
Endoplasmic stress response, the unfolded protein response (UPR), is a homeostatic signaling pathway comprising transmembrane sensors that get activated upon alterations in ER luminal environment. Studies suggest a relation between activated UPR pathways and several disease states such as Parkinson, Alzheimer, inflammatory bowel disease, tumor growth, and metabolic syndrome. Diabetic peripheral neuropathy (DPN), a common microvascular complication of diabetes-related chronic hyperglycemia, causes chronic pain, loss of sensation, foot ulcers, amputations, allodynia, hyperalgesia, paresthesia, and spontaneous pain. Factors like disrupted calcium signaling, dyslipidemia, hyperglycemia, inflammation, insulin signaling, and oxidative stress disturb the UPR sensor levels manifesting as DPN. We discuss new effective therapeutic alternatives for DPN that can be developed by targeting UPR pathways like synthetic ER stress inhibitors like 4-PhenylButyric acid (4-PBA), Sephin 1, Salubrinal and natural ER stress inhibitors like Tauroursodeoxycholic acid (TUDCA), Cordycepin, Proanthocyanidins, Crocin, Purple Rice extract and cyanidin and Caffeic Acid Phenethyl Ester (CAPE).
Collapse
Affiliation(s)
- Shivangi Patel
- Department of Pharmacology, Bombay College of Pharmacy, Kalina, Mumbai, 400098, India
| | - Arnika Pangarkar
- Department of Pharmacology, Bombay College of Pharmacy, Kalina, Mumbai, 400098, India
| | - Sakshi Mahajan
- Department of Pharmacology, Bombay College of Pharmacy, Kalina, Mumbai, 400098, India
| | - Anuradha Majumdar
- Department of Pharmacology, Bombay College of Pharmacy, Kalina, Mumbai, 400098, India.
| |
Collapse
|
8
|
Baumanns S, Muehlemeyer F, Miesbauer LC, Baake J, Roloff EM, Beis DM, Wenzel U. 4-Phenylbutyric acid attenuates amyloid-β proteotoxicity through activation of HSF-1 in an Alzheimer's disease model of the nematode Caenorhabditiselegans. Biochem Biophys Res Commun 2023; 673:16-22. [PMID: 37354655 DOI: 10.1016/j.bbrc.2023.06.064] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 06/19/2023] [Indexed: 06/26/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder and the most common form of dementia. The pathogenesis is a complex process, in which the proteotoxicity of amyloid-β (Aβ) was identified as a major factor. 4-Phenylbutyric acid (4-PBA) is an aromatic short-chain fatty acid that may attenuate Aβ proteotoxicity through its already shown properties as a chemical chaperone or by inhibition of histone deacetylases (HDACs). In the present study, we investigated the molecular effects of 4-PBA on Aβ proteotoxicity using the nematode Caenorhabditis elegans as a model. Computer-based analysis of motility was used as a measure of Aβ proteotoxicity in the transgenic strain GMC101, expressing human Aβ1-42 in body wall muscle cells. Aβ aggregation was quantified using the fluorescent probe NIAD-4 to correlate the effects of 4-PBA on motility with the amount of the proteotoxic protein. Furthermore, these approaches were supplemented by gene regulation via RNA interference (RNAi) to identify molecular targets of 4-PBA. 4-PBA improved the motility of GMC101 nematodes and reduced Aβ aggregation significantly. Knockdown of hsf-1, encoding an ortholog essential for the cytosolic heat shock response, prevented the increase in motility and decrease in Aβ aggregation by 4-PBA incubation. RNAi for hda-1, encoding an ortholog of histone deacetylase 2, also increased motility. Double RNAi for hsf-1 and hda-1 revealed a dominant effect of hsf-1 RNAi. Moreover, 4-PBA failed to further increase motility under hda-1 RNAi. Accordingly, the results suggest that 4-PBA attenuates Aβ proteotoxicity in an AD-model of C. elegans through activation of HSF-1 via inhibition of HDA-1.
Collapse
Affiliation(s)
- Stefan Baumanns
- Molecular Nutrition Research, Interdisciplinary Research Center, Justus-Liebig-University of Giessen, Heinrich-Buff-Ring 26-32, D-35392, Giessen, Germany
| | - Felix Muehlemeyer
- Molecular Nutrition Research, Interdisciplinary Research Center, Justus-Liebig-University of Giessen, Heinrich-Buff-Ring 26-32, D-35392, Giessen, Germany
| | - Laura C Miesbauer
- Molecular Nutrition Research, Interdisciplinary Research Center, Justus-Liebig-University of Giessen, Heinrich-Buff-Ring 26-32, D-35392, Giessen, Germany
| | - Jonas Baake
- Molecular Nutrition Research, Interdisciplinary Research Center, Justus-Liebig-University of Giessen, Heinrich-Buff-Ring 26-32, D-35392, Giessen, Germany
| | - Eva M Roloff
- Molecular Nutrition Research, Interdisciplinary Research Center, Justus-Liebig-University of Giessen, Heinrich-Buff-Ring 26-32, D-35392, Giessen, Germany
| | - Daniel M Beis
- Molecular Nutrition Research, Interdisciplinary Research Center, Justus-Liebig-University of Giessen, Heinrich-Buff-Ring 26-32, D-35392, Giessen, Germany
| | - Uwe Wenzel
- Molecular Nutrition Research, Interdisciplinary Research Center, Justus-Liebig-University of Giessen, Heinrich-Buff-Ring 26-32, D-35392, Giessen, Germany.
| |
Collapse
|
9
|
Villani S, Dematteis G, Tapella L, Gagliardi M, Lim D, Corazzari M, Aprile S, Del Grosso E. Quantification of the Chemical Chaperone 4-Phenylbutyric Acid (4-PBA) in Cell Culture Media via LC-HRMS: Applications in Fields of Neurodegeneration and Cancer. Pharmaceuticals (Basel) 2023; 16:298. [PMID: 37259441 PMCID: PMC9960049 DOI: 10.3390/ph16020298] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 02/01/2023] [Accepted: 02/10/2023] [Indexed: 04/10/2024] Open
Abstract
In recent years, 4-phenylbutyric acid (4-PBA), an FDA-approved drug, has increasingly been used as a nonspecific chemical chaperone in vitro and in vitro, but its pharmacodynamics is still not clear. In this context, we developed and validated a Liquid Chromatography-High Resolution Mass Spectrometry (LC-HRMS) method to quantify 4-PBA in NeuroBasal-A and Dulbecco's Modified Eagle widely used cell culture media. Samples were injected on a Luna® 3 µm PFP(2) 100 Å (100 × 2.0 mm) column maintained at 40 °C. Water and methanol both with 0.1% formic acid served as mobile phases in a step gradient mode. The mass acquisition was performed by selected ion monitoring (SIM) in negative mode for a total run time of 10.5 min at a flow rate of 0.300 mL/min. The analogue 4-(4-Nitrophenyl)-Butyric Acid served as internal standard. Validation parameters were verified according to FDA and EMA guidelines. The quantification ranges from 0.38-24 µM. Inter and intraday RSDs (Relative Standard Deviations) were within 15%. The developed LC-HRMS method allowed the estimation of 4-PBA absorption and adsorption kinetics in vitro in two experimental systems: (i) 4-PBA improvement of protein synthesis in an Alzheimer's disease astrocytic cell model; and (ii) 4-PBA reduction of endoplasmic reticulum stress in thapsigargin-treated melanoma cell lines.
Collapse
Affiliation(s)
- Salvatore Villani
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Largo Donegani 2, 28100 Novara, Italy
| | - Giulia Dematteis
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Largo Donegani 2, 28100 Novara, Italy
| | - Laura Tapella
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Largo Donegani 2, 28100 Novara, Italy
| | - Mara Gagliardi
- Department of Health Science (DSS), Center for Translational Research on Autoimmune and Allergic Disease (CAAD) & Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale, 28100 Novara, Italy
| | - Dmitry Lim
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Largo Donegani 2, 28100 Novara, Italy
| | - Marco Corazzari
- Department of Health Science (DSS), Center for Translational Research on Autoimmune and Allergic Disease (CAAD) & Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale, 28100 Novara, Italy
| | - Silvio Aprile
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Largo Donegani 2, 28100 Novara, Italy
| | - Erika Del Grosso
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Largo Donegani 2, 28100 Novara, Italy
| |
Collapse
|
10
|
Kasture AS, Fischer FP, Kunert L, Burger ML, Burgstaller AC, El-Kasaby A, Hummel T, Sucic S. Drosophila melanogaster as a model for unraveling unique molecular features of epilepsy elicited by human GABA transporter 1 variants. Front Neurosci 2023; 16:1074427. [PMID: 36741049 PMCID: PMC9893286 DOI: 10.3389/fnins.2022.1074427] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 12/21/2022] [Indexed: 01/20/2023] Open
Abstract
Mutations in the human γ-aminobutyric acid (GABA) transporter 1 (hGAT-1) can instigate myoclonic-atonic and other generalized epilepsies in the afflicted individuals. We systematically examined fifteen hGAT-1 disease variants, all of which dramatically reduced or completely abolished GABA uptake activity. Many of these loss-of-function variants were absent from their regular site of action at the cell surface, due to protein misfolding and/or impaired trafficking machinery (as verified by confocal microscopy and de-glycosylation experiments). A modest fraction of the mutants displayed correct targeting to the plasma membrane, but nonetheless rendered the mutated proteins devoid of GABA transport, possibly due to structural alterations in the GABA binding site/translocation pathway. We here focused on a folding-deficient A288V variant. In flies, A288V reiterated its impeded expression pattern, closely mimicking the ER-retention demonstrated in transfected HEK293 cells. Functionally, A288V presented a temperature-sensitive seizure phenotype in fruit flies. We employed diverse small molecules to restore the expression and activity of folding-deficient hGAT-1 epilepsy variants, in vitro (in HEK293 cells) and in vivo (in flies). We identified three compounds (chemical and pharmacological chaperones) conferring moderate rescue capacity for several variants. Our data grant crucial new insights into: (i) the molecular basis of epilepsy in patients harboring hGAT-1 mutations, and (ii) a proof-of-principle that protein folding deficits in disease-associated hGAT-1 variants can be corrected using the pharmacochaperoning approach. Such innovative pharmaco-therapeutic prospects inspire the rational design of novel drugs for alleviating the clinical symptoms triggered by the numerous emerging pathogenic mutations in hGAT-1.
