1
|
Zhang R, Hu Z, Wei D, Li R, Li Y, Zhang Z. Carboplatin restricts peste des petits ruminants virus replication by suppressing the STING-mediated autophagy. Front Vet Sci 2024; 11:1383927. [PMID: 38812563 PMCID: PMC11133560 DOI: 10.3389/fvets.2024.1383927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 04/30/2024] [Indexed: 05/31/2024] Open
Abstract
Peste des petits ruminants virus (PPRV) is a morbillivirus that causes the acute and highly pathogenic infectious disease peste des petits ruminants (PPR) in small ruminants and poses a major threat to the goat and sheep industries. Currently, there is no effective treatment for PPRV infection. Here, we propose Carboplatin, a platinum-based regimen designed to treat a range of malignancies, as a potential antiviral agent. We showed that Carboplatin exhibits significant antiviral activity against PPRV in a cell culture model. The mechanism of action of Carboplatin against PPRV is mainly attributed to its ability to block STING mediated autophagy. Together, our study supports the discovery of Carboplatin as an antiviral against PPRV and potentially other closely related viruses, sheds light on its mode of action, and establishes STING as a valid and attractive target to counteract viral infection.
Collapse
Affiliation(s)
| | | | | | | | - Yanmin Li
- College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu, Sichuan, China
| | - Zhidong Zhang
- College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu, Sichuan, China
| |
Collapse
|
2
|
Kapuy O. Mechanism of Decision Making between Autophagy and Apoptosis Induction upon Endoplasmic Reticulum Stress. Int J Mol Sci 2024; 25:4368. [PMID: 38673953 PMCID: PMC11050573 DOI: 10.3390/ijms25084368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 04/10/2024] [Accepted: 04/15/2024] [Indexed: 04/28/2024] Open
Abstract
Dynamic regulation of the cellular proteome is mainly controlled in the endoplasmic reticulum (ER). Accumulation of misfolded proteins due to ER stress leads to the activation of unfolded protein response (UPR). The primary role of UPR is to reduce the bulk of damages and try to drive back the system to the former or a new homeostatic state by autophagy, while an excessive level of stress results in apoptosis. It has already been proven that the proper order and characteristic features of both surviving and self-killing mechanisms are controlled by negative and positive feedback loops, respectively. The new results suggest that these feedback loops are found not only within but also between branches of the UPR, fine-tuning the response to ER stress. In this review, we summarize the recent knowledge of the dynamical characteristic of endoplasmic reticulum stress response mechanism by using both theoretical and molecular biological techniques. In addition, this review pays special attention to describing the mechanism of action of the dynamical features of the feedback loops controlling cellular life-and-death decision upon ER stress. Since ER stress appears in diseases that are common worldwide, a more detailed understanding of the behaviour of the stress response is of medical importance.
Collapse
Affiliation(s)
- Orsolya Kapuy
- Department of Molecular Biology, Institute of Biochemistry and Molecular Biology, Semmelweis University, H-1085 Budapest, Hungary
| |
Collapse
|
3
|
Liu X, Li T, Sun J, Wang Z. The Role of Endoplasmic Reticulum Stress in Calcific Aortic Valve Disease. Can J Cardiol 2023; 39:1571-1580. [PMID: 37516250 DOI: 10.1016/j.cjca.2023.07.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 06/28/2023] [Accepted: 07/23/2023] [Indexed: 07/31/2023] Open
Abstract
Calcific aortic valve disease (CAVD), which is involved in osteogenic reprogramming of valvular interstitial cells, is the most common form of valve disease. It still lacks effective pharmacologic intervention, as its cellular biological mechanisms remain unclear. Congenital abnormality (bicuspid valve) and older age are considered to be the most powerful risk factors for CAVD. Aortic valve sclerosis (AVS) and calcific aortic stenosis (CAS), 2 subclinical forms of CAVD, represent 2 distinct stages of aortic valve calcification. During the AVS stage, the disease is characterised by endothelial activation/damage, inflammatory response, and lipid infiltration accompanied by microcalcification. The CAS stage is dominated by calcification, resulting in valvular dysfunction and severe obstruction to cardiac outflow, which is life threatening if surgery is not performed in time. Endoplasmic reticulum (ER) stress, a state in which conditions disrupting ER homeostasis cause an accumulation of unfolded and misfolded proteins in the ER lumen, has been shown to promote osteogenic differentiation and aortic valve calcification. Therefore, identifying targets or drugs for suppressing ER stress may be a novel approach for CAVD treatment.
Collapse
Affiliation(s)
- Xiaolin Liu
- Department of Cardiac Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China; Medicial Science and Technology Innovation Center, Shandong First Medical University, Jinan, Shandong, China
| | - Ting Li
- School of Life Science, Shandong First Medical University (Shandong Academy of Medical Sciences), Taian, Shandong, China
| | - Jun Sun
- Department of Cardiac Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Zhengjun Wang
- Department of Cardiac Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
| |
Collapse
|
4
|
Parkkinen I, Their A, Asghar MY, Sree S, Jokitalo E, Airavaara M. Pharmacological Regulation of Endoplasmic Reticulum Structure and Calcium Dynamics: Importance for Neurodegenerative Diseases. Pharmacol Rev 2023; 75:959-978. [PMID: 37127349 DOI: 10.1124/pharmrev.122.000701] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 03/27/2023] [Accepted: 04/04/2023] [Indexed: 05/03/2023] Open
Abstract
The endoplasmic reticulum (ER) is the largest organelle of the cell, composed of a continuous network of sheets and tubules, and is involved in protein, calcium (Ca2+), and lipid homeostasis. In neurons, the ER extends throughout the cell, both somal and axodendritic compartments, and is highly important for neuronal functions. A third of the proteome of a cell, secreted and membrane-bound proteins, are processed within the ER lumen and most of these proteins are vital for neuronal activity. The brain itself is high in lipid content, and many structural lipids are produced, in part, by the ER. Cholesterol and steroid synthesis are strictly regulated in the ER of the blood-brain barrier protected brain cells. The high Ca2+ level in the ER lumen and low cytosolic concentration is needed for Ca2+-based intracellular signaling, for synaptic signaling and Ca2+ waves, and for preparing proteins for correct folding in the presence of high Ca2+ concentrations to cope with the high concentrations of extracellular milieu. Particularly, ER Ca2+ is controlled in axodendritic areas for proper neurito- and synaptogenesis and synaptic plasticity and remodeling. In this review, we cover the physiologic functions of the neuronal ER and discuss it in context of common neurodegenerative diseases, focusing on pharmacological regulation of ER Ca2+ Furthermore, we postulate that heterogeneity of the ER, its protein folding capacity, and ensuring Ca2+ regulation are crucial factors for the aging and selective vulnerability of neurons in various neurodegenerative diseases. SIGNIFICANCE STATEMENT: Endoplasmic reticulum (ER) Ca2+ regulators are promising therapeutic targets for degenerative diseases for which efficacious drug therapies do not exist. The use of pharmacological probes targeting maintenance and restoration of ER Ca2+ can provide restoration of protein homeostasis (e.g., folding of complex plasma membrane signaling receptors) and slow down the degeneration process of neurons.