Collapse
Affiliation(s)
- Ameya S. Kasture
- Institute of Pharmacology, Medical University of Vienna, Vienna, Austria,Department of Neuroscience and Developmental Biology, University of Vienna, Vienna, Austria
| | - Florian P. Fischer
- Institute of Pharmacology, Medical University of Vienna, Vienna, Austria,Department of Epileptology and Neurology, University of Aachen, Aachen, Germany
| | - Lisa Kunert
- Department of Neuroscience and Developmental Biology, University of Vienna, Vienna, Austria
| | - Melanie L. Burger
- Institute of Pharmacology, Medical University of Vienna, Vienna, Austria
| | | | - Ali El-Kasaby
- Institute of Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Thomas Hummel
- Department of Neuroscience and Developmental Biology, University of Vienna, Vienna, Austria
| | - Sonja Sucic
- Institute of Pharmacology, Medical University of Vienna, Vienna, Austria,*Correspondence: Sonja Sucic,
| |
Collapse
|
11
|
Effect of Dietary 4-Phenylbuthyric Acid Supplementation on Acute Heat-Stress-Induced Hyperthermia in Broiler Chickens. Animals (Basel) 2022; 12:ani12162056. [PMID: 36009646 PMCID: PMC9404993 DOI: 10.3390/ani12162056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/08/2022] [Accepted: 08/10/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Heat stress (HS) induces endoplasmic reticulum (ER) stress and disrupts the ER and cellular homeostasis. A recent study showed that ER stress was induced in broiler chickens under severe and acute HS; however, it was unclear how the alleviation of ER stress affects the physiological state of broiler chickens. Therefore, this study aimed to investigate the ameliorative effects of an ER stress alleviator, 4-phenylbutyric acid (4-PBA), which is a chemical chaperone that reduces ER stress, on the body temperature response, energy metabolic state, and cellular ER stress in HS-exposed birds. 4-PBA supplementation did not negatively affect the growth rate. In addition, 4-PBA suppressed the HS-induced ER stress response in skeletal muscle. Surprisingly, 4-PBA significantly decreased body temperature elevation in HS birds. The present study showed that the ER stress, alleviated by 4-PBA, might contribute to the induction of heat tolerance in broiler chickens. Abstract Hot, humid weather causes heat stress (HS) in broiler chickens, which can lead to high mortality. A recent study found that HS causes endoplasmic reticulum (ER) stress. However, the possible involvement of ER stress in HS-induced physiological alterations in broiler chickens is unclear. This study aimed to evaluate the effect of the dietary supplementation of 4-phenylbutyric acid (4-PBA), an alleviator of ER stress, in acute HS-exposed young broiler chickens. Twenty-eight 14-day-old male broiler chickens (ROSS 308) were divided into two groups and fed either a control diet or a diet containing 4-PBA (5.25 g per kg of diet feed) for 10 days. At 24 days old, each group of chickens was kept in thermoneutral (24 ± 0.5 °C) or acute HS (36 ± 0.5 °C) conditions for 2 h. The results showed that thermoneutral birds supplemented with 4-PBA exhibited no negative effects in terms of broiler body weight gain and tissue weight compared to non-supplemental birds. HS increased body temperature in both the control and 4-PBA groups, but the elevation was significantly lower in the 4-PBA group than in the control group. The plasma non-esterified fatty acid concentration was significantly increased by HS treatment in non-supplemental groups, while the increase was partially attenuated in the 4-PBA group. Moreover, 4-PBA prevented HS-induced gene elevation of the ER stress markers GRP78 and GRP94 in the skeletal muscle. These findings suggest that the 4-PBA effect may be specific to the skeletal muscle in HS-exposed birds and that 4-PBA supplementation attenuated HS-induced muscle ER stress, which could be associated with a supplementation of the body temperature elevation and lipolysis.
Collapse
|
12
|
Gomez-Navarro N, Maldutyte J, Poljak K, Peak-Chew SY, Orme J, Bisnett BJ, Lamb CH, Boyce M, Gianni D, Miller EA. Selective inhibition of protein secretion by abrogating receptor-coat interactions during ER export. Proc Natl Acad Sci U S A 2022; 119:e2202080119. [PMID: 35901214 PMCID: PMC9351455 DOI: 10.1073/pnas.2202080119] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 06/01/2022] [Indexed: 01/03/2023] Open
Abstract
Protein secretion is an essential process that drives cell growth, movement, and communication. Protein traffic within the secretory pathway occurs via transport intermediates that bud from one compartment and fuse with a downstream compartment to deliver their contents. Here, we explore the possibility that protein secretion can be selectively inhibited by perturbing protein-protein interactions that drive capture into transport vesicles. Human proprotein convertase subtilisin/kexin type 9 (PCSK9) is a determinant of cholesterol metabolism whose secretion is mediated by a specific cargo adaptor protein, SEC24A. We map a series of protein-protein interactions between PCSK9, its endoplasmic reticulum (ER) export receptor SURF4, and SEC24A that mediate secretion of PCSK9. We show that the interaction between SURF4 and SEC24A can be inhibited by 4-phenylbutyrate (4-PBA), a small molecule that occludes a cargo-binding domain of SEC24. This inhibition reduces secretion of PCSK9 and additional SURF4 clients that we identify by mass spectrometry, leaving other secreted cargoes unaffected. We propose that selective small-molecule inhibition of cargo recognition by SEC24 is a potential therapeutic intervention for atherosclerosis and other diseases that are modulated by secreted proteins.
Collapse
Affiliation(s)
- Natalia Gomez-Navarro
- Cell Biology Division, Medical Research Council Laboratory of Molecular Biology, Cambridge, CB2 0QH, United Kingdom
| | - Julija Maldutyte
- Cell Biology Division, Medical Research Council Laboratory of Molecular Biology, Cambridge, CB2 0QH, United Kingdom
| | - Kristina Poljak
- Cell Biology Division, Medical Research Council Laboratory of Molecular Biology, Cambridge, CB2 0QH, United Kingdom
| | - Sew-Yeu Peak-Chew
- Cell Biology Division, Medical Research Council Laboratory of Molecular Biology, Cambridge, CB2 0QH, United Kingdom
| | - Jonathon Orme
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, CB2 0AA, United Kingdom
| | - Brittany J. Bisnett
- Department of Biochemistry, Duke University School of Medicine, Durham, NC27710, USA
| | - Caitlin H. Lamb
- Department of Biochemistry, Duke University School of Medicine, Durham, NC27710, USA
| | - Michael Boyce
- Department of Biochemistry, Duke University School of Medicine, Durham, NC27710, USA
| | - Davide Gianni
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, CB2 0AA, United Kingdom
| | - Elizabeth A. Miller
- Cell Biology Division, Medical Research Council Laboratory of Molecular Biology, Cambridge, CB2 0QH, United Kingdom
| |
Collapse
|
13
|
Miglioranza Scavuzzi B, van Drongelen V, Kaur B, Fox JC, Liu J, Mesquita-Ferrari RA, Kahlenberg JM, Farkash EA, Benavides F, Miller FW, Sawalha AH, Holoshitz J. The lupus susceptibility allele DRB1*03:01 encodes a disease-driving epitope. Commun Biol 2022; 5:751. [PMID: 35902632 PMCID: PMC9334592 DOI: 10.1038/s42003-022-03717-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 07/14/2022] [Indexed: 12/14/2022] Open
Abstract
The HLA-DRB1*03:01 allele is a major genetic risk factor in systemic lupus erythematosus (SLE), but the mechanistic basis of the association is unclear. Here we show that in the presence of interferon gamma (IFN-γ), a short DRB1*03:01-encoded allelic epitope activates a characteristic lupus transcriptome in mouse and human macrophages. It also triggers a cascade of SLE-associated cellular aberrations, including endoplasmic reticulum stress, unfolded protein response, mitochondrial dysfunction, necroptotic cell death, and production of pro-inflammatory cytokines. Parenteral administration of IFN-γ to naïve DRB1*03:01 transgenic mice causes increased serum levels of anti-double stranded DNA antibodies, glomerular immune complex deposition and histopathological renal changes that resemble human lupus nephritis. This study provides evidence for a noncanonical, antigen presentation-independent mechanism of HLA-disease association in SLE and could lay new foundations for our understanding of key molecular mechanisms that trigger and propagate this devastating autoimmune disease.
Collapse
Affiliation(s)
| | | | - Bhavneet Kaur
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | | | - Jianhua Liu
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | | | | | - Evan A Farkash
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Fernando Benavides
- Department of Epigenetics and Molecular Carcinogenesis, MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Frederick W Miller
- Environmental Autoimmunity Group, National Institute of Environmental Health Sciences, Research Triangle Park, NC, 27709, USA
| | - Amr H Sawalha
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
- Departments of Pediatrics and Internal Medicine, University of Pittsburgh, Pittsburgh, PA, 15224, USA
| | - Joseph Holoshitz
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
14
|
Suga K, Yamamoto-Hijikata S, Terao Y, Akagawa K, Ushimaru M. Golgi stress induces upregulation of the ER-Golgi SNARE Syntaxin-5, altered βAPP processing, and Caspase-3-dependent apoptosis in NG108-15 cells. Mol Cell Neurosci 2022; 121:103754. [PMID: 35842170 DOI: 10.1016/j.mcn.2022.103754] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 05/29/2022] [Accepted: 07/07/2022] [Indexed: 01/06/2023] Open
Abstract
The involvement of secretory pathways and Golgi dysfunction in neuronal cells during Alzheimer's disease progression is poorly understood. Our previous overexpression and knockdown studies revealed that the intracellular protein level of Syntaxin-5, an endoplasmic reticulum-Golgi soluble N-ethylmaleimide-sensitive factor-attachment protein receptor (SNARE), modulates beta-amyloid precursor protein processing in neuronal cells. We recently showed that changes in endogenous Syntaxin-5 protein expression occur under stress induction. Syntaxin-5 was upregulated by endoplasmic reticulum stress but was degraded by Caspase-3 during apoptosis in neuronal cells. In addition, we showed that sustained endoplasmic reticulum stress promotes Caspase-3-dependent apoptosis during the later phase of the endoplasmic reticulum stress response in NG108-15 cells. In this study, to elucidate the consequences of secretory pathway dysfunction in beta-amyloid precursor protein processing that lead to neuronal cell death, we examined the effect of various stresses on endoplasmic reticulum-Golgi SNARE expression and beta-amyloid precursor protein processing. By using compounds to disrupt Golgi function, we show that Golgi stress promotes upregulation of the endoplasmic reticulum-Golgi SNARE Syntaxin-5, and prolonged stress causes Caspase-3-dependent apoptosis. Golgi stress induced intracellular beta-amyloid precursor protein accumulation and a concomitant decrease in total amyloid-beta production. We also examined the protective effect of the chemical chaperone 4-phenylbutylate on changes in amyloid-beta production and the activation of Caspase-3 induced by endoplasmic reticulum and Golgi stress. The compound alleviated the increase in the amyloid-beta 1-42/amyloid-beta 1-40 ratio induced by endoplasmic reticulum and Golgi stress. Furthermore, 4-phenylbutylate could rescue Caspase-3-dependent apoptosis induced by prolonged organelle stress. These results suggest that organelle stress originating from the endoplasmic reticulum and Golgi has a substantial impact on the amyloidogenic processing of beta-amyloid precursor protein and Caspase-3-dependent apoptosis, leading to neuronal cell death.
Collapse
Affiliation(s)
- Kei Suga
- Department of Chemistry, Kyorin University, Faculty of Medicine, Mitaka, Tokyo 181-8611, Japan; Department of Medical Physiology, Kyorin University, Faculty of Medicine, Mitaka, Tokyo 181-8611, Japan.
| | | | - Yasuo Terao
- Department of Medical Physiology, Kyorin University, Faculty of Medicine, Mitaka, Tokyo 181-8611, Japan
| | - Kimio Akagawa
- Department of Medical Physiology, Kyorin University, Faculty of Medicine, Mitaka, Tokyo 181-8611, Japan
| | - Makoto Ushimaru
- Department of Chemistry, Kyorin University, Faculty of Medicine, Mitaka, Tokyo 181-8611, Japan
| |
Collapse
|
15
|
Vats A, Xi Y, Feng B, Clinger OD, St Leger AJ, Liu X, Ghosh A, Dermond CD, Lathrop KL, Tochtrop GP, Picaud S, Chen Y. Non-retinoid chaperones improve rhodopsin homeostasis in a mouse model of retinitis pigmentosa. JCI Insight 2022; 7:153717. [PMID: 35472194 PMCID: PMC9220944 DOI: 10.1172/jci.insight.153717] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 04/20/2022] [Indexed: 11/17/2022] Open
Abstract
Rhodopsin-associated (RHO-associated) retinitis pigmentosa (RP) is a progressive retinal disease that currently has no cure. RHO protein misfolding leads to disturbed proteostasis and the death of rod photoreceptors, resulting in decreased vision. We previously identified nonretinoid chaperones of RHO, including YC-001 and F5257-0462, by small-molecule high-throughput screening. Here, we profile the chaperone activities of these molecules toward the cell-surface level of 27 RP-causing human RHO mutants in NIH3T3 cells. Furthermore, using retinal explant culture, we show that YC-001 improves retinal proteostasis by supporting RHO homeostasis in RhoP23H/+ mouse retinae, which results in thicker outer nuclear layers (ONL), indicating delayed photoreceptor degeneration. Interestingly, YC-001 ameliorated retinal immune responses and reduced the number of microglia/macrophages in the RhoP23H/+ retinal explants. Similarly, F5257-0462 also protects photoreceptors in RhoP23H/+ retinal explants. In vivo, intravitreal injection of YC-001 or F5257-0462 microparticles in PBS shows that F5257-0462 has a higher efficacy in preserving photoreceptor function and delaying photoreceptor death in RhoP23H/+ mice. Collectively, we provide proof of principle that nonretinoid chaperones are promising drug candidates in treating RHO-associated RP.