Collapse
Affiliation(s)
- Ilmari Parkkinen
- Neuroscience Center (I.P., A.T., M.A.), Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy (I.P., M.A.), Cell and Tissue Dynamics Research Program, Institute of Biotechnology, Helsinki Institute of Life Sciences (M.Y.A., S.S., E.J.), and Electron Microscopy Unit, Institute of Biotechnology, Helsinki Institute of Life Sciences (E.J.), University of Helsinki, Helsinki, Finland
| | - Anna Their
- Neuroscience Center (I.P., A.T., M.A.), Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy (I.P., M.A.), Cell and Tissue Dynamics Research Program, Institute of Biotechnology, Helsinki Institute of Life Sciences (M.Y.A., S.S., E.J.), and Electron Microscopy Unit, Institute of Biotechnology, Helsinki Institute of Life Sciences (E.J.), University of Helsinki, Helsinki, Finland
| | - Muhammad Yasir Asghar
- Neuroscience Center (I.P., A.T., M.A.), Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy (I.P., M.A.), Cell and Tissue Dynamics Research Program, Institute of Biotechnology, Helsinki Institute of Life Sciences (M.Y.A., S.S., E.J.), and Electron Microscopy Unit, Institute of Biotechnology, Helsinki Institute of Life Sciences (E.J.), University of Helsinki, Helsinki, Finland
| | - Sreesha Sree
- Neuroscience Center (I.P., A.T., M.A.), Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy (I.P., M.A.), Cell and Tissue Dynamics Research Program, Institute of Biotechnology, Helsinki Institute of Life Sciences (M.Y.A., S.S., E.J.), and Electron Microscopy Unit, Institute of Biotechnology, Helsinki Institute of Life Sciences (E.J.), University of Helsinki, Helsinki, Finland
| | - Eija Jokitalo
- Neuroscience Center (I.P., A.T., M.A.), Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy (I.P., M.A.), Cell and Tissue Dynamics Research Program, Institute of Biotechnology, Helsinki Institute of Life Sciences (M.Y.A., S.S., E.J.), and Electron Microscopy Unit, Institute of Biotechnology, Helsinki Institute of Life Sciences (E.J.), University of Helsinki, Helsinki, Finland
| | - Mikko Airavaara
- Neuroscience Center (I.P., A.T., M.A.), Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy (I.P., M.A.), Cell and Tissue Dynamics Research Program, Institute of Biotechnology, Helsinki Institute of Life Sciences (M.Y.A., S.S., E.J.), and Electron Microscopy Unit, Institute of Biotechnology, Helsinki Institute of Life Sciences (E.J.), University of Helsinki, Helsinki, Finland
| |
Collapse
|
5
|
Chen Z, Zhang SL. Endoplasmic Reticulum Stress: A Key Regulator of Cardiovascular Disease. DNA Cell Biol 2023. [PMID: 37140435 DOI: 10.1089/dna.2022.0532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2023] Open
Abstract
The problems associated with economic development and social progress have led to an increase in the occurrence of cardiovascular diseases (CVDs), which affect the health of an increasing number of people and are a leading cause of disease and population mortality worldwide. Endoplasmic reticulum stress (ERS), a hot topic of interest for scholars in recent years, has been confirmed in numerous studies to be an important pathogenetic basis for many metabolic diseases and play an important role in maintaining physiological processes. The endoplasmic reticulum (ER) is a major organelle that is involved in protein folding and modification synthesis, and ERS occurs when several physiological and pathological factors allow excessive amounts of unfolded/misfolded proteins to accumulate. ERS often leads to initiation of the unfolded protein response (UPR) in a bid to re-establish tissue homeostasis; however, UPR has been documented to induce vascular remodeling and cardiomyocyte damage under various pathological conditions, leading to or accelerating the development of CVDs such as hypertension, atherosclerosis, and heart failure. In this review, we summarize the latest knowledge gained concerning ERS in terms of cardiovascular system pathophysiology, and discuss the feasibility of targeting ERS as a novel therapeutic target for the treatment of CVDs. Investigation of ERS has immense potential as a new direction for future research involving lifestyle intervention, the use of existing drugs, and the development of novel drugs that target and inhibit ERS.
Collapse
Affiliation(s)
- Zhao Chen
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Shi-Liang Zhang
- Section 4, Department of Cardiology, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
6
|
Chen L, Huan X, Jia F, Zhang Z, Bi M, Fu L, Du X, Chen X, Yan C, Jiao Q, Jiang H. Deubiquitylase OTUD3 Mediates Endoplasmic Reticulum Stress through Regulating Fortilin Stability to Restrain Dopaminergic Neurons Apoptosis. Antioxidants (Basel) 2023; 12:antiox12040809. [PMID: 37107185 PMCID: PMC10135230 DOI: 10.3390/antiox12040809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 03/19/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023] Open
Abstract
OTU domain-containing protein 3 (OTUD3) knockout mice exhibited loss of nigral dopaminergic neurons and Parkinsonian symptoms. However, the underlying mechanisms are largely unknown. In this study, we observed that the inositol-requiring enzyme 1α (IRE1α)-induced endoplasmic reticulum (ER) stress was involved in this process. We found that the ER thickness and the expression of protein disulphide isomerase (PDI) were increased, and the apoptosis level was elevated in the dopaminergic neurons of OTUD3 knockout mice. These phenomena were ameliorated by ER stress inhibitor tauroursodeoxycholic acid (TUDCA) treatment. The ratio of p-IRE1α/IRE1α, and the expression of X-box binding protein 1-spliced (XBP1s) were remarkably increased after OTUD3 knockdown, which was inhibited by IRE1α inhibitor STF-083010 treatment. Moreover, OTUD3 regulated the ubiquitination level of Fortilin through binding with the OTU domain. OTUD3 knockdown resulted in a decrease in the interaction ability of IRE1α with Fortilin and finally enhanced the activity of IRE1α. Taken together, we revealed that OTUD3 knockout-induced injury of dopaminergic neurons might be caused by activating IRE1α signaling in ER stress. These findings demonstrated that OTUD3 played a critical role in dopaminergic neuron neurodegeneration, which provided new evidence for the multiple and tissue-dependent functions of OTUD3.
Collapse
Affiliation(s)
- Ling Chen
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines, Physiology, School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Xuejie Huan
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines, Physiology, School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Fengju Jia
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines, Physiology, School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Zhen Zhang
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines, Physiology, School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Mingxia Bi
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines, Physiology, School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Lin Fu
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines, Physiology, School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Xixun Du
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines, Physiology, School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Xi Chen
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines, Physiology, School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Chunling Yan
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines, Physiology, School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Qian Jiao
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines, Physiology, School of Basic Medicine, Qingdao University, Qingdao 266071, China
- Correspondence: (Q.J.); (H.J.); Tel.: +86-532-8595-0188 (H.J.)
| | - Hong Jiang
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines, Physiology, School of Basic Medicine, Qingdao University, Qingdao 266071, China
- College of Health and Life Science, University of Health and Rehabilitation Sciences, Qingdao 266071, China
- Correspondence: (Q.J.); (H.J.); Tel.: +86-532-8595-0188 (H.J.)
| |
Collapse
|
7
|
Liu J, Maxwell M, Cuddihy T, Crawford T, Bassetti M, Hyde C, Peigneur S, Tytgat J, Undheim EAB, Mobli M. ScrepYard: An online resource for disulfide-stabilized tandem repeat peptides. Protein Sci 2023; 32:e4566. [PMID: 36644825 PMCID: PMC9885460 DOI: 10.1002/pro.4566] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 01/05/2023] [Accepted: 01/12/2023] [Indexed: 01/17/2023]
Abstract
Receptor avidity through multivalency is a highly sought-after property of ligands. While readily available in nature in the form of bivalent antibodies, this property remains challenging to engineer in synthetic molecules. The discovery of several bivalent venom peptides containing two homologous and independently folded domains (in a tandem repeat arrangement) has provided a unique opportunity to better understand the underpinning design of multivalency in multimeric biomolecules, as well as how naturally occurring multivalent ligands can be identified. In previous work, we classified these molecules as a larger class termed secreted cysteine-rich repeat-proteins (SCREPs). Here, we present an online resource; ScrepYard, designed to assist researchers in identification of SCREP sequences of interest and to aid in characterizing this emerging class of biomolecules. Analysis of sequences within the ScrepYard reveals that two-domain tandem repeats constitute the most abundant SCREP domain architecture, while the interdomain "linker" regions connecting the functional domains are found to be abundant in amino acids with short or polar sidechains and contain an unusually high abundance of proline residues. Finally, we demonstrate the utility of ScrepYard as a virtual screening tool for discovery of putatively multivalent peptides, by using it as a resource to identify a previously uncharacterized serine protease inhibitor and confirm its predicted activity using an enzyme assay.