Collapse
Affiliation(s)
- Abhishek Vats
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, United States of America
| | - Yibo Xi
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, United States of America
| | - Bing Feng
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, United States of America
| | - Owen D Clinger
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, United States of America
| | - Anthony J St Leger
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, United States of America
| | - Xujie Liu
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, United States of America
| | - Archisha Ghosh
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, United States of America
| | - Chase D Dermond
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, United States of America
| | - Kira L Lathrop
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, United States of America
| | - Gregory P Tochtrop
- Department of Chemistry, Case Western Reserve University, Cleveland, United States of America
| | - Serge Picaud
- Institut de la Vision, Sorbonne Université, Paris, France
| | - Yuanyuan Chen
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, United States of America
| |
Collapse
|
16
|
Nagata T, Shinagawa S, Nakajima S, Noda Y, Mimura M. Pharmacotherapeutic combinations for the treatment of Alzheimer's disease. Expert Opin Pharmacother 2022; 23:727-737. [PMID: 35230200 DOI: 10.1080/14656566.2022.2042514] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Alzheimer's disease (AD) is the most common form of dementia, and four medications are currently available as symptomatic therapies: three cholinesterase inhibitors (ChEI) and memantine. In June 2021, aducanumab was approved in the United States under an accelerated approval pathway as the first novel putative disease-modifying therapy (p-DMT) targeting the β-amyloid (Aβ) cascade in the brain. The combination of several monotherapies to address the multifactorial pathogenesis of neurodegenerative diseases is an anticipated next step. AREAS COVERED The cholinergic hypothesis and the amyloid cascade hypothesis have been proposed as explanations for the pathogenesis of AD. Given the limited effectiveness of monotherapies based on these hypotheses, approaches using combination therapy are attempting to address the complexity of AD pathogenesis, including putative causative proteins-related neurodegeneration, neurotransmitters, and neuroinflammation, in a comprehensive manner. EXPERT OPINION The efficacy of an initial or add-on combination approach to counteracting neurodegenerative processes and functional deterioration has been investigated. The combination of symptomatic therapies with approved anti-dementia medicines (one ChEI and memantine) has been found to be functionally effective for a moderately severe disease stage. Furthermore, combination strategies involving p-DMTs, symptomatic therapies, and neuro-regeneration may be useful in the future.
Collapse
Affiliation(s)
- Tomoyuki Nagata
- Department of Psychiatry, The Jikei University School of Medicine, Tokyo, Japan.,Department of Psychiatry, Airanomori Hospital, Kagoshima, Japan
| | | | - Shinichiro Nakajima
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Yoshihiro Noda
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Masaru Mimura
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
17
|
Zhang F, Zeng QY, Xu H, Xu AN, Liu DJ, Li NZ, Chen Y, Jin Y, Xu CH, Feng CZ, Zhang YL, Liu D, Liu N, Xie YY, Yu SH, Yuan H, Xue K, Shi JY, Liu TX, Xu PF, Zhao WL, Zhou Y, Wang L, Huang QH, Chen Z, Chen SJ, Zhou XL, Sun XJ. Selective and competitive functions of the AAR and UPR pathways in stress-induced angiogenesis. Cell Discov 2021; 7:98. [PMID: 34697290 PMCID: PMC8547220 DOI: 10.1038/s41421-021-00332-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 08/31/2021] [Indexed: 12/30/2022] Open
Abstract
The amino acid response (AAR) and unfolded protein response (UPR) pathways converge on eIF2α phosphorylation, which is catalyzed by Gcn2 and Perk, respectively, under different stresses. This close interconnection makes it difficult to specify different functions of AAR and UPR. Here, we generated a zebrafish model in which loss of threonyl-tRNA synthetase (Tars) induces angiogenesis dependent on Tars aminoacylation activity. Comparative transcriptome analysis of the tars-mutant and wild-type embryos with/without Gcn2- or Perk-inhibition reveals that only Gcn2-mediated AAR is activated in the tars-mutants, whereas Perk functions predominantly in normal development. Mechanistic analysis shows that, while a considerable amount of eIF2α is normally phosphorylated by Perk, the loss of Tars causes an accumulation of uncharged tRNAThr, which in turn activates Gcn2, leading to phosphorylation of an extra amount of eIF2α. The partial switchover of kinases for eIF2α largely overwhelms the functions of Perk in normal development. Interestingly, although inhibition of Gcn2 and Perk in this stress condition both can reduce the eIF2α phosphorylation levels, their functional consequences in the regulation of target genes and in the rescue of the angiogenic phenotypes are dramatically different. Indeed, genetic and pharmacological manipulations of these pathways validate that the Gcn2-mediated AAR, but not the Perk-mediated UPR, is required for tars-deficiency induced angiogenesis. Thus, the interconnected AAR and UPR pathways differentially regulate angiogenesis through selective functions and mutual competitions, reflecting the specificity and efficiency of multiple stress response pathways that evolve integrally to enable an organism to sense/respond precisely to various types of stresses.
Collapse
Affiliation(s)
- Fan Zhang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qi-Yu Zeng
- State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Hao Xu
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ai-Ning Xu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dian-Jia Liu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.,School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Ning-Zhe Li
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.,School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Yi Chen
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi Jin
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chun-Hui Xu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Chang-Zhou Feng
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuan-Liang Zhang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dan Liu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Key Laboratory of Systems Biomedicine, Ministry of Education, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Na Liu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.,CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yin-Yin Xie
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shan-He Yu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hao Yuan
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kai Xue
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing-Yi Shi
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ting Xi Liu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.,CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Peng-Fei Xu
- Division of Human Reproduction and Developmental Genetics, Women's Hospital, and Institute of Genetics and Department of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Wei-Li Zhao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi Zhou
- Stem Cell Program, Hematology/Oncology Program at Children's Hospital Boston and Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Lan Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qiu-Hua Huang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhu Chen
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Sai-Juan Chen
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Xiao-Long Zhou
- State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China.
| | - Xiao-Jian Sun
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
18
|
Persaud AK, Nair S, Rahman MF, Raj R, Weadick B, Nayak D, McElroy C, Shanmugam M, Knoblaugh S, Cheng X, Govindarajan R. Facilitative lysosomal transport of bile acids alleviates ER stress in mouse hematopoietic precursors. Nat Commun 2021; 12:1248. [PMID: 33623001 PMCID: PMC7902824 DOI: 10.1038/s41467-021-21451-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 01/27/2021] [Indexed: 12/27/2022] Open
Abstract
Mutations in human equilibrative nucleoside transporter 3 (ENT3) encoded by SLC29A3 results in anemia and erythroid hypoplasia, suggesting that ENT3 may regulate erythropoiesis. Here, we demonstrate that lysosomal ENT3 transport of taurine-conjugated bile acids (TBA) facilitates TBA chemical chaperone function and alleviates endoplasmic reticulum (ER) stress in expanding mouse hematopoietic stem and progenitor cells (HSPCs). Slc29a3−/− HSPCs accumulate less TBA despite elevated levels of TBA in Slc29a3−/− mouse plasma and have elevated basal ER stress, reactive oxygen species (ROS), and radiation-induced apoptosis. Reintroduction of ENT3 allows for increased accumulation of TBA into HSPCs, which results in TBA-mediated alleviation of ER stress and erythroid apoptosis. Transplanting TBA-preconditioned HSPCs expressing ENT3 into Slc29a3−/− mice increase bone marrow repopulation capacity and erythroid pool size and prevent early mortalities. Together, these findings suggest a putative role for a facilitative lysosomal transporter in the bile acid regulation of ER stress in mouse HSPCs which may have implications in erythroid biology, the treatment of anemia observed in ENT3-mutated human genetic disorders, and nucleoside analog drug therapy. Mutations in ENT3, encoded by SLC29A3, result in anaemia and erythroid hypoplasia, suggesting roles in erythropoiesis. Here the authors show that ENT3 acts as a lysosomal bile acid transporter, and mutation compromises taurine conjugated bile acid transport in erythroid progenitors leading to ER stress, and anaemia.
Collapse
Affiliation(s)
- Avinash K Persaud
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, Ohio State University, Columbus, OH, 43210, USA
| | - Sreenath Nair
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, Ohio State University, Columbus, OH, 43210, USA
| | - Md Fazlur Rahman
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, Ohio State University, Columbus, OH, 43210, USA
| | - Radhika Raj
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, Ohio State University, Columbus, OH, 43210, USA
| | - Brenna Weadick
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, Ohio State University, Columbus, OH, 43210, USA
| | - Debasis Nayak
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, Ohio State University, Columbus, OH, 43210, USA
| | - Craig McElroy
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, Ohio State University, Columbus, OH, 43210, USA
| | - Muruganandan Shanmugam
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, Ohio State University, Columbus, OH, 43210, USA
| | - Sue Knoblaugh
- Depatment of Veterinary Biosciences, College of Veterinary Medicine, Ohio State University, Columbus, OH, 43210, USA
| | - Xiaolin Cheng
- Division of Medicinal Chemistry & Pharmacognosy, College of Pharmacy, Ohio State University, Columbus, OH, 43210, USA
| | - Rajgopal Govindarajan
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, Ohio State University, Columbus, OH, 43210, USA. .,Translational Therapeutics, Ohio State University Comprehensive Cancer Center, Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
19
|
Imbimbo BP, Lozupone M, Watling M, Panza F. Discontinued disease-modifying therapies for Alzheimer's disease: status and future perspectives. Expert Opin Investig Drugs 2020; 29:919-933. [PMID: 32657175 DOI: 10.1080/13543784.2020.1795127] [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: 12/17/2022]
Abstract
INTRODUCTION Alzheimer's disease (AD) is the main cause of dementia and represents a huge burden for patients, carers, and healthcare systems. Extensive efforts for over 20 years have failed to find effective disease-modifying drugs. Although amyloid-β (Aβ) accumulation in the brain predicts cognitive decline, effective reduction of plaque load by numerous drug candidates has not yielded significant clinical benefits. A similar pattern is now emerging for drugs which target hyperphosphorylated tau, and trials with anti-inflammatory drugs have been negative despite neuroinflammation appearing to have a crucial role in AD pathogenesis. AREAS COVERED This article reviews key drugs that have been discontinued while in development for AD and delineates the future landscape for present and alternative approaches. EXPERT OPINION Anti-Aβ drugs have failed to validate the Aβ cascade hypothesis of AD. Early findings suggest that the same is happening with therapeutics targeting tau and focussing future research solely on anti-tau drugs is inappropriate. Alternative targets should be pursued, including apolipoprotein E, immunomodulation, plasma exchange, protein autophagy and clearance, mitochondrial dysfunction, abnormal glucose metabolism, neurovascular unit support, epigenetic dysregulation, synaptic loss and dysfunction, microbiota dysbiosis, and combination therapies. Meanwhile, repurposing of drugs approved for other indications is justified where scientific rationale and robust preclinical evidence exist.
Collapse
Affiliation(s)
- Bruno P Imbimbo
- Department of Research and Development, Chiesi Farmaceutici , Parma, Italy
| | - Madia Lozupone
- Unit of Epidemiological Research on Aging "Greatage Study", National Institute of Gastroenterology and Research Hospital IRCCS "S. de Bellis" , Bari, Italy.,Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, and Sense Organs, University of Bari Aldo Moro , Bari, Italy
| | - Mark Watling
- CNS & Pain Department, TranScrip Partners , Reading, UK
| | - Francesco Panza
- Unit of Epidemiological Research on Aging "Greatage Study", National Institute of Gastroenterology and Research Hospital IRCCS "S. de Bellis" , Bari, Italy
| |
Collapse
|
20
|
Ho N, Yap WS, Xu J, Wu H, Koh JH, Goh WWB, George B, Chong SC, Taubert S, Thibault G. Stress sensor Ire1 deploys a divergent transcriptional program in response to lipid bilayer stress. J Cell Biol 2020; 219:e201909165. [PMID: 32349127 PMCID: PMC7337508 DOI: 10.1083/jcb.201909165] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 02/26/2020] [Accepted: 04/07/2020] [Indexed: 12/11/2022] Open
Abstract
Membrane integrity at the endoplasmic reticulum (ER) is tightly regulated, and its disturbance is implicated in metabolic diseases. Using an engineered sensor that activates the unfolded protein response (UPR) exclusively when normal ER membrane lipid composition is compromised, we identified pathways beyond lipid metabolism that are necessary to maintain ER integrity in yeast and in C. elegans. To systematically validate yeast mutants that disrupt ER membrane homeostasis, we identified a lipid bilayer stress (LBS) sensor in the UPR transducer protein Ire1, located at the interface of the amphipathic and transmembrane helices. Furthermore, transcriptome and chromatin immunoprecipitation analyses pinpoint the UPR as a broad-spectrum compensatory response wherein LBS and proteotoxic stress deploy divergent transcriptional UPR programs. Together, these findings reveal the UPR program as the sum of two independent stress responses, an insight that could be exploited for future therapeutic intervention.
Collapse
Affiliation(s)
- Nurulain Ho
- Lipid Regulation and Cell Stress Group, School of Biological Sciences, Nanyang Technological University, Singapore
| | - Wei Sheng Yap
- Lipid Regulation and Cell Stress Group, School of Biological Sciences, Nanyang Technological University, Singapore
| | - Jiaming Xu
- Centre for Molecular Medicine and Therapeutics, British Columbia Children’s Hospital Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, Canada
| | - Haoxi Wu
- Lipid Regulation and Cell Stress Group, School of Biological Sciences, Nanyang Technological University, Singapore
| | - Jhee Hong Koh
- Lipid Regulation and Cell Stress Group, School of Biological Sciences, Nanyang Technological University, Singapore
| | - Wilson Wen Bin Goh
- Bio-Data Science and Education Research Group, School of Biological Sciences, Nanyang Technological University, Singapore
| | - Bhawana George
- Lipid Regulation and Cell Stress Group, School of Biological Sciences, Nanyang Technological University, Singapore
| | - Shu Chen Chong
- Lipid Regulation and Cell Stress Group, School of Biological Sciences, Nanyang Technological University, Singapore
| | - Stefan Taubert
- Centre for Molecular Medicine and Therapeutics, British Columbia Children’s Hospital Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, Canada
| | - Guillaume Thibault
- Lipid Regulation and Cell Stress Group, School of Biological Sciences, Nanyang Technological University, Singapore
- Institute of Molecular and Cell Biology, A*STAR, Singapore
| |
Collapse
|
21
|
Arrieta A, Blackwood EA, Stauffer WT, Santo Domingo M, Bilal AS, Thuerauf DJ, Pentoney AN, Aivati C, Sarakki AV, Doroudgar S, Glembotski CC. Mesencephalic astrocyte-derived neurotrophic factor is an ER-resident chaperone that protects against reductive stress in the heart. J Biol Chem 2020; 295:7566-7583. [PMID: 32327487 DOI: 10.1074/jbc.ra120.013345] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 04/17/2020] [Indexed: 12/13/2022] Open
Abstract
We have previously demonstrated that ischemia/reperfusion (I/R) impairs endoplasmic reticulum (ER)-based protein folding in the heart and thereby activates an unfolded protein response sensor and effector, activated transcription factor 6α (ATF6). ATF6 then induces mesencephalic astrocyte-derived neurotrophic factor (MANF), an ER-resident protein with no known structural homologs and unclear ER function. To determine MANF's function in the heart in vivo, here we developed a cardiomyocyte-specific MANF-knockdown mouse model. MANF knockdown increased cardiac damage after I/R, which was reversed by AAV9-mediated ectopic MANF expression. Mechanistically, MANF knockdown in cultured neonatal rat ventricular myocytes (NRVMs) impaired protein folding in the ER and cardiomyocyte viability during simulated I/R. However, this was not due to MANF-mediated protection from reactive oxygen species generated during reperfusion. Because I/R impairs oxygen-dependent ER protein disulfide formation and such impairment can be caused by reductive stress in the ER, we examined the effects of the reductive ER stressor DTT. MANF knockdown in NRVMs increased cell death from DTT-mediated reductive ER stress, but not from nonreductive ER stresses caused by thapsigargin-mediated ER Ca2+ depletion or tunicamycin-mediated inhibition of ER protein glycosylation. In vitro, recombinant MANF exhibited chaperone activity that depended on its conserved cysteine residues. Moreover, in cells, MANF bound to a model ER protein exhibiting improper disulfide bond formation during reductive ER stress but did not bind to this protein during nonreductive ER stress. We conclude that MANF is an ER chaperone that enhances protein folding and myocyte viability during reductive ER stress.
Collapse
Affiliation(s)
- Adrian Arrieta
- San Diego State University Heart Institute and the Department of Biology, San Diego State University, San Diego, California, USA
| | - Erik A Blackwood
- San Diego State University Heart Institute and the Department of Biology, San Diego State University, San Diego, California, USA
| | - Winston T Stauffer
- San Diego State University Heart Institute and the Department of Biology, San Diego State University, San Diego, California, USA
| | - Michelle Santo Domingo
- San Diego State University Heart Institute and the Department of Biology, San Diego State University, San Diego, California, USA
| | - Alina S Bilal
- San Diego State University Heart Institute and the Department of Biology, San Diego State University, San Diego, California, USA
| | - Donna J Thuerauf
- San Diego State University Heart Institute and the Department of Biology, San Diego State University, San Diego, California, USA
| | - Amber N Pentoney
- San Diego State University Heart Institute and the Department of Biology, San Diego State University, San Diego, California, USA
| | - Cathrine Aivati
- San Diego State University Heart Institute and the Department of Biology, San Diego State University, San Diego, California, USA
| | - Anup V Sarakki
- San Diego State University Heart Institute and the Department of Biology, San Diego State University, San Diego, California, USA
| | - Shirin Doroudgar
- San Diego State University Heart Institute and the Department of Biology, San Diego State University, San Diego, California, USA.,Department of Cardiology, Angiology, and Pneumology, University Hospital Heidelberg, Innere Medizin III, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Christopher C Glembotski
- San Diego State University Heart Institute and the Department of Biology, San Diego State University, San Diego, California, USA
| |
Collapse
|
22
|
Kim JS, Hwang SI, Ryu JL, Hong HS, Lee JM, Lee SM, Jin X, Han C, Kim JH, Han J, Lee MR, Woo DH. ER stress reliever enhances functionalities of in vitro cultured hepatocytes. Stem Cell Res 2020; 43:101732. [DOI: 10.1016/j.scr.2020.101732] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 02/04/2020] [Accepted: 02/06/2020] [Indexed: 10/25/2022] Open
|
23
|
Masroor A, Zaidi N, Chandel TI, Aqueel Z, Malik S, Khan RH. Probing the Nongeneralized Amyloid Inhibitory Mechanism of Hydrophobic Chaperone. ACS Chem Neurosci 2020; 11:373-384. [PMID: 31935057 DOI: 10.1021/acschemneuro.9b00593] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Increasing prevalence of protein misfolding disorders urges the search for effective therapies. Although several antiaggregation molecules have been identified, their molecular process of aggregation and clinical trials are underway. The present study is focused on the mechanism through which phenyl butyrate (PB), a chemical chaperone, triggers inhibition of human serum albumin (HSA) fibrillation. Turbidity and Rayleigh light scattering (RLS) measurements reveal the marked presence of aggregates in HSA that were confirmed as amyloid fibrils by thioflavin T (ThT) and Congo red (CR) and were subsequently inhibited by PB in a dose dependent manner. ThT fluorescence kinetics reveals a decrease in the apparent rate constant, Kapp, in the presence of PB without triggering a lag phase in HSA suggesting PB's interference with the elongation phase. Dynamic light scattering (DLS) results display a reduction in the aggregate size in the presence of PB. Isothermal titration calorimetry (ITC) data reveals strong binding of PB at site II both at 25 °C (Kb ≈ 1.94 × 105 M-1) and 65 °C (Kb ≈ 2.90 × 104 M-1), mediated by hydrogen bonding. Overall, our finding establishes that PB stabilizes partially unfolded HSA molecules through hydrogen bonding, thereby preventing establishment of hydrogen bonds between them and hindering their progression into amyloid fibrils. This is in contrast to its chaperone effect manifested with other proteins.
Collapse
Affiliation(s)
- Aiman Masroor
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh 202002, India
| | - Nida Zaidi
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh 202002, India
| | - Tajalli Ilm Chandel
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh 202002, India
| | - Zoha Aqueel
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh 202002, India
| | - Sadia Malik
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh 202002, India
| | - Rizwan Hasan Khan
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh 202002, India
| |
Collapse
|
24
|
Scheffer D, Kulcsár G, Nagyéri G, Kiss-Merki M, Rékási Z, Maloy M, Czömpöly T. Active mixture of serum-circulating small molecules selectively inhibits proliferation and triggers apoptosis in cancer cells via induction of ER stress. Cell Signal 2019; 65:109426. [PMID: 31669205 DOI: 10.1016/j.cellsig.2019.109426] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 09/19/2019] [Indexed: 12/14/2022]
Abstract
Genetic and epigenetic regulation as well as immune surveillance are known defense mechanisms to protect organisms from developing cancer. Based on experimental evidence, we proposed that small metabolically active molecules accumulating in cancer cells may play a role in an alternative antitumor surveillance system. Previously, we reported that treatment with a mixture of experimentally selected small molecules, usually found in the serum (defined 'active mixture', AM), selectively induces apoptosis in cancer cells and significantly inhibits tumor formation in vivo. In this study, we show that the AM elicits gene expression changes characteristic of endoplasmic reticulum (ER) stress in HeLa, MCF-7, PC-3 and Caco-2 cancer cells, but not in primary human renal epithelial cells. The activation of the ER stress pathway was confirmed by the upregulation of ATF3, ATF4, CHAC1, DDIT3 and GDF15 proteins. Mechanistically, our investigation revealed that eIF2α, PERK and IRE1α are phosphorylated upon treatment with the AM, linking the induction of ER stress to the antiproliferative and proapoptotic effects of the AM previously demonstrated. Inhibition of ER stress in combination with BBC3 and PMAIP1 knockdown completely abrogated the effect of the AM. Moreover, we also demonstrated that the AM induces mIR-3189-3p, which in turn enhances the expression of ATF3 and DDIT3, thus representing a possible new feedback mechanism in the regulation of ATF3 and DDIT3 during ER stress. Our results highlight small molecules as attractive anticancer agents and warrant further evaluation of the AM in cancer therapy, either alone or in combination with other ER stress inducing agents.