Collapse
Affiliation(s)
- Junyu Liu
- Centre for Advanced ImagingThe University of QueenslandSt. LuciaQueenslandAustralia
| | - Michael Maxwell
- Centre for Advanced ImagingThe University of QueenslandSt. LuciaQueenslandAustralia
| | - Thom Cuddihy
- Queensland Cyber Infrastructure Foundation Ltd.The University of QueenslandSt. LuciaQueenslandAustralia,Centre for Clinical ResearchThe University of QueenslandSt. LuciaQueenslandAustralia
| | - Theo Crawford
- Centre for Advanced ImagingThe University of QueenslandSt. LuciaQueenslandAustralia
| | - Madeline Bassetti
- Queensland Cyber Infrastructure Foundation Ltd.The University of QueenslandSt. LuciaQueenslandAustralia
| | - Cameron Hyde
- Queensland Cyber Infrastructure Foundation Ltd.The University of QueenslandSt. LuciaQueenslandAustralia,University of the Sunshine CoastMaroochydoreQueenslandAustralia
| | - Steve Peigneur
- Toxicology and PharmacologyUniversity of Leuven (KU Leuven)LeuvenBelgium
| | - Jan Tytgat
- Toxicology and PharmacologyUniversity of Leuven (KU Leuven)LeuvenBelgium
| | - Eivind A. B. Undheim
- Centre for Advanced ImagingThe University of QueenslandSt. LuciaQueenslandAustralia,Centre for Ecological and Evolutionary Synthesis, Department of BiosciencesUniversity of OsloOsloNorway
| | - Mehdi Mobli
- Centre for Advanced ImagingThe University of QueenslandSt. LuciaQueenslandAustralia
| |
Collapse
|
8
|
Bertini L, Proietti S, Fongaro B, Holfeld A, Picotti P, Falconieri GS, Bizzarri E, Capaldi G, Polverino de Laureto P, Caruso C. Environmental Signals Act as a Driving Force for Metabolic and Defense Responses in the Antarctic Plant Colobanthus quitensis. PLANTS (BASEL, SWITZERLAND) 2022; 11:3176. [PMID: 36432905 PMCID: PMC9695728 DOI: 10.3390/plants11223176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/14/2022] [Accepted: 11/16/2022] [Indexed: 06/16/2023]
Abstract
During evolution, plants have faced countless stresses of both biotic and abiotic nature developing very effective mechanisms able to perceive and counteract adverse signals. The biggest challenge is the ability to fine-tune the trade-off between plant growth and stress resistance. The Antarctic plant Colobanthus quitensis has managed to survive the adverse environmental conditions of the white continent and can be considered a wonderful example of adaptation to prohibitive conditions for millions of other plant species. Due to the progressive environmental change that the Antarctic Peninsula has undergone over time, a more comprehensive overview of the metabolic features of C. quitensis becomes particularly interesting to assess its ability to respond to environmental stresses. To this end, a differential proteomic approach was used to study the response of C. quitensis to different environmental cues. Many differentially expressed proteins were identified highlighting the rewiring of metabolic pathways as well as defense responses. Finally, a different modulation of oxidative stress response between different environmental sites was observed. The data collected in this paper add knowledge on the impact of environmental stimuli on plant metabolism and stress response by providing useful information on the trade-off between plant growth and defense mechanisms.
Collapse
Affiliation(s)
- Laura Bertini
- Department of Ecological and Biological Sciences, University of Tuscia, 01100 Viterbo, Italy
| | - Silvia Proietti
- Department of Ecological and Biological Sciences, University of Tuscia, 01100 Viterbo, Italy
| | - Benedetta Fongaro
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35100 Padova, Italy
| | - Aleš Holfeld
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland
| | - Paola Picotti
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland
| | | | - Elisabetta Bizzarri
- Department of Ecological and Biological Sciences, University of Tuscia, 01100 Viterbo, Italy
| | - Gloria Capaldi
- Department of Ecological and Biological Sciences, University of Tuscia, 01100 Viterbo, Italy
| | | | - Carla Caruso
- Department of Ecological and Biological Sciences, University of Tuscia, 01100 Viterbo, Italy
| |
Collapse
|
9
|
Yu Y, Yang A, Yu G, Wang H. Endoplasmic Reticulum Stress in Chronic Obstructive Pulmonary Disease: Mechanisms and Future Perspectives. Biomolecules 2022; 12:1637. [PMID: 36358987 PMCID: PMC9687722 DOI: 10.3390/biom12111637] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 10/31/2022] [Accepted: 11/01/2022] [Indexed: 09/08/2024] Open
Abstract
The endoplasmic reticulum (ER) is an integral organelle for maintaining protein homeostasis. Multiple factors can disrupt protein folding in the lumen of the ER, triggering ER stress and activating the unfolded protein response (UPR), which interrelates with various damage mechanisms, such as inflammation, apoptosis, and autophagy. Numerous studies have linked ER stress and UPR to the progression of chronic obstructive pulmonary disease (COPD). This review focuses on the mechanisms of other cellular processes triggered by UPR and summarizes drug intervention strategies targeting the UPR pathway in COPD to explore new therapeutic approaches and preventive measures for COPD.
Collapse
Affiliation(s)
| | | | - Ganggang Yu
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Haoyan Wang
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| |
Collapse
|
10
|
Liu X, Xie X, Li D, Liu Z, Niu Y, Shen B, Zhang B, Song Y, Ma J, Zhang M, Shi Z, Shen C. Transcriptome reveals the dysfunction of pancreatic islets after wound healing in severely burned mice. J Trauma Acute Care Surg 2022; 93:712-718. [PMID: 36301128 DOI: 10.1097/ta.0000000000003697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Severely burned patients have a higher risk of diabetes mellitus after healing, but its mechanism remains unclear. Therefore, the purpose of the study was to explore the influence of burns on pancreatic islets of mice after wound healing. METHODS Forty-two male C57BL/6 mice were randomized into a sham group and a burn group and subjected to sham treatment or a third-degree burn model of 30% total body surface area. Fasting blood glucose was detected weekly for 8 weeks after severe burns. Glucose-stimulated insulin secretion was measured 8 weeks post severe burns. Islets of the two groups were isolated and mRNA libraries were sequenced by the Illumina sequencing platform. The expressions of differentially expressed genes (DEGs) related to the cell cycle and the amounts of mitochondrial DNA were detected by quantitative real-time polymerase chain reaction after gene ontology, gene set enrichment analysis, and protein-protein network analysis. Hematoxylin-eosin staining of pancreatic tail tissue and adenosine triphosphate (ATP) assay of islets were performed. RESULTS The levels of fasting blood glucose were significantly higher within 8 weeks post severe burns. Glucose-stimulated insulin secretion was impaired at the eighth week post severe burns. Totally 128 DEGs were selected. Gene ontology and gene set enrichment analysis indicated that the pathways related to the cell cycle, protein processing, and oxidative phosphorylation were downregulated. The expressions of DEGs related to the cell cycle showed a consistent trend with mRNA sequencing data, and most of them were downregulated post severe burns. The cell mass of the burn group was less than that of the sham group. Also, the concentration of ATP and the amount of mitochondrial DNA were lower in the burn group. CONCLUSION In the model of severe-burned mice, disorders in glucose metabolism persist for 8 weeks after burns, which may be related to low islet cell proliferation, downregulation of protein processing, and less ATP production.
Collapse
Affiliation(s)
- Xinzhu Liu
- From the Department of Burns and Plastic Surgery (X.L., J.M., D.L., Z.L., Y.N., B.S., B.Z., Y.S., M.Z., Z.S., C.S.), the Fourth Medical Center, Chinese PLA General Hospital; and Medical School of Chinese PLA (X.X., X.L., J.M.), Beijing, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Abstract
Peste des petits ruminants virus (PPRV) infection leads to autophagy, and the molecular mechanisms behind this phenomenon are unclear. Here, we demonstrate that PPRV infection results in morphological changes of the endoplasmic reticulum (ER) and activation of activating transcription factor 6 (ATF6) of the ER stress unfolded protein response (UPR). Knockdown of ATF6 blocked the autophagy process, suggesting ATF6 is necessary for PPRV-mediated autophagy induction. Further study showed that PPRV infection upregulates expression of the ER-anchored adaptor protein stimulator of interferon genes (STING), which is well-known for its pivotal roles in restricting DNA viruses. Knockdown of STING suppressed ATF6 activation and autophagy induction, implying that STING functions upstream of ATF6 to induce autophagy. Moreover, the STING-mediated autophagy response originated from the cellular pattern recognition receptor melanoma differentiation-associated gene 5 (MDA5). The absence of MDA5 abolished the upregulation of STING and the activation of autophagy. The deficiency of autophagy-related genes (ATG) repressed the autophagy process and PPRV replication, while it had no effect on MDA5 or STING expression. Overall, our work revealed that MDA5 works upstream of STING to activate ATF6 to induce autophagy. IMPORTANCEPPRV infection induces cellular autophagy; however, the intracellular responses and signaling mechanisms that occur upon PPRV infection are obscure, and whether innate immune responses are linked with autophagy to regulate viral replication is largely unknown. Here, we uncovered that the innate immune sensor MDA5 initiated the signaling cascade by upregulating STING, which is best known for its role in anti-DNA virus infection by inducing interferon expression. We first provide evidence that STING regulates PPRV replication by activating the ATF6 pathway of unfolded protein responses (UPRs) to induce autophagy. Our results revealed that in addition to mediating responses to foreign DNA, STING can cross talk with MDA5 to regulate the cellular stress response and autophagy induced by RNA viruses; thus, STING works as an adaptor protein for cellular stress responses and innate immune responses. Modulation of STING represents a promising approach to control both DNA and RNA viruses.