Collapse
Affiliation(s)
- Dalma Scheffer
- Cancer Research and Drug Development Center, Culevit Ltd., Pécs, Finn u. 1/1., 7630, Hungary
| | - Gyula Kulcsár
- Cancer Research and Drug Development Center, Culevit Ltd., Pécs, Finn u. 1/1., 7630, Hungary
| | - György Nagyéri
- Soft Flow Hungary Ltd., Pécs, Ürögi fasor 2/A, 7628, Hungary
| | | | - Zoltán Rékási
- Department of Anatomy, Medical School, University of Pécs, Pécs, Szigeti u. 12., 7624, Hungary
| | - Magnus Maloy
- Department of Anatomy, Medical School, University of Pécs, Pécs, Szigeti u. 12., 7624, Hungary
| | - Tamás Czömpöly
- Cancer Research and Drug Development Center, Culevit Ltd., Pécs, Finn u. 1/1., 7630, Hungary.
| |
Collapse
|
25
|
Mimori S, Kawada K, Saito R, Takahashi M, Mizoi K, Okuma Y, Hosokawa M, Kanzaki T. Indole-3-propionic acid has chemical chaperone activity and suppresses endoplasmic reticulum stress-induced neuronal cell death. Biochem Biophys Res Commun 2019; 517:623-628. [PMID: 31378367 DOI: 10.1016/j.bbrc.2019.07.074] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 07/19/2019] [Indexed: 12/14/2022]
Abstract
Insoluble aggregated proteins are often associated with neurodegenerative diseases. Previously, we investigated chemical chaperones that prevent the aggregation of denatured proteins. Among these, 4-phenyl butyric acid (4-PBA) has well-documented chemical chaperone activity, but is required at doses that have multiple effects on cells, warranting further optimization of treatment regimens. In this study, we demonstrate chemical chaperone activities of the novel compound indole-3-propionic acid (IPA). Although it has already been reported that IPA prevents β-amyloid aggregation, herein we show that this compound suppresses aggregation of denatured proteins. Our experiments with a cell culture model of Parkinson's disease are the first to show that IPA prevents endoplasmic reticulum (ER) stress and thereby protects against neuronal cell death. We suggest that IPA has potential for the treatment of neurodegenerative diseases and other diseases for which ER stress has been implicated.
Collapse
Affiliation(s)
- Seisuke Mimori
- Department of Clinical Medicine, Faculty of Pharmacy, Chiba Institute of Science, 15-8 Shiomicho, Choshi, Chiba, 288-0025, Japan.
| | - Koichi Kawada
- Department of Pharmacology, Faculty of Pharmacy, Chiba Institute of Science, 15-8 Shiomicho, Choshi, Chiba, 288-0025, Japan
| | - Ryo Saito
- Advanced Clinical Research Center, Southern Tohoku Research Institute for Neuroscience, 255 Furusawa-tsuko, Asao-ku, Kawasaki, Kanagawa, 215-0026, Japan
| | - Masato Takahashi
- Laboratory of Drug Metabolism and Biopharmaceutics, Faculty of Pharmaceutical Sciences, Chiba Institute of Science, 15-8 Shiomicho, Choshi, Chiba, 288-0025, Japan
| | - Kenta Mizoi
- Department of Pharmacy, Faculty of Pharmacy, Takasaki University of Health and Welfare, 60, Nakaorui-machi, Takasaki, Gunma, 377-0033, Japan
| | - Yasunobu Okuma
- Department of Pharmacology, Faculty of Pharmacy, Chiba Institute of Science, 15-8 Shiomicho, Choshi, Chiba, 288-0025, Japan
| | - Masakiyo Hosokawa
- Laboratory of Drug Metabolism and Biopharmaceutics, Faculty of Pharmaceutical Sciences, Chiba Institute of Science, 15-8 Shiomicho, Choshi, Chiba, 288-0025, Japan
| | - Tetsuto Kanzaki
- Department of Drug Informatics, Graduate School and Faculty of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8675, Japan
| |
Collapse
|
26
|
Huang GH, Chen K, Sun YY, Zhu L, Sun ZL, Feng DF. 4-Phenylbutyrate Ameliorates Anxiety Disorder by Inhibiting Endoplasmic Reticulum Stress after Diffuse Axonal Injury. J Neurotrauma 2019; 36:1856-1868. [PMID: 30582423 DOI: 10.1089/neu.2018.6048] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Diffuse axonal injury (DAI) is accompanied frequently by adverse sequelae and psychiatric disorders, such as anxiety, leading to a decreased quality of life, social isolation, and poor outcomes in patients. The mechanisms regulating psychiatric disorders post-DAI are not well elucidated, however. Previous studies showed that endoplasmic reticulum (ER) stress functions as a pivotal factor in neurodegeneration disease. In this study, we showed that DAI can trigger ER stress and unfolded protein response (UPR) activation in both the acute and chronic periods, leading to cell death and anxiety disorder. Treatment with 4-phenylbutyrate (4-PBA) is able to inhibit the UPR and cell apoptosis and relieve the anxiety disorder in our DAI model. Later (14 days post-DAI) 4-PBA treatment, however, can restore only the related gene expression of ER stress and UPR but not the psychiatric disorder. Therefore, the early (5 min after DAI) administration of 4-PBA might be a therapeutic approach for blocking the ER stress/UPR-induced cell death and anxiety disorder after DAI.
Collapse
Affiliation(s)
- Guo-Hui Huang
- 1 Department of Neurosurgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,2 Institute of Traumatic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kui Chen
- 1 Department of Neurosurgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,2 Institute of Traumatic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi-Yu Sun
- 1 Department of Neurosurgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,2 Institute of Traumatic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liang Zhu
- 1 Department of Neurosurgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhao-Liang Sun
- 1 Department of Neurosurgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dong-Fu Feng
- 1 Department of Neurosurgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,2 Institute of Traumatic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
27
|
Mai CT, Le QG, Ishiwata-Kimata Y, Takagi H, Kohno K, Kimata Y. 4-Phenylbutyrate suppresses the unfolded protein response without restoring protein folding in Saccharomyces cerevisiae. FEMS Yeast Res 2019; 18:4858386. [PMID: 29452364 DOI: 10.1093/femsyr/foy016] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Accepted: 02/13/2018] [Indexed: 12/16/2022] Open
Abstract
Accumulation of unfolded secretory proteins in the endoplasmic reticulum (ER), namely ER stress, is hazardous to eukaryotic cells and promotes the unfolded protein response (UPR). Ire1 is an ER-located transmembrane protein that senses ER stress and triggers the UPR. According to previous in vitro experiments, 4-phenylbutyrate (4-PBA) works as a chemical molecular chaperone. Since 4-PBA attenuates the UPR in mammalian tissue cultures, this chemical may have clinical potential for restoring ER-stressing conditions. In this study, we investigated 4-PBA's mode of action using the yeast Saccharomyces cerevisiae as a model organism. Although 4-PBA blocked a dithiothreitol (DTT)-induced UPR, it did not appear to restore impairment of ER protein folding that was caused by DTT. Moreover, even under non-stress conditions, 4-PBA attenuated UPR that was induced by an Ire1 mutant that exhibits a substantial activity without sensing ER accumulation of unfolded proteins. We also found that 4-PBA drastically promotes the degradation of Ire1. These observations indicate that at least in the case of yeast cells, 4-PBA suppresses the UPR not through restoration of the ER function to correctly fold proteins. Instead, the accelerated degradation of Ire1 possibly explains the reason why the UPR is attenuated by 4-PBA.
Collapse
Affiliation(s)
- Chi Thanh Mai
- Graduate School of Biological Sciences, Nara Institute of Science and Technology, 8916-5 Takayama, Ikoma, Nara 630-0192, Japan
| | - Quynh Giang Le
- Graduate School of Biological Sciences, Nara Institute of Science and Technology, 8916-5 Takayama, Ikoma, Nara 630-0192, Japan
| | - Yuki Ishiwata-Kimata
- Graduate School of Biological Sciences, Nara Institute of Science and Technology, 8916-5 Takayama, Ikoma, Nara 630-0192, Japan
| | - Hiroshi Takagi
- Graduate School of Biological Sciences, Nara Institute of Science and Technology, 8916-5 Takayama, Ikoma, Nara 630-0192, Japan
| | - Kenji Kohno
- Institute for Research Initiatives, Nara Institute of Science and Technology, 8916-5 Takayama, Ikoma, Nara 630-0192, Japan
| | - Yukio Kimata
- Graduate School of Biological Sciences, Nara Institute of Science and Technology, 8916-5 Takayama, Ikoma, Nara 630-0192, Japan
| |
Collapse
|
28
|
Noguchi T, Suzuki M, Mutoh N, Hirata Y, Tsuchida M, Miyagawa S, Hwang GW, Aoki J, Matsuzawa A. Nuclear-accumulated SQSTM1/p62-based ALIS act as microdomains sensing cellular stresses and triggering oxidative stress-induced parthanatos. Cell Death Dis 2018; 9:1193. [PMID: 30546061 PMCID: PMC6294141 DOI: 10.1038/s41419-018-1245-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 11/13/2018] [Accepted: 11/27/2018] [Indexed: 12/25/2022]
Abstract
Aggresome-like induced structures (ALIS) have been described as ubiquitinated protein-containing aggresomes transiently formed in response to various stresses. In this study, we provide evidence that ALIS composed of SQSTM1/p62 act as a key determinant of oxidative stress-induced parthanatos, which is newly discovered and distinct from regular programmed cell death. Interestingly, we first found that chemical stresses induced by particular chemical drugs, such as several cephalosporin antibiotics, cause oxidative stress-mediated parthanatos, accompanied by the ALIS formation. Blocking the ALIS formation potently suppressed the parthanatos, and p62 knockout cells exhibited the attenuated ALIS formation and high resistance to parthanatos. Moreover, we also found that the redox-sensing activity of p62 is required for nuclear accumulation of the p62-based ALIS, resulting in the induction of parthanatos. Together, our results demonstrate unexpected functions of p62 and ALIS as cell death mediators sensing oxidative stress, and thus uncover a novel mechanism whereby p62 mediates parthanatos.
Collapse
Affiliation(s)
- Takuya Noguchi
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba-6-3, Aramaki, Aoba-ku, Sendai, 980-8578, Japan
| | - Midori Suzuki
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba-6-3, Aramaki, Aoba-ku, Sendai, 980-8578, Japan
| | - Natsumi Mutoh
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba-6-3, Aramaki, Aoba-ku, Sendai, 980-8578, Japan
| | - Yusuke Hirata
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba-6-3, Aramaki, Aoba-ku, Sendai, 980-8578, Japan
| | - Mei Tsuchida
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba-6-3, Aramaki, Aoba-ku, Sendai, 980-8578, Japan
| | - Sayoko Miyagawa
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba-6-3, Aramaki, Aoba-ku, Sendai, 980-8578, Japan
| | - Gi-Wook Hwang
- Laboratory of Molecular and Biochemical Toxicology, Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba-6-3, Aramaki, Aoba-ku, Sendai, 980-8578, Japan
| | - Junken Aoki
- Laboratory of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba-6-3, Aramaki, Aoba-ku, Sendai, 980-8578, Japan
| | - Atsushi Matsuzawa
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba-6-3, Aramaki, Aoba-ku, Sendai, 980-8578, Japan.
| |
Collapse
|
29
|
Bozi LH, Takano AP, Campos JC, Rolim N, Dourado PM, Voltarelli VA, Wisløff U, Ferreira JC, Barreto-Chaves ML, Brum PC. Endoplasmic reticulum stress impairs cardiomyocyte contractility through JNK-dependent upregulation of BNIP3. Int J Cardiol 2018; 272:194-201. [DOI: 10.1016/j.ijcard.2018.08.070] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 08/22/2018] [Indexed: 12/22/2022]
|
30
|
Hertz E, Terenius L, Vukojević V, Svenningsson P. GPR37 and GPR37L1 differently interact with dopamine 2 receptors in live cells. Neuropharmacology 2018; 152:51-57. [PMID: 30423289 PMCID: PMC6599889 DOI: 10.1016/j.neuropharm.2018.11.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 10/26/2018] [Accepted: 11/08/2018] [Indexed: 02/09/2023]
Abstract
Receptor-receptor interactions are essential to fine tune receptor responses and new techniques enable closer characterization of the interactions between involved proteins directly in the plasma membrane. Fluorescence cross-correlation spectroscopy (FCCS), which analyses concurrent movement of bound molecules with single-molecule detection limit, was here used to, in live N2a cells, study interactions between the Parkinson's disease (PD) associated orphan receptor GPR37, its homologue GPR37L1, and the two splice variants of the dopamine 2 receptor (D2R). An interaction between GPR37 and both splice forms of D2R was detected. 4-phenylbutyrate (4-PBA), a neuroprotective chemical chaperone known to increase GPR37 expression at the cell surface, increased the fraction of interacting molecules. The interaction was also increased by pramipexole, a D2R agonist commonly used in the treatment of PD, indicating a possible clinically relevance. Cross-correlation, indicating interaction between GPR37L1 and the short isoform of D2R, was also detected. However, this interaction was not changed with 4-PBA or pramipexole treatment. Overall, these data provide further evidence that heteromeric GPR37-D2R exist and can be pharmacologically modulated, which is relevant for the treatment of PD. This article is part of the Special Issue entitled ‘Receptor heteromers and their allosteric receptor-receptor interactions’. GPCR interaction is studied with fluorescence cross-correlation spectroscopy. Interaction between GPR37 and both isoforms of D2R is detected in live cells. GPR37's homologue GPR37L1 is detected to interact with D2RS in live cells. GPR37-D2R interaction is increased by D2-like agonist and 4-PBA treatment.