Collapse
|
12
|
Cheng B, Wang Q, Wei Z, He Y, Li R, Liu G, Zeng S, Meng Z. MHBSt 167 induced autophagy promote cell proliferation and EMT by activating the immune response in L02 cells. Virol J 2022; 19:110. [PMID: 35761331 PMCID: PMC9235077 DOI: 10.1186/s12985-022-01840-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 06/03/2022] [Indexed: 11/13/2022] Open
Abstract
Background Hepatitis B virus can induce hepatocellular carcinoma (HCC) by inducing a host immune response against infected hepatocytes. C-terminally truncated middle surface protein (MHBSt) has been reported to contribute to HCC through transcriptional activation in epidemiology studies, while the underlying mechanism of MHBSt-induced HCC is unknown. Methods In this study, a premature stop at codon 167 in MHBS (MHBSt167) was investigated into eukaryotic expression plasmid pcDNA3.1(-). MHBSt167 expressed plasmid was transfected into the L02 cell line, cell proliferation was analyzed by CCK-8 and high-content screening assays, the cell cycle was analyzed by flow cytometry, and epithelial-to-mesenchymal transition and autophagy were analyzed by immunoblotting and immunofluorescence. NF-κB activation and the MHBSt167-induced immune response were analyzed by immunoblotting and immunofluorescence. IFN-α, IFN-β and IL-1α expression were analyzed by qPCR. Autophagy inhibitors were used to analyze the relationship between the immune response and autophagy. Results The results showed that MHBSt167 promoted L02 cell proliferation, accelerated cell cycle progression from the S to G2 phase and promoted epithelial-to-mesenchymal transition through ER-stress, leading to autophagy and NF-κB activation and increased immune-related factor expression. The MHBSt167-induced acceleration of cell proliferation and the cell cycle was abolished by autophagy or NF-κB inhibitors. Conclusion In summary, MHBSt167 could promote cell proliferation, accelerate cell cycle progression, induce EMT and activate autophagy through ER-stress to induce the host immune response, supporting a potential role of MHBSt167 in contributing to carcinogenesis. Supplementary Information The online version contains supplementary material available at 10.1186/s12985-022-01840-z.
Collapse
Affiliation(s)
- Bin Cheng
- Institute of Biomedical Research, Hubei Clinical Research Center for Precise Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China.,Hubei Key Laboratory of Embryonic Stem Cell Research, Shiyan, 442000, Hubei, China
| | - Qiong Wang
- Institute of Biomedical Research, Hubei Clinical Research Center for Precise Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China.,Department of Infectious Diseases, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Zhiqiang Wei
- Institute of Biomedical Research, Hubei Clinical Research Center for Precise Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China.,Hubei Key Laboratory of Embryonic Stem Cell Research, Shiyan, 442000, Hubei, China
| | - Yulin He
- Institute of Biomedical Research, Hubei Clinical Research Center for Precise Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China.,Hubei Key Laboratory of Embryonic Stem Cell Research, Shiyan, 442000, Hubei, China
| | - Ruiming Li
- Institute of Biomedical Research, Hubei Clinical Research Center for Precise Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China.,Hubei Key Laboratory of Embryonic Stem Cell Research, Shiyan, 442000, Hubei, China
| | - Guohua Liu
- Institute of Biomedical Research, Hubei Clinical Research Center for Precise Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China.,Hubei Key Laboratory of Embryonic Stem Cell Research, Shiyan, 442000, Hubei, China
| | - Shaobo Zeng
- Department of Hepatobiliary Pancreatic Surgery, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Zhongji Meng
- Institute of Biomedical Research, Hubei Clinical Research Center for Precise Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China. .,Department of Infectious Diseases, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China. .,Hubei Key Laboratory of Embryonic Stem Cell Research, Shiyan, 442000, Hubei, China.
| |
Collapse
|
13
|
Yang CY, Liu SH, Su CC, Fang KM, Yang TY, Liu JM, Chen YW, Chang KC, Chuang HL, Wu CT, Lee KI, Huang CF. Methylmercury Induces Mitochondria- and Endoplasmic Reticulum Stress-Dependent Pancreatic β-Cell Apoptosis via an Oxidative Stress-Mediated JNK Signaling Pathway. Int J Mol Sci 2022; 23:2858. [PMID: 35270009 PMCID: PMC8910963 DOI: 10.3390/ijms23052858] [Citation(s) in RCA: 12] [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: 01/05/2022] [Revised: 02/21/2022] [Accepted: 03/01/2022] [Indexed: 12/21/2022] Open
Abstract
Methylmercury (MeHg), a long-lasting organic pollutant, is known to induce cytotoxic effects in mammalian cells. Epidemiological studies have suggested that environmental exposure to MeHg is linked to the development of diabetes mellitus (DM). The exact molecular mechanism of MeHg-induced pancreatic β-cell cytotoxicity is still unclear. Here, we found that MeHg (1-4 μM) significantly decreased insulin secretion and cell viability in pancreatic β-cell-derived RIN-m5F cells. A concomitant elevation of mitochondrial-dependent apoptotic events was observed, including decreased mitochondrial membrane potential and increased proapoptotic (Bax, Bak, p53)/antiapoptotic (Bcl-2) mRNA ratio, cytochrome c release, annexin V-Cy3 binding, caspase-3 activity, and caspase-3/-7/-9 activation. Exposure of RIN-m5F cells to MeHg (2 μM) also induced protein expression of endoplasmic reticulum (ER) stress-related signaling molecules, including C/EBP homologous protein (CHOP), X-box binding protein (XBP-1), and caspase-12. Pretreatment with 4-phenylbutyric acid (4-PBA; an ER stress inhibitor) and specific siRNAs for CHOP and XBP-1 significantly inhibited their expression and caspase-3/-12 activation in MeHg-exposed RIN-mF cells. MeHg could also evoke c-Jun N-terminal kinase (JNK) activation and reactive oxygen species (ROS) generation. Antioxidant N-acetylcysteine (NAC; 1mM) or 6-hydroxy-2,5,7,8-tetramethylchroman-2-carboxylic acid (trolox; 100 μM) markedly prevented MeH-induced ROS generation and decreased cell viability in RIN-m5F cells. Furthermore, pretreatment of cells with SP600125 (JNK inhibitor; 10 μM) or NAC (1 mM) or transfection with JNK-specific siRNA obviously attenuated the MeHg-induced JNK phosphorylation, CHOP and XBP-1 protein expression, apoptotic events, and insulin secretion dysfunction. NAC significantly inhibited MeHg-activated JNK signaling, but SP600125 could not effectively reduce MeHg-induced ROS generation. Collectively, these findings demonstrate that the induction of ROS-activated JNK signaling is a crucial mechanism underlying MeHg-induced mitochondria- and ER stress-dependent apoptosis, ultimately leading to β-cell death.
Collapse
Affiliation(s)
- Ching-Yao Yang
- Department of Surgery, National Taiwan University Hospital, Taipei 100, Taiwan;
- Department of Surgery, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Shing-Hwa Liu
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei 100, Taiwan;
| | - Chin-Chuan Su
- Department of Otorhinolaryngology, Head and Neck Surgery, Changhua Christian Hospital, Changhua County 500, Taiwan;
| | - Kai-Min Fang
- Department of Otolaryngology, Far Eastern Memorial Hospital, New Taipei City 220, Taiwan;
| | - Tsung-Yuan Yang
- Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung 402, Taiwan;
- School of Medicine, Institute of Medicine, Chung-Shan Medical University, Taichung 402, Taiwan
| | - Jui-Ming Liu
- Department of Urology, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan 330, Taiwan;
- Department of Obstetrics and Gynecology, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Ya-Wen Chen
- Department of Physiology, School of Medicine, College of Medicine, China Medical University, Taichung 404, Taiwan;
| | - Kai-Chih Chang
- Center for Digestive Medicine, Department of Internal Medicine, China Medical University Hospital, Taichung 404, Taiwan;
| | - Haw-Ling Chuang
- Department of Emergency, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung 427, Taiwan;
| | - Cheng-Tien Wu
- Department of Nutrition and Master Program of Food and Drug Safety, China Medical University, Taichung 40402, Taiwan;
| | - Kuan-I Lee
- Department of Emergency, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung 427, Taiwan;
| | - Chun-Fa Huang
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung 404, Taiwan
- Department of Nursing, College of Medical and Health Science, Asia University, Taichung 413, Taiwan
| |
Collapse
|
14
|
Sahin GS, Lee H, Engin F. An accomplice more than a mere victim: The impact of β-cell ER stress on type 1 diabetes pathogenesis. Mol Metab 2021; 54:101365. [PMID: 34728341 PMCID: PMC8606542 DOI: 10.1016/j.molmet.2021.101365] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/23/2021] [Accepted: 10/26/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Pancreatic β-cells are the insulin factory of an organism with a mission to regulate glucose homeostasis in the body. Due to their high secretory activity, β-cells rely on a functional and intact endoplasmic reticulum (ER). Perturbations to ER homeostasis and unmitigated stress lead to β-cell dysfunction and death. Type 1 diabetes (T1D) is a chronic inflammatory disease caused by the autoimmune-mediated destruction of β-cells. Although autoimmunity is an essential component of T1D pathogenesis, accumulating evidence suggests an important role of β-cell ER stress and aberrant unfolded protein response (UPR) in disease initiation and progression. SCOPE OF REVIEW In this article, we introduce ER stress and the UPR, review β-cell ER stress in various mouse models, evaluate its involvement in inflammation, and discuss the effects of ER stress on β-cell plasticity and demise, and islet autoimmunity in T1D. We also highlight the relationship of ER stress with other stress response pathways and provide insight into ongoing clinical studies targeting ER stress and the UPR for the prevention or treatment of T1D. MAJOR CONCLUSIONS Evidence from ex vivo studies, in vivo mouse models, and tissue samples from patients suggest that β-cell ER stress and a defective UPR contribute to T1D pathogenesis. Thus, restoration of β-cell ER homeostasis at various stages of disease presents a plausible therapeutic strategy for T1D. Identifying the specific functions and regulation of each UPR sensor in β-cells and uncovering the crosstalk between stressed β-cells and immune cells during T1D progression would provide a better understanding of the molecular mechanisms of disease process, and may reveal novel targets for development of effective therapies for T1D.
Collapse
Affiliation(s)
- Gulcan Semra Sahin
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, 53706, USA
| | - Hugo Lee
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, 53706, USA
| | - Feyza Engin
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, 53706, USA; Department of Medicine, Division of Endocrinology, Diabetes & Metabolism, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, 53705, USA; Department of Cell & Regenerative Biology, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, 53705, USA.
| |
Collapse
|
15
|
Pick T, Beck A, Gamayun I, Schwarz Y, Schirra C, Jung M, Krause E, Niemeyer BA, Zimmermann R, Lang S, Anken EV, Cavalié A. Remodelling of Ca 2+ homeostasis is linked to enlarged endoplasmic reticulum in secretory cells. Cell Calcium 2021; 99:102473. [PMID: 34560367 DOI: 10.1016/j.ceca.2021.102473] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 08/27/2021] [Accepted: 09/08/2021] [Indexed: 11/30/2022]
Abstract
The endoplasmic reticulum (ER) is extensively remodelled during the development of professional secretory cells to cope with high protein production. Since ER is the principal Ca2+ store in the cell, we characterised the Ca2+ homeostasis in NALM-6 and RPMI 8226 cells, which are commonly used as human pre-B and antibody secreting plasma cell models, respectively. Expression levels of Sec61 translocons and the corresponding Sec61-mediated Ca2+ leak from ER, Ca2+ storage capacity and store-operated Ca2+ entry were significantly enlarged in the secretory RPMI 8226 cell line. Using an immunoglobulin M heavy chain producing HeLa cell model, we found that the enlarged Ca2+ storage capacity and Ca2+ leak from ER are linked to ER expansion. Our data delineates a developmental remodelling of Ca2+ homeostasis in professional secretory cells in which a high Sec61-mediated Ca2+ leak and, thus, a high Ca2+ turnover in the ER is backed up by enhanced store-operated Ca2+ entry.
Collapse
Affiliation(s)
- Tillman Pick
- Experimental and Clinical Pharmacology and Toxicology, Pre-clinical Center for Molecular Signalling (PZMS), Saarland University, 66421 Homburg, Germany.
| | - Andreas Beck
- Experimental and Clinical Pharmacology and Toxicology, Pre-clinical Center for Molecular Signalling (PZMS), Saarland University, 66421 Homburg, Germany
| | - Igor Gamayun
- Experimental and Clinical Pharmacology and Toxicology, Pre-clinical Center for Molecular Signalling (PZMS), Saarland University, 66421 Homburg, Germany
| | - Yvonne Schwarz
- Molecular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University, 66421 Homburg, Germany
| | - Claudia Schirra
- Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University, 66421 Homburg, Germany
| | - Martin Jung
- Medical Biochemistry and Molecular Biology, Pre-clinical Centre for Molecular Signalling (PZMS), Saarland University, 66421 Homburg, Germany
| | - Elmar Krause
- Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University, 66421 Homburg, Germany
| | - Barbara A Niemeyer
- Molecular Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University, 66421 Homburg, Germany
| | - Richard Zimmermann
- Medical Biochemistry and Molecular Biology, Pre-clinical Centre for Molecular Signalling (PZMS), Saarland University, 66421 Homburg, Germany
| | - Sven Lang
- Medical Biochemistry and Molecular Biology, Pre-clinical Centre for Molecular Signalling (PZMS), Saarland University, 66421 Homburg, Germany
| | - Eelco van Anken
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute and Università Vita-Salute San Raffaele, Milan, Italy
| | - Adolfo Cavalié
- Experimental and Clinical Pharmacology and Toxicology, Pre-clinical Center for Molecular Signalling (PZMS), Saarland University, 66421 Homburg, Germany.
| |
Collapse
|
16
|
Roboti P, O'Keefe S, Duah KB, Shi WQ, High S. Ipomoeassin-F disrupts multiple aspects of secretory protein biogenesis. Sci Rep 2021; 11:11562. [PMID: 34079010 PMCID: PMC8173012 DOI: 10.1038/s41598-021-91107-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/11/2021] [Indexed: 12/12/2022] Open
Abstract
The Sec61 complex translocates nascent polypeptides into and across the membrane of the endoplasmic reticulum (ER), providing access to the secretory pathway. In this study, we show that Ipomoeassin-F (Ipom-F), a selective inhibitor of protein entry into the ER lumen, blocks the in vitro translocation of certain secretory proteins and ER lumenal folding factors whilst barely affecting others such as albumin. The effects of Ipom-F on protein secretion from HepG2 cells are twofold: reduced ER translocation combined, in some cases, with defective ER lumenal folding. This latter issue is most likely a consequence of Ipom-F preventing the cell from replenishing its ER lumenal chaperones. Ipom-F treatment results in two cellular stress responses: firstly, an upregulation of stress-inducible cytosolic chaperones, Hsp70 and Hsp90; secondly, an atypical unfolded protein response (UPR) linked to the Ipom-F-mediated perturbation of ER function. Hence, although levels of spliced XBP1 and CHOP mRNA and ATF4 protein increase with Ipom-F, the accompanying increase in the levels of ER lumenal BiP and GRP94 seen with tunicamycin are not observed. In short, although Ipom-F reduces the biosynthetic load of newly synthesised secretory proteins entering the ER lumen, its effects on the UPR preclude the cell restoring ER homeostasis.