Collapse
Affiliation(s)
- E Hertz
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden.
| | - L Terenius
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - V Vukojević
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - P Svenningsson
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
31
|
Kumari N, Kumar A, Thapa BR, Modi M, Pal A, Prasad R. Characterization of mutation spectrum and identification of novel mutations in ATP7B gene from a cohort of Wilson disease patients: Functional and therapeutic implications. Hum Mutat 2018; 39:1926-1941. [PMID: 30120852 DOI: 10.1002/humu.23614] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 08/09/2018] [Accepted: 08/13/2018] [Indexed: 12/13/2022]
Abstract
Wilson disease (WD), a copper metabolism disorder, occurs due to the presence of mutations in the gene encoding ATP7B, a protein that primarily facilitates hepatic copper excretion. A better understanding of spectrum and functional significance of ATP7B variants is critical to formulating targeted and personalized therapies. Henceforth, we screened and sequenced 21 exons of ATP7B gene from 50 WD patients and 60 healthy subjects. We identified 28 variants comprising, seven novels in 20% alleles, while eight variations affecting 23% alleles were first time reported in Indian cohort. The c.813C>A, p.(Cys271*) (10%) was the most frequent mutation. Bioinformatics analysis revealed five of seven novel variants viz. c.1600C>A, p.(Pro534Thr); c.1616C>A, p.(Pro539His); c.1924G>T, p.(Asp642Tyr); c.2168G>C, p.(Arg723Thr); c.2174G>C, p.(Arg725Thr) resulted in protein misfolding. Sequence conservation analysis of ATP7B regions containing novel variants documented an evolutionarily conserved nature. Functional analysis of these novel variants in five different cell lines lacking inherent ATP7B expression demonstrated sensitivity to CuCl2 -treatment, experiencing augmented cellular copper retention and decreased copper excretion as well as ceruloplasmin secretion to that of wildtype-ATP7B expressing cells. Interestingly, pharmacological chaperone 4-phenylbutyrate, a clinically approved compound, partially restored protein function of ATP7B mutants. These findings might enable novel treatment strategies in WD by clinically enhancing the protein expression of mutant ATP7B with residual copper export activity.
Collapse
Affiliation(s)
- Niti Kumari
- Department of Biochemistry, PGIMER, Chandigarh, India
| | - Aman Kumar
- Department of Biochemistry, PGIMER, Chandigarh, India
| | - Babu Ram Thapa
- Department of Paediatrics Gastroenterology, PGIMER, Chandigarh, India
| | - Manish Modi
- Department of Neurology, PGIMER, Chandigarh, India
| | - Arnab Pal
- Department of Biochemistry, PGIMER, Chandigarh, India
| | | |
Collapse
|
32
|
Roberts NB, Alqazzaz A, Hwang JR, Qi X, Keegan AD, Kim AJ, Winkles JA, Woodworth GF. Oxaliplatin disrupts pathological features of glioma cells and associated macrophages independent of apoptosis induction. J Neurooncol 2018; 140:497-507. [PMID: 30132163 DOI: 10.1007/s11060-018-2979-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 08/06/2018] [Indexed: 11/29/2022]
Abstract
INTRODUCTION Emerging evidence suggests that effective treatment of glioblastoma (GBM), the most common and deadly form of adult primary brain cancer, will likely require concurrent treatment of multiple aspects of tumor pathobiology to overcome tumor heterogeneity and the complex tumor-supporting microenvironment. Recent studies in non-central nervous system (CNS) tumor cells have demonstrated that oxaliplatin (OXA) can induce multi-faceted anti-tumor effects, in particular at drug concentrations below those required to induce apoptosis. These findings motivated re-investigation of OXA for the treatment of GBM. METHODS The effects of OXA on murine KR158 and GL261 glioma cells including cell growth, cell death, inhibition of signal transducer and activator of transcription (STAT) activity, O-6-methylguanine-DNA methyltransferase (MGMT) expression, and immunogenic cell death (ICD) initiation, were evaluated by cytotoxicity assays, Western blot analysis, STAT3-luciferase reporter assays, qRT-PCR assays, and flow cytometry. Chemical inhibitors of endoplasmic reticulum (ER) stress were used to investigate the contribution of this cell damage response to the observed OXA effects. The effect of OXA on bone marrow-derived macrophages (BMDM) exposed to glioma conditioned media (GCM) was also analyzed by Western blot analysis. RESULTS We identified the OXA concentration threshold for induction of apoptosis and from this determined the drug dose and treatment period for sub-cytotoxic treatments of glioma cells. Under these experimental conditions, OXA reduced STAT3 activity, reduced MGMT levels and increased temozolomide sensitivity. In addition, there was evidence of immunogenic cell death (elevated EIF2α phosphorylation and calreticulin exposure) following prolonged OXA treatment. Notably, inhibition of ER stress reversed the OXA-mediated inhibition of STAT3 activity and MGMT expression in the tumor cells. In BMDMs exposed to GCM, OXA also reduced levels of phosphorylated STAT3 and decreased expression of Arginase 1, an enzyme known to contribute to pro-tumor functions in the tumor-immune environment. CONCLUSIONS OXA can induce notable multi-faceted biological effects in glioma cells and BMDMs at relatively low drug concentrations. These findings may have significant therapeutic relevance against GBM and warrant further investigation.
Collapse
Affiliation(s)
- Nathan B Roberts
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.,University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, 21201, USA
| | - Aymen Alqazzaz
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.,University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, 21201, USA
| | - Jacqueline R Hwang
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.,University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, 21201, USA
| | - Xiulan Qi
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Achsah D Keegan
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.,Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.,Research and Development Service, U.S. Department of Veterans Affairs, Veterans Affairs Maryland Health Care System, Baltimore, MD, 21201, USA
| | - Anthony J Kim
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.,University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, 21201, USA
| | - Jeffrey A Winkles
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, 21201, USA. .,Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA. .,Department of Surgery, University of Maryland School of Medicine, 800 West Baltimore Street, Baltimore, MD, 21201, USA.
| | - Graeme F Woodworth
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.,University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, 21201, USA
| |
Collapse
|
33
|
Jiang X, Fang G, Dong L, Jin P, Ding L, Zhang H, Fan J, Mao S, Fan X, Gong Y, Wang Y. Chemical chaperone 4-phenylbutyric acid alleviates the aggregation of human familial pulmonary fibrosis-related mutant SP-A2 protein in part through effects on GRP78. Biochim Biophys Acta Mol Basis Dis 2018; 1864:3546-3557. [PMID: 30293573 DOI: 10.1016/j.bbadis.2018.08.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 08/06/2018] [Accepted: 08/07/2018] [Indexed: 12/13/2022]
Abstract
G231V and F198S mutations in surfactant protein A2 (SP-A2) are associated with familial pulmonary fibrosis. These mutations cause defects in dimer/trimer assembly, trafficking, and secretion, as well as cause mutant protein aggregation. We investigated the effects and mechanisms of chemical chaperones on the cellular and biochemical properties of mutant SP-A2. Chemical chaperones, including 4-phenyl butyric acid (4-PBA), could enhance secretion and decrease intracellular aggregation of mutant SP-A2 in a dose-dependent manner. Interestingly, increased levels of aggregated mutant SP-A2, resulting from MG-132-mediated proteasome inhibition, could also be alleviated by 4-PBA. 4-PBA treatment reduced the degradation of mutant SP-A2 to chymotrypsin digestion in CHO-K1 cells and up-regulated GRP78 (BiP) expression. Overexpression of GRP78 in SP-A2 G231V- or F198S-expressing cells reduced, whereas shRNA-mediated knockdown of GRP78 enhanced aggregation of mutant SP-A2, suggesting that GRP78 regulates aggregation of mutant SP-A2. Together, these data indicate chemical chaperone 4-PBA and upregulation of GRP78 can alleviate aggregation to stabilize and facilitate secretion of mutant SP-A2. The up-regulation expression of GRP78 might partially contribute to the aggregate-alleviating effect of 4-PBA.
Collapse
Affiliation(s)
- Xu Jiang
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Guodong Fang
- Department of Pathology, Shaanxi Provincial People's Hospital, The Third Affiliated Hospital of Xi'an JiaoTong University, Xi'an 710068, Shaanxi, China
| | - Li Dong
- Department of Respiratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, Zhejiang, China
| | - Peifeng Jin
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, Zhejiang, China
| | - Lu Ding
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Haizeng Zhang
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Junming Fan
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Sunzhong Mao
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaofang Fan
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yongsheng Gong
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Yongyu Wang
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
34
|
Tochiya M, Hagiwara D, Azuma Y, Miyata T, Morishita Y, Suga H, Onoue T, Tsunekawa T, Takagi H, Ito Y, Iwama S, Goto M, Banno R, Arima H. Chemical chaperone 4-phenylbutylate reduces mutant protein accumulation in the endoplasmic reticulum of arginine vasopressin neurons in a mouse model for familial neurohypophysial diabetes insipidus. Neurosci Lett 2018; 682:50-55. [DOI: 10.1016/j.neulet.2018.06.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 05/28/2018] [Accepted: 06/06/2018] [Indexed: 12/12/2022]
|
35
|
Wilding AS, Patte-Mensah C, Taleb O, Brun S, Kemmel V, Mensah-Nyagan AG. Protective effect of 4-Phenylbutyrate against proteolipid protein mutation-induced endoplasmic reticulum stress and oligodendroglial cell death. Neurochem Int 2018; 118:185-194. [PMID: 29936187 DOI: 10.1016/j.neuint.2018.06.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 06/19/2018] [Accepted: 06/20/2018] [Indexed: 01/08/2023]
Abstract
Proteolipid protein (PLP) mutation causes oligodendrocyte degeneration and myelin disorders including Pelizaeus-Merzbacher Disease (PMD). As the pathophysiological mechanisms involved in PMD are poorly known, the development of therapies remains difficult. To elucidate the pathogenic pathways, an immortalized oligodendroglial cell line (158JP) expressing PLP mutation has been generated. Previous investigations revealed that 158JP oligodendrocytes exhibit several abnormalities including aberrant PLP insertion into the plasma membrane, cAMP, plasmalogen and cell cycle deficits. However, further clarifications of abnormal PLP-induced oligodendrocyte degeneration are required in order to identify relevant mechanisms to target for efficient protection against oligodendrocyte death. Because PLP overexpression may lead to its accumulation inside the endoplasmic reticulum (ER) and cause ER-stress, we explored whether ER-stress may pivotally determine 158JP cell survival/death. Viability assays, RT-qPCR, western blot and flow cytometry were combined to compare cell survival, ER-stress and apoptotic markers in 158JP and control (158N) oligodendrocytes. We observed a significant decreased viability/survival of 158JP compared to 158N cells. Consistently, ER-stress markers (BiP, caspase-12) increased in 158JP (+30%) compared to the controls. mRNA and protein ratios of apoptotic modulators (Bax/Bcl2) are higher in 158JP oligodendrocytes which are also more vulnerable than 158N cells to tunicamycin-induced ER-stress. Interestingly, 4-Phenylbutyrate (ER-stress inhibitor), which decreased ER-stress and apoptotic markers in 158JP cells, significantly increased their survival. Our results, which show a direct link between the viability and endogenous levels of ER-stress and apoptotic markers in 158JP cells, also suggest that 4-Phenylbutyrate-based strategy may contribute to develop effective strategies against oligodendrocyte dysfunctions/death and myelin disorders.
Collapse
Affiliation(s)
- Anne-Sophie Wilding
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, France
| | - Christine Patte-Mensah
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, France
| | - Omar Taleb
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, France
| | - Susana Brun
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, France
| | - Véronique Kemmel
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, France
| | - Ayikoe-Guy Mensah-Nyagan
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, France.
| |
Collapse
|
36
|
Endoplasmic reticulum stress induced by an ethanol extract of Coicis semen in Chang liver cells. Altern Ther Health Med 2018; 18:100. [PMID: 29554897 PMCID: PMC5859727 DOI: 10.1186/s12906-018-2175-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 03/15/2018] [Indexed: 12/21/2022]
Abstract
Background It is well known that endoplasmic reticulum (ER) stress plays a huge role in development of metabolic diseases. Specially, ER stress-induced cellular dysfunction has a significant involvement in the pathogenesis of human chronic disorders. This study was designed to study to assess whether an ethanol extract of Coicis Semen (CSE) and coixol induces the ER stress in Chang liver cells. Methods Coicis Semen was mixed with 95% ethanol at a ratio of 1:10 (w/v) and freeze dried. Chang liver cells were seeded to 96-well plates and treated with or without CSE (100, 200, 300, 500, or 1000 μg/mL) or coixol (100, 200, 300, 500, 750, or 1000 μg/mL). cell viability was analyzed with MTT assay. Effects of CSE and coixol on expression of the genes for ER stress markers were determined with qRT-PCR and the expression of the protein levels of ER stress markers were determined with western blotting. Results The concentration causing 50% inhibition (IC50) for CSE and coixol was 250 and 350 μg/mL, respectively. The CSE and coixol increased the gene expression of BiP and CHOP in a dose-dependent manner. Furthermore, CSE and coixol dose-dependently increased the the expression of XBP1. Conclusions CSE or coixol may have cytotoxic effect to Chang liver cells and, may induce ER stress and stimulate the UPR via activation of the PERK and IRE1 pathways in normal liver cells. Electronic supplementary material The online version of this article (10.1186/s12906-018-2175-z) contains supplementary material, which is available to authorized users.
Collapse
|
37
|
Mycobacterium fortuitum-induced ER-Mitochondrial calcium dynamics promotes calpain/caspase-12/caspase-9 mediated apoptosis in fish macrophages. Cell Death Discov 2018. [PMID: 29531827 PMCID: PMC5841318 DOI: 10.1038/s41420-018-0034-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Mycobacterium fortuitum is a natural fish pathogen. It induces apoptosis in headkidney macrophages (HKM) of catfish, Clarias sp though the mechanism remains largely unknown. We observed M. fortuitum triggers calcium (Ca2+) insult in the sub-cellular compartments which elicits pro-apototic ER-stress factor CHOP. Alleviating ER-stress inhibited CHOP and attenuated HKM apoptosis implicating ER-stress in the pathogenesis of M. fortuitum. ER-stress promoted calpain activation and silencing the protease inhibited caspase-12 activation. The study documents the primal role of calpain/caspase-12 axis on caspase-9 activation in M. fortuitum-pathogenesis. Mobilization of Ca2+ from ER to mitochondria led to increased mitochondrial Ca2+ (Ca2+)m load,, mitochondrial permeability transition (MPT) pore opening, altered mitochondrial membrane potential (ΔΨm) and cytochrome c release eventually activating the caspase-9/-3 cascade. Ultra-structural studies revealed close apposition of ER and mitochondria and pre-treatment with (Ca2+)m-uniporter (MUP) blocker ruthenium red, reduced Ca2+ overload suggesting (Ca2+)m fluxes are MUP-driven and the ER-mitochondria tethering orchestrates the process. This is the first report implicating role of sub-cellular Ca2+ in the pathogenesis of M. fortuitum. We summarize, the dynamics of Ca2+ in sub-cellular compartments incites ER-stress and mitochondrial dysfunction, leading to activation of pro-apoptotic calpain/caspase-12/caspase-9 axis in M. fortuitum-infected HKM.
Collapse
|
38
|
Kaur B, Bhat A, Chakraborty R, Adlakha K, Sengupta S, Roy S, Chakraborty K. Proteomic profile of 4-PBA treated human neuronal cells during ER stress. Mol Omics 2018; 14:53-63. [PMID: 29570205 DOI: 10.1039/c7mo00114b] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Perturbations affecting the homoeostasis of endoplasmic reticulum (ER) activate an adaptive signaling known as the unfolded protein response or UPR. Many studies have reported the association between neurological disorders and ER stress. Decreasing ER stress may therefore aid in therapeutic control of neuronal diseases. Sodium 4-phenylbutyrate (4-PBA), a small molecule, has been shown to alleviate ER stress and various neurological diseases, but the mechanistic basis of its action is not well understood. Using an iTRAQ based LC-MS technique we have delineated the effect of 4-PBA on the proteome of human neuroblastoma cells (SK-N-SH) during Tunicamycin-induced ER stress. The proteomic profile of 4-PBA-treated cells revealed that 4-PBA does not alter the cellular proteome to adapt towards ER stress. However, it can alleviate both the toxicity and proteomic alterations, induced by an ER stress inducer. Hence, the therapeutic effect of 4-PBA is primarily due to its ability to resolve ER stress rather than its ability to alter the expression of proteins required for maintaining ER proteostasis. Thus, we posit here that 4-PBA acts as an authentic chemical chaperone by aiding protein folding in the ER.
Collapse
Affiliation(s)
- Bhavneet Kaur
- Genomics and Molecular Medicine, CSIR-IGIB, Mathura Road, New Delhi, India.
| | | | | | | | | | | | | |
Collapse
|
39
|
Morphine-Mediated Brain Region-Specific Astrocytosis Involves the ER Stress-Autophagy Axis. Mol Neurobiol 2018; 55:6713-6733. [PMID: 29344928 DOI: 10.1007/s12035-018-0878-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 01/07/2018] [Indexed: 01/08/2023]
Abstract
A recent study from our lab has revealed a link between morphine-mediated autophagy and synaptic impairment. The current study was aimed at investigating whether morphine-mediated activation of astrocytes involved the ER stress/autophagy axis. Our in vitro findings demonstrated upregulation of GFAP indicating astrocyte activation with a concomitant increase in the production of proinflammatory cytokines in morphine-exposed human astrocytes. Using both pharmacological and gene-silencing approaches, it was demonstrated that morphine-mediated defective autophagy involved upstream activation of ER stress with subsequent downstream astrocyte activation via the μ-opioid receptor (MOR). In vivo validation demonstrated preferential activation of ER stress/autophagy axis in the areas of the brain not associated with pain such as the basal ganglia, frontal cortex, occipital cortex, and the cerebellum of morphine-dependent rhesus macaques, and this correlated with increased astrocyte activation and neuroinflammation. Interventions aimed at blocking either the MOR or ER stress could thus likely be developed as promising therapeutic targets for abrogating morphine-mediated astrocytosis.
Collapse
|
40
|
Alzheimer's disease pathology and the unfolded protein response: prospective pathways and therapeutic targets. Behav Pharmacol 2018; 28:161-178. [PMID: 28252521 DOI: 10.1097/fbp.0000000000000299] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Many vital interdependent cellular functions including proteostasis, lipogenesis and Ca homeostasis are executed by the endoplasmic reticulum (ER). Exogenous insults can impair ER performance: this must be rapidly corrected or cell death will ensue. Protective adaptations can boost the functional capacity of the ER and form the basis of the unfolded protein response (UPR). Activated in response to the accumulation of misfolded proteins, the UPR can halt protein translation while increasing protein-handling chaperones and the degradation of erroneous proteins through a conserved three-tier molecular cascade. However, prolonged activation of the UPR can result in the maladaptation of the system, resulting in the activation of inflammatory and apoptotic effectors. Recently, UPR and its involvement in neurodegenerative disease has attracted much interest and numerous potentially 'drugable' points of crosstalk are now emerging. Here, we summarize the functions of the ER and UPR, and highlight evidence for its potential role in the pathogenesis of Alzheimer's disease, before discussing several key targets with therapeutic potential.
Collapse
|
41
|
Phenyl butyrate inhibits pyruvate dehydrogenase kinase 1 and contributes to its anti-cancer effect. Eur J Pharm Sci 2017; 110:93-100. [DOI: 10.1016/j.ejps.2017.04.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 04/21/2017] [Accepted: 04/23/2017] [Indexed: 12/15/2022]
|
42
|
Crosstalk between endoplasmic reticulum stress and oxidative stress in schizophrenia: The dawn of new therapeutic approaches. Neurosci Biobehav Rev 2017; 83:589-603. [DOI: 10.1016/j.neubiorev.2017.08.025] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 08/09/2017] [Accepted: 08/30/2017] [Indexed: 01/15/2023]
|
43
|
Schoenhacker-Alte B, Mohr T, Pirker C, Kryeziu K, Kuhn PS, Buck A, Hofmann T, Gerner C, Hermann G, Koellensperger G, Keppler BK, Berger W, Heffeter P. Sensitivity towards the GRP78 inhibitor KP1339/IT-139 is characterized by apoptosis induction via caspase 8 upon disruption of ER homeostasis. Cancer Lett 2017; 404:79-88. [PMID: 28716523 DOI: 10.1016/j.canlet.2017.07.009] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 07/05/2017] [Accepted: 07/07/2017] [Indexed: 11/30/2022]
Abstract
The ruthenium drug and GRP78 inhibitor KP1339/IT-139 has already demonstrated promising anticancer activity in a phase I clinical trial. This study aimed to identify mechanisms underlying increased sensitivity to KP1339 treatment. Based on a screen utilizing 23 cell lines, a small panel was selected to compare KP1339-sensitive and low-responsive models. KP1339 sensitivity was neither based on differences in ruthenium accumulation, nor sensitivity to oxidative stress or constituents of KP1339 (ruthenium chloride and indazole). Subsequently, the biochemical response to KP1339 was analyzed using whole genome expression arrays indicating that, while sensitive cell lines were characterized by "response to chemical stimuli" and "regulation of cell death", low-responsive cells preferentially activated pathways controlling cell cycle, DNA repair, and metabolism. Cell culture experiments confirmed that, while low-responsive cells executed cell cycle arrest in G2 phase, pronounced apoptosis induction via activation of caspase 8 was found in sensitive cells. Cell death induction is based on a unique disruption of the ER homeostasis by depletion of key cellular chaperones including GRP78 in combination with enhanced KP1339-mediated protein damage.