Collapse
Affiliation(s)
- Peristera Roboti
- Division of Molecular and Cellular Function, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK.
| | - Sarah O'Keefe
- Division of Molecular and Cellular Function, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK
| | - Kwabena B Duah
- Department of Chemistry, Ball State University, Muncie, IN, 47306, USA
| | - Wei Q Shi
- Department of Chemistry, Ball State University, Muncie, IN, 47306, USA
| | - Stephen High
- Division of Molecular and Cellular Function, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK.
| |
Collapse
|
17
|
Burillo J, Marqués P, Jiménez B, González-Blanco C, Benito M, Guillén C. Insulin Resistance and Diabetes Mellitus in Alzheimer's Disease. Cells 2021; 10:1236. [PMID: 34069890 PMCID: PMC8157600 DOI: 10.3390/cells10051236] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/11/2021] [Accepted: 05/13/2021] [Indexed: 12/12/2022] Open
Abstract
Type 2 diabetes mellitus is a progressive disease that is characterized by the appearance of insulin resistance. The term insulin resistance is very wide and could affect different proteins involved in insulin signaling, as well as other mechanisms. In this review, we have analyzed the main molecular mechanisms that could be involved in the connection between type 2 diabetes and neurodegeneration, in general, and more specifically with the appearance of Alzheimer's disease. We have studied, in more detail, the different processes involved, such as inflammation, endoplasmic reticulum stress, autophagy, and mitochondrial dysfunction.
Collapse
Affiliation(s)
- Jesús Burillo
- Department of Biochemistry, Complutense University, 28040 Madrid, Spain; (J.B.); (P.M.); (B.J.); (C.G.-B.); (M.B.)
- Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28040 Madrid, Spain
- Mechanisms of Insulin Resistance (MOIR2), General Direction of Universities and Investigation (CCMM), 28040 Madrid, Spain
| | - Patricia Marqués
- Department of Biochemistry, Complutense University, 28040 Madrid, Spain; (J.B.); (P.M.); (B.J.); (C.G.-B.); (M.B.)
- Mechanisms of Insulin Resistance (MOIR2), General Direction of Universities and Investigation (CCMM), 28040 Madrid, Spain
| | - Beatriz Jiménez
- Department of Biochemistry, Complutense University, 28040 Madrid, Spain; (J.B.); (P.M.); (B.J.); (C.G.-B.); (M.B.)
- Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28040 Madrid, Spain
- Mechanisms of Insulin Resistance (MOIR2), General Direction of Universities and Investigation (CCMM), 28040 Madrid, Spain
| | - Carlos González-Blanco
- Department of Biochemistry, Complutense University, 28040 Madrid, Spain; (J.B.); (P.M.); (B.J.); (C.G.-B.); (M.B.)
- Mechanisms of Insulin Resistance (MOIR2), General Direction of Universities and Investigation (CCMM), 28040 Madrid, Spain
| | - Manuel Benito
- Department of Biochemistry, Complutense University, 28040 Madrid, Spain; (J.B.); (P.M.); (B.J.); (C.G.-B.); (M.B.)
- Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28040 Madrid, Spain
- Mechanisms of Insulin Resistance (MOIR2), General Direction of Universities and Investigation (CCMM), 28040 Madrid, Spain
| | - Carlos Guillén
- Department of Biochemistry, Complutense University, 28040 Madrid, Spain; (J.B.); (P.M.); (B.J.); (C.G.-B.); (M.B.)
- Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28040 Madrid, Spain
- Mechanisms of Insulin Resistance (MOIR2), General Direction of Universities and Investigation (CCMM), 28040 Madrid, Spain
| |
Collapse
|
18
|
van Anken E, Bakunts A, Hu CCA, Janssens S, Sitia R. Molecular Evaluation of Endoplasmic Reticulum Homeostasis Meets Humoral Immunity. Trends Cell Biol 2021; 31:529-541. [PMID: 33685797 DOI: 10.1016/j.tcb.2021.02.004] [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: 11/27/2020] [Revised: 02/02/2021] [Accepted: 02/03/2021] [Indexed: 12/16/2022]
Abstract
The biosynthesis of about one third of the human proteome, including membrane receptors and secreted proteins, occurs in the endoplasmic reticulum (ER). Conditions that perturb ER homeostasis activate the unfolded protein response (UPR). An 'optimistic' UPR output aims at restoring homeostasis by reinforcement of machineries that guarantee efficiency and fidelity of protein biogenesis in the ER. Yet, once the UPR 'deems' that ER homeostatic readjustment fails, it transitions to a 'pessimistic' output, which, depending on the cell type, will result in apoptosis. In this article, we discuss emerging concepts on how the UPR 'evaluates' ER stress, how the UPR is repurposed, in particular in B cells, and how UPR-driven counter-selection of cells undergoing homeostatic failure serves organismal homeostasis and humoral immunity.
Collapse
Affiliation(s)
- Eelco van Anken
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy.
| | - Anush Bakunts
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | | | - Sophie Janssens
- Laboratory for Endoplasmic Reticulum (ER) Stress and Inflammation, VIB Center for Inflammation Research, and Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Roberto Sitia
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
19
|
Jin N, Jin N, Wang Z, Liu L, Meng L, Li D, Li X, Zhou D, Liu J, Bu W, Sun H, Yang B. Osteopromotive carbon dots promote bone regeneration through the PERK-eIF2α-ATF4 pathway. Biomater Sci 2021; 8:2840-2852. [PMID: 32307492 DOI: 10.1039/d0bm00424c] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Bone defects are still an unsolved clinical issue that must be overcome. Carbon dots have shown very promising effects in biological therapy. In the current study, we explored their effects on osteogenesis. Furthermore, we revealed the mechanisms in order to develop novel therapeutic approaches to manage the bone defect. For this study, ascorbic acid carbon dots (CDs) were created by a one-step microwave-assisted method. Results showed that the CDs effectively enhanced matrix mineralization, promoted osteogenic differentiation in vitro, and promoted new bone regeneration in the skull defect model in vivo. Furthermore, our data demonstrated that the ER stress and PERK-eIF2α-ATF4 pathway were activated by the CD-induced increase in intracellular calcium. Taken together, our findings suggest that the PERK pathway plays a critical role in CD-induced osteogenic differentiation, and the CDs created herein have the potential to be used to repair bone defects in clinical practice.
Collapse
Affiliation(s)
- Nianqiang Jin
- Department of Oral Pathology, School and Hospital of Stomatology, China Medical University, 110001, Shenyang, China.
| | - Nuo Jin
- Department of Oral Pathology, School and Hospital of Stomatology, China Medical University, 110001, Shenyang, China.
| | - Zilin Wang
- Department of Oral Pathology, School and Hospital of Stomatology, Jilin University, 130000, Changchun, China.
| | - Lili Liu
- Department of Oral Pathology, School and Hospital of Stomatology, Jilin University, 130000, Changchun, China.
| | - Lin Meng
- Department of Oral Pathology, School and Hospital of Stomatology, Jilin University, 130000, Changchun, China.
| | - Daowei Li
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School of Stomatology, Jilin University, Changchun, 130021, China.
| | - Xing Li
- Department of Oral Pathology, School and Hospital of Stomatology, China Medical University, 110001, Shenyang, China.
| | - Dabo Zhou
- School and Hospital of Stomatology, China Medical University, 117 Nanjing North Street, Shenyang, 110001, China.
| | - Jie Liu
- Department of Head and Neck Tumor Surgery, School of Stomatology, Wuhan University, Wuhan, 430000, China.
| | - Wenhuan Bu
- Department of Dental Materials, School of Stomatology, China Medical University, Shenyang 110001, China. and Department of Center Laboratory, School of Stomatology, China Medical University, Shenyang 110001, China
| | - Hongchen Sun
- Department of Oral Pathology, School and Hospital of Stomatology, China Medical University, 110001, Shenyang, China.
| | - Bai Yang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, 130012, Changchun, China
| |
Collapse
|
20
|
Devi S, Kim JJ, Singh AP, Kumar S, Dubey AK, Singh SK, Singh RS, Kumar V. Proteotoxicity: A Fatal Consequence of Environmental Pollutants-Induced Impairments in Protein Clearance Machinery. J Pers Med 2021; 11:69. [PMID: 33503824 PMCID: PMC7912547 DOI: 10.3390/jpm11020069] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/20/2021] [Accepted: 01/22/2021] [Indexed: 02/08/2023] Open
Abstract
A tightly regulated protein quality control (PQC) system maintains a healthy balance between correctly folded and misfolded protein species. This PQC system work with the help of a complex network comprised of molecular chaperones and proteostasis. Any intruder, especially environmental pollutants, disrupt the PQC network and lead to PQCs disruption, thus generating damaged and infectious protein. These misfolded/unfolded proteins are linked to several diseases such as Parkinson's disease, Alzheimer's disease, Huntington's disease, and cataracts. Numerous studies on proteins misfolding and disruption of PQCs by environmental pollutants highlight the necessity of detailed knowledge. This review represents the PQCs network and environmental pollutants' impact on the PQC network, especially through the protein clearance system.
Collapse
Affiliation(s)
- Shweta Devi
- Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research, Lucknow 226001, India;
| | - Jong-Joo Kim
- Department of Biotechnology, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Korea;
| | - Anand Prakash Singh
- Division of Cardiovascular Disease, The University of Alabama at Birmingham (UAB), 1720 2nd Ave South, Birmingham, AL 35294-1913, USA;
| | - Surendra Kumar
- Cytogenetics Lab, Department of Anatomy, All India Institute of Medical Sciences, New Delhi 110029, India;
| | | | | | - Ravi Shankar Singh
- Department of Biochemistry, Microbiology & Immunology, University of Saskatchewan, Room 4D40, Health Sciences Building, 107 Wiggins Road, Saskatoon, SK S7N 5E5, Canada
| | - Vijay Kumar
- Department of Biotechnology, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Korea;
| |
Collapse
|
21
|
Eastman AJ, Moore RE, Townsend SD, Gaddy JA, Aronoff DM. The Influence of Obesity and Associated Fatty Acids on Placental Inflammation. Clin Ther 2021; 43:265-278. [PMID: 33487441 DOI: 10.1016/j.clinthera.2020.12.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/24/2020] [Accepted: 12/31/2020] [Indexed: 12/11/2022]
Abstract
PURPOSE Maternal obesity, affecting nearly 1 in 4 pregnancies, is associated with increased circulating saturated fatty acids, such as palmitate. These fatty acids are implicated in placental inflammation, which may in turn exacerbate both maternal-fetal tolerance and responses to pathogens, such as group B Streptococcus. In this review, we address the question, "How do obesity and associated fatty acids influence placental inflammation?" METHODS In this narrative review, we searched PubMed and Google Scholar using combinations of the key words placental inflammation or pregnancy and lipids, fatty acids, obesity, palmitate, or other closely related search terms. We also used references found within these articles that may have been absent from our original search queries. We analyzed methods and key results of these articles to compare and contrast their findings, which were occasionally at odds with each other. FINDINGS Although obesity can be studied as a whole, complex phenomena with in vivo mouse models and human samples from patients with obesity, in vitro modeling often relies on the treatment of cells or tissues with ≥1 fatty acids and occasionally other compounds (eg, glucose and insulin). We found that palmitate, most commonly used in vitro to recreate hallmarks of obesity, induces apoptosis, oxidative stress, mitochondrial dysfunction, autophagy defects, and inflammasome activation in many placental cell types. We compare this to in vivo models of obesity wherever possible. We found that obesity as a whole may have more complex regulation of these phenomena (apoptosis, oxidative stress, mitochondrial dysfunction, autophagy defects, and inflammasome activation) compared with in vitro models of fatty acid treatment (primarily palmitate) because of the presence of unsaturated fatty acids (ie, oleate), which may have anti-inflammatory effects. IMPLICATIONS The interaction of unsaturated fatty acids with saturated fatty acids may ameliorate many inflammatory effects of saturated fatty acids alone, which complicates interpretation of in vitro studies that focus on a particular fatty acid in isolation. This complication may explain why certain studies of obesity in vivo have differing outcomes from studies of specific fatty acids in vitro.
Collapse
Affiliation(s)
- Alison J Eastman
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Rebecca E Moore
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA
| | | | - Jennifer A Gaddy
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA; Tennessee Valley Healthcare Systems, Department of Veterans Affairs, Nashville, TN, USA
| | - David M Aronoff
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
22
|
Koksal AR, Verne GN, Zhou Q. Endoplasmic reticulum stress in biological processing and disease. J Investig Med 2021; 69:309-315. [PMID: 33472886 DOI: 10.1136/jim-2020-001570] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/13/2020] [Indexed: 12/12/2022]
Abstract
The ability of translated cellular proteins to perform their functions requires their proper folding after synthesis. The endoplasmic reticulum (ER) is responsible for coordinating protein folding and maturation. Infections, genetic mutations, environmental factors and many other conditions can lead to challenges to the ER known as ER stress. Altering ER homeostasis results in accumulation of misfolded or unfolded proteins. To eliminate this problem, a response is initiated by the cell called the unfolded protein response (UPR), which involves multiple signaling pathways. Prolonged ER stress or a dysregulated UPR can lead to premature apoptosis and an exaggerated inflammatory response. Following these discoveries, ER stress was shown to be related to several chronic diseases, such as diabetes mellitus, neurodegenerative disorders, fatty liver disease and inflammatory bowel disease that have not yet been clearly demonstrated pathophysiologically. Here, we review the field and present up-to-date information on the relationship between biological processing, ER stress, UPR, and several chronic diseases.
Collapse
Affiliation(s)
- Ali Riza Koksal
- Gastroenterology & Hepatology, Tulane University School of Medicine, New Orleans, Lousiana, USA
| | | | - QiQi Zhou
- Medicine, Division of Gastroenterology, UTHSC COM, Memphis, Tennessee, USA
| |
Collapse
|
23
|
Li X, Sun S, Appathurai S, Sundaram A, Plumb R, Mariappan M. A Molecular Mechanism for Turning Off IRE1α Signaling during Endoplasmic Reticulum Stress. Cell Rep 2020; 33:108563. [PMID: 33378667 PMCID: PMC7809255 DOI: 10.1016/j.celrep.2020.108563] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 09/14/2020] [Accepted: 12/06/2020] [Indexed: 12/15/2022] Open
Abstract
Misfolded proteins in the endoplasmic reticulum (ER) activate IRE1α endoribonuclease in mammalian cells, which mediates XBP1 mRNA splicing to produce an active transcription factor. This promotes the expression of specific genes to alleviate ER stress, thereby attenuating IRE1α. Although sustained activation of IRE1α is linked to human diseases, it is not clear how IRE1α is attenuated during ER stress. Here, we identify that Sec63 is a subunit of the previously identified IRE1α/Sec61 translocon complex. We find that Sec63 recruits and activates BiP ATPase through its luminal J-domain to bind onto IRE1α. This leads to inhibition of higher-order oligomerization and attenuation of IRE1α RNase activity during prolonged ER stress. In Sec63-deficient cells, IRE1α remains activated for a long period of time despite the presence of excess BiP in the ER. Thus, our data suggest that the Sec61 translocon bridges IRE1α with Sec63/BiP to regulate the dynamics of IRE1α signaling in cells. The stress sensor IRE1α is attenuated during prolonged ER stress by a poorly understood mechanism. Li et al. show that IRE1α forms a complex with the Sec61/Sec63 translocon in cells. Sec63 mediates BiP binding to IRE1α and thereby inhibits IRE1α oligomerization and attenuates IRE1α signaling during prolonged ER stress.
Collapse
Affiliation(s)
- Xia Li
- Department of Cell Biology, Nanobiology Institute, Yale School of Medicine, Yale West Campus, West Haven, CT 06516, USA
| | - Sha Sun
- Department of Cell Biology, Nanobiology Institute, Yale School of Medicine, Yale West Campus, West Haven, CT 06516, USA
| | - Suhila Appathurai
- Department of Cell Biology, Nanobiology Institute, Yale School of Medicine, Yale West Campus, West Haven, CT 06516, USA
| | - Arunkumar Sundaram
- Department of Cell Biology, Nanobiology Institute, Yale School of Medicine, Yale West Campus, West Haven, CT 06516, USA
| | - Rachel Plumb
- Department of Cell Biology, Nanobiology Institute, Yale School of Medicine, Yale West Campus, West Haven, CT 06516, USA
| | - Malaiyalam Mariappan
- Department of Cell Biology, Nanobiology Institute, Yale School of Medicine, Yale West Campus, West Haven, CT 06516, USA.
| |
Collapse
|
24
|
Zhang J, Ye ZW, Chen W, Culpepper J, Jiang H, Ball LE, Mehrotra S, Blumental-Perry A, Tew KD, Townsend DM. Altered redox regulation and S-glutathionylation of BiP contribute to bortezomib resistance in multiple myeloma. Free Radic Biol Med 2020; 160:755-767. [PMID: 32937189 PMCID: PMC7704679 DOI: 10.1016/j.freeradbiomed.2020.09.013] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 09/10/2020] [Indexed: 12/13/2022]
Abstract
Multiple myeloma (MM) cells have high rates of secretion of proteins rich in disulfide bonds and depend upon compartmentalized redox balance for accurate protein folding. The proteasome inhibitor bortezomib (Btz) is a successful frontline treatment for the disease, but its long-term efficacy is restricted by the acquisition of resistance. We found that MM cell lines resistant to Btz maintain high levels of oxidative stress and are cross resistant to endoplasmic reticulum (ER) stress-inducing agents thapsigargin (ThG), and tunicamycin (TuM). Moreover, cells expressing high/wild type levels of glutathione S-transferase P (GSTP) are more resistant than Gstp1/p2 knockout cells. In agreement, basal levels of S-glutathionylated proteins and redox regulation enzymes, including GSTP are elevated at mRNA and protein levels in resistant cells. GSTP mediated S-glutathionylation (SSG) regulates the activities of a number of redox active ER proteins. Here we demonstrated that the post-translational modification determines the balance between foldase and ATPase activities of the binding immunoglobulin protein (BiP), with Cys41-SSG important for ATPase, and Cys420-SSG for foldase. BiP expression and S-glutathionylation are increased in clinical specimens of bone marrow from MM patients compared to non-cancerous samples. Preventing S-glutathionylation in MM cells with a GSTP specific inhibitor restored BiP activities and reversed resistance to Btz. Therefore, S-glutathionylation of BiP confers pro-survival advantages and represents a novel mechanism of drug resistance in MM cells. We conclude that altered GSTP expression leads to S-glutathionylation of BiP, and contributes to acquired resistance to Btz in MM.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, 173 Ashley Avenue, MSC 509/BSB 358, Charleston, SC, 29425, USA.
| | - Zhi-Wei Ye
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, 173 Ashley Avenue, MSC 509/BSB 358, Charleston, SC, 29425, USA
| | - Wei Chen
- Clinical Research Center, the Second Hospital of Nanjing, Nanjing University of Chinese Medicine, 1-1 Zhongfu Road, Nangjing, 21003, China
| | - John Culpepper
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, 173 Ashley Avenue, MSC 509/BSB 358, Charleston, SC, 29425, USA
| | - Haiming Jiang
- Intensive Care Unit, Yantai Affiliated Hospital of Binzhou Medical University, No. 717, Jinbu Road, Muping District, Yantai City, Shandong, 264100, PR China
| | - Lauren E Ball
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, 173 Ashley Avenue, MSC 509/BSB 358, Charleston, SC, 29425, USA
| | - Shikhar Mehrotra
- Department of Surgery, Medical University of South Carolina, 86 Jonathan Lucas Street, HCC512H, Charleston, SC, 29425, USA
| | - Anna Blumental-Perry
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, 14203, USA
| | - Kenneth D Tew
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, 173 Ashley Avenue, MSC 509/BSB 358, Charleston, SC, 29425, USA
| | - Danyelle M Townsend
- Department of Pharmaceutical and Biomedical Sciences, Medical University of South Carolina, 274 Calhoun Street, MSC 141, Charleston, SC, 29425, USA.
| |
Collapse
|
25
|
da Costa CA, Manaa WE, Duplan E, Checler F. The Endoplasmic Reticulum Stress/Unfolded Protein Response and Their Contributions to Parkinson's Disease Physiopathology. Cells 2020; 9:cells9112495. [PMID: 33212954 PMCID: PMC7698446 DOI: 10.3390/cells9112495] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/13/2020] [Accepted: 11/16/2020] [Indexed: 12/11/2022] Open
Abstract
Parkinson’s disease (PD) is a multifactorial age-related movement disorder in which defects of both mitochondria and the endoplasmic reticulum (ER) have been reported. The unfolded protein response (UPR) has emerged as a key cellular dysfunction associated with the etiology of the disease. The UPR involves a coordinated response initiated in the endoplasmic reticulum that grants the correct folding of proteins. This review gives insights on the ER and its functioning; the UPR signaling cascades; and the link between ER stress, UPR activation, and physiopathology of PD. Thus, post-mortem studies and data obtained by either in vitro and in vivo pharmacological approaches or by genetic modulation of PD causative genes are described. Further, we discuss the relevance and impact of the UPR to sporadic and genetic PD pathology.
Collapse
|
26
|
Bhardwaj M, Leli NM, Koumenis C, Amaravadi RK. Regulation of autophagy by canonical and non-canonical ER stress responses. Semin Cancer Biol 2020; 66:116-128. [PMID: 31838023 PMCID: PMC7325862 DOI: 10.1016/j.semcancer.2019.11.007] [Citation(s) in RCA: 137] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 11/05/2019] [Accepted: 11/26/2019] [Indexed: 12/12/2022]
Abstract
Cancer cells encounter numerous stresses that pose a threat to their survival. Tumor microenviroment stresses that perturb protein homeostasis can produce endoplasmic reticulum (ER) stress, which can be counterbalanced by triggering the unfolded protein response (UPR) which is considered the canonical ER stress response. The UPR is characterized by three major proteins that lead to specific changes in transcriptional and translational programs in stressed cells. Activation of the UPR can induce apoptosis, but also can induce cytoprotective programs such as autophagy. There is increasing appreciation for the role that UPR-induced autophagy plays in supporting tumorigenesis and cancer therapy resistance. More recently several new pathways that connect cell stresses, components of the UPR and autophagy have been reported, which together can be viewed as non-canonical ER stress responses. Here we review recent findings on the molecular mechanisms by which canonical and non-canonical ER stress responses can activate cytoprotective autophagy and contribute to tumor growth and therapy resistance. Autophagy has been identified as a druggable pathway, however the components of autophagy (ATG genes) have proven difficult to drug. It may be the case that targeting the UPR or non-canonical ER stress programs can more effectively block cytoprotective autophagy to enhance cancer therapy. A deeper understanding of these pathways could provide new therapeutic targets in cancer.
Collapse
Affiliation(s)
- Monika Bhardwaj
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Nektaria Maria Leli
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Constantinos Koumenis
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, 19104, USA; Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Ravi K Amaravadi
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA; Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
27
|
Kalmankar NV, Venkatesan R, Balaram P, Sowdhamini R. Transcriptomic profiling of the medicinal plant Clitoria ternatea: identification of potential genes in cyclotide biosynthesis. Sci Rep 2020; 10:12658. [PMID: 32728092 PMCID: PMC7391643 DOI: 10.1038/s41598-020-69452-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 07/10/2020] [Indexed: 01/20/2023] Open
Abstract
Clitoria ternatea a perennial climber of the Fabaceae family, is well known for its agricultural and medical applications. It is also currently the only known member of the Fabaceae family that produces abundant amounts of the ultra-stable macrocyclic peptides, cyclotides, across all tissues. Cyclotides are a class of gene-encoded, disulphide-rich, macrocyclic peptides (26–37 residues) acting as defensive metabolites in several plant species. Previous transcriptomic studies have demonstrated the genetic origin of cyclotides from the Fabaceae plant family to be embedded in the albumin-1 genes, unlike its counterparts in other plant families. However, the complete mechanism of its biosynthesis and the repertoire of enzymes involved in cyclotide folding and processing remains to be understood. In this study, using RNA-Seq data and de novo transcriptome