Collapse
Affiliation(s)
- Beatrix Schoenhacker-Alte
- Institute of Inorganic Chemistry, University of Vienna, Waehringer Str. 42, A-1090, Vienna, Austria; Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center of the Medical University, Medical University of Vienna, Borschkegasse 8a, A-1090, Vienna, Austria
| | - Thomas Mohr
- Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center of the Medical University, Medical University of Vienna, Borschkegasse 8a, A-1090, Vienna, Austria
| | - Christine Pirker
- Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center of the Medical University, Medical University of Vienna, Borschkegasse 8a, A-1090, Vienna, Austria
| | - Kushtrim Kryeziu
- Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center of the Medical University, Medical University of Vienna, Borschkegasse 8a, A-1090, Vienna, Austria
| | - Paul-Steffen Kuhn
- Institute of Inorganic Chemistry, University of Vienna, Waehringer Str. 42, A-1090, Vienna, Austria
| | - Alicia Buck
- Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center of the Medical University, Medical University of Vienna, Borschkegasse 8a, A-1090, Vienna, Austria
| | - Thilo Hofmann
- Department of Environmental Geosciences, University of Vienna, Althanstraße 14, A-1090, Vienna, Austria
| | - Christopher Gerner
- Department of Analytical Chemistry, University of Vienna, Waehringer Str. 38, A-1090, Vienna, Austria
| | - Gerrit Hermann
- Department of Analytical Chemistry, University of Vienna, Waehringer Str. 38, A-1090, Vienna, Austria
| | - Gunda Koellensperger
- Department of Analytical Chemistry, University of Vienna, Waehringer Str. 38, A-1090, Vienna, Austria
| | - Bernhard K Keppler
- Institute of Inorganic Chemistry, University of Vienna, Waehringer Str. 42, A-1090, Vienna, Austria; Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center of the Medical University, Medical University of Vienna, Borschkegasse 8a, A-1090, Vienna, Austria
| | - Walter Berger
- Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center of the Medical University, Medical University of Vienna, Borschkegasse 8a, A-1090, Vienna, Austria; Research Cluster "Translational Cancer Therapy Research", Vienna, Austria
| | - Petra Heffeter
- Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center of the Medical University, Medical University of Vienna, Borschkegasse 8a, A-1090, Vienna, Austria; Research Cluster "Translational Cancer Therapy Research", Vienna, Austria.
| |
Collapse
|
44
|
Vauthier V, Housset C, Falguières T. Targeted pharmacotherapies for defective ABC transporters. Biochem Pharmacol 2017; 136:1-11. [DOI: 10.1016/j.bcp.2017.02.020] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 02/23/2017] [Indexed: 02/07/2023]
|
45
|
Leinartaité L, Svenningsson P. Folding Underlies Bidirectional Role of GPR37/Pael-R in Parkinson Disease. Trends Pharmacol Sci 2017. [PMID: 28629580 DOI: 10.1016/j.tips.2017.05.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Since conformational flexibility, which is required for the function of a protein, comes at the expense of structural stability, many proteins, including G-protein-coupled receptors (GPCRs), are under constant risk of misfolding and aggregation. In this regard GPR37 (also named PAEL-R and ETBR-LP-1) takes a prominent role, particularly in relation to Parkinson disease (PD). GPR37 is a substrate for parkin and accumulates abnormally in autosomal recessive juvenile parkinsonism, contributing to endoplasmic reticulum stress and death of dopaminergic neurons. GPR37 also constitutes a core structure of Lewy bodies, demonstrating a more general involvement in PD pathology. However, if folded and matured properly, GPR37 seems to be neuroprotective. Moreover, GPR37 modulates functionality of the dopamine transporter and the dopamine D2 receptor and stimulates dopamine neurotransmission. Here we review the multiple roles of GPR37 with relevance to potential disease modification and symptomatic therapies of PD and highlight unsolved issues in this field.
Collapse
Affiliation(s)
- Lina Leinartaité
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, SE-171 76, Stockholm, Sweden.
| | - Per Svenningsson
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, SE-171 76, Stockholm, Sweden.
| |
Collapse
|
46
|
ER stress dependent microparticles derived from smooth muscle cells promote endothelial dysfunction during thoracic aortic aneurysm and dissection. Clin Sci (Lond) 2017; 131:1287-1299. [PMID: 28468950 PMCID: PMC5461939 DOI: 10.1042/cs20170252] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Revised: 04/20/2017] [Accepted: 05/02/2017] [Indexed: 12/11/2022]
Abstract
The degeneration of vascular smooth muscle cell(s) (SMC) is one of the key features of thoracic aortic aneurysm and dissection (TAAD). We and others have shown that elevated endoplasmic reticulum (ER) stress causes SMC loss and TAAD formation, however, the mechanism of how SMC dysfunction contributes to intimal damage, leading to TAAD, remains to be explored. In the present study, in vitro assay demonstrated that elevated mechanical stretch (18% elongation, 3600 cycles/h) stimulated the ER stress response and microparticle(s) (MP) production from both SMC and endothelial cell(s) (EC) in a time-dependent manner. Treatment of EC with isolated MP led to anoikis, which was determined by measuring the fluorescence of the ethidium homodimer (EthD-1) and Calcein AM cultured in hydrogel-coated plates and control plates. MP stimulation of EC also up-regulated the mRNA levels of inflammatory molecules (i.e. Vascular cellular adhesion molecular-1 (VCAM-1)), intercellular adhesion molecular-1 (ICAM-1), interleukin-1β (IL-1β), and interleukin-6 (IL-6)). Use of an ER stress inhibitor or knockout of CHOP decreased mechanical stretch-induced MP production in SMC. In vivo, administration of an ER stress inhibitor or knockout of CHOP suppressed both apoptosis of EC and the infiltration of inflammatory cells. Moreover, TAAD formation was also suppressed by the administration of an ER stress inhibitor. In conclusion, our study demonstrates that elevated mechanical stretch induces MP formation in SMC leading to endothelial dysfunction, which is ER stress dependent. The inhibition of ER stress suppressed EC apoptosis, inflammation in the aorta, and TAAD development.
Collapse
|
47
|
Fu HY, Mukai M, Awata N, Sakata Y, Hori M, Minamino T. Protein Quality Control Dysfunction in Cardiovascular Complications Induced by Anti-Cancer Drugs. Cardiovasc Drugs Ther 2017; 31:109-117. [PMID: 28120277 DOI: 10.1007/s10557-016-6709-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Cardiovascular complications, including heart failure, hypertension, ischemic syndromes and venous thromboembolism, have been identified in patients treated with anti-cancer drugs. Oxidative stress, mitochondrial dysfunction and DNA synthesis inhibition are considered to be responsible for the cardiotoxicity induced by these agents. Protein quality control (PQC) has 3 major components, including the endoplasmic reticulum (ER), the ubiquitin-proteasome system (UPS) and the autophagy-lysosome system, and participates in protein folding and degradation to maintain protein homeostasis. We have demonstrated that PQC dysfunction is a new causal mechanism for the development of cardiac hypertrophy and failure. Increasing evidence shows that anti-cancer drugs, such as tyrosine kinase inhibitors, proteasome inhibitors, anthracyclines and autophagy inhibitors, cause PQC dysfunction. Here, we provide an overview of the potential role of PQC dysfunction in the development of cardiovascular complications induced by anti-cancer drugs.
Collapse
Affiliation(s)
- Hai Ying Fu
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan.,Department of Clinical Research and Development, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Mikio Mukai
- Osaka Medical Center for Cancer and Cardiovascular Diseases, Osaka, Japan
| | - Nobuhisa Awata
- Osaka Medical Center for Cancer and Cardiovascular Diseases, Osaka, Japan
| | - Yasushi Sakata
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Masatsugu Hori
- Osaka Medical Center for Cancer and Cardiovascular Diseases, Osaka, Japan
| | - Tetsuo Minamino
- Department of Cardiorenal and Cerebrovascular Medicine, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Mikicho, Kita-gun, Kagawa Prefecture, 761-0793, Japan.
| |
Collapse
|
48
|
Li X, Nabeka H, Saito S, Shimokawa T, Khan MSI, Yamamiya K, Shan F, Gao H, Li C, Matsuda S. Expression of prosaposin and its receptors in the rat cerebellum after kainic acid injection. IBRO Rep 2017; 2:31-40. [PMID: 30135931 PMCID: PMC6084904 DOI: 10.1016/j.ibror.2017.02.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 02/02/2017] [Accepted: 02/21/2017] [Indexed: 11/16/2022] Open
Abstract
Prosaposin (PSAP), a highly conserved glycoprotein, is a precursor of saposins A–D. Accumulating evidence suggests that PSAP is a neurotrophic factor that induces differentiation and prevents death in a variety of neuronal cells through the active region within the saposin C domain both in vivo and in vitro. Recently, GPR37 and GPR37L1 were recognized as PSAP receptors. In this study, we examined the alteration in expression of PSAP and its receptors in the cerebellum using rats injected with kainic acid (KA). The results show that PSAP was strongly expressed in the cytoplasm of Purkinje cells and interneurons in the molecular layer, and that PSAP expression in both types of neurons was markedly enhanced following KA treatment. Immunoblotting revealed that the expression of GPR37 was diminished significantly three days after KA injection compared with control rats; however, no changes were observed through immunostaining. No discernable changes were found in GPR37L1. These findings may help us to understand the role of PSAP and the GPR37 and GPR37L1 receptors in alleviating the neural damage caused by KA.
Collapse
Key Words
- BSA, bovine serum albumin
- Cerebellum
- ER, endoplasmic reticulum
- GPCR, G protein-coupled receptor
- GPR37
- GPR37L1
- H-E staining, hematoxylin-eosin staining
- IF, immunofluorescence
- IHC, immunohistochemistry
- ISH, in situ hybridization
- KA, kainic acid
- Kainic acid
- Neurodegeneration
- PSAP, prosaposin
- Prosaposin
- SSC, standard saline citrate
Collapse
Affiliation(s)
- Xuan Li
- Department of Anatomy and Embryology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Hiroaki Nabeka
- Department of Anatomy and Embryology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Shouichiro Saito
- Laboratory of Veterinary Anatomy, Faculty of Applied Biological Sciences, Gifu University, Yanagido, Gifu, Japan
| | - Tetsuya Shimokawa
- Department of Anatomy and Embryology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Md Sakirul Islam Khan
- Department of Anatomy and Embryology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Kimiko Yamamiya
- Department of Anatomy and Embryology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Fengping Shan
- Department of Immunology, School of Basic Medical Science, China Medical University, Shenyang, PR China
| | - Huiling Gao
- College of Life and Health Science, Northeastern University, Shenyang, PR China
| | - Cheng Li
- Department of Immunology, School of Basic Medical Science, China Medical University, Shenyang, PR China
| | - Seiji Matsuda
- Department of Anatomy and Embryology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| |
Collapse
|
49
|
Barger SW. Gene regulation and genetics in neurochemistry, past to future. J Neurochem 2016; 139 Suppl 2:24-57. [PMID: 27747882 DOI: 10.1111/jnc.13629] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2015] [Revised: 03/01/2016] [Accepted: 03/30/2016] [Indexed: 12/14/2022]
Abstract
Ask any neuroscientist to name the most profound discoveries in the field in the past 60 years, and at or near the top of the list will be a phenomenon or technique related to genes and their expression. Indeed, our understanding of genetics and gene regulation has ushered in whole new systems of knowledge and new empirical approaches, many of which could not have even been imagined prior to the molecular biology boon of recent decades. Neurochemistry, in the classic sense, intersects with these concepts in the manifestation of neuropeptides, obviously dependent upon the central dogma (the established rules by which DNA sequence is eventually converted into protein primary structure) not only for their conformation but also for their levels and locales of expression. But, expanding these considerations to non-peptide neurotransmitters illustrates how gene regulatory events impact neurochemistry in a much broader sense, extending beyond the neurochemicals that translate electrical signals into chemical ones in the synapse, to also include every aspect of neural development, structure, function, and pathology. From the beginning, the mutability - yet relative stability - of genes and their expression patterns were recognized as potential substrates for some of the most intriguing phenomena in neurobiology - those instances of plasticity required for learning and memory. Near-heretical speculation was offered in the idea that perhaps the very sequence of the genome was altered to encode memories. A fascinating component of the intervening progress includes evidence that the central dogma is not nearly as rigid and consistent as we once thought. And this mutability extends to the potential to manipulate that code for both experimental and clinical purposes. Astonishing progress has been made in the molecular biology of neurochemistry during the 60 years since this journal debuted. Many of the gains in conceptual understanding have been driven by methodological progress, from automated high-throughput sequencing instruments to recombinant-DNA vectors that can convey color-coded genetic modifications in the chromosomes of live adult animals. This review covers the highlights of these advances, both theoretical and technological, along with a brief window into the promising science ahead. This article is part of the 60th Anniversary special issue.
Collapse
Affiliation(s)
- Steven W Barger
- Department of Geriatrics, Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA. .,Geriatric Research Education and Clinical Center, Central Arkansas Veterans Healthcare System, Little Rock, Arkansas, USA.
| |
Collapse
|
50
|
Mercado G, Castillo V, Soto P, Sidhu A. ER stress and Parkinson's disease: Pathological inputs that converge into the secretory pathway. Brain Res 2016; 1648:626-632. [DOI: 10.1016/j.brainres.2016.04.042] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 04/15/2016] [Accepted: 04/16/2016] [Indexed: 12/20/2022]
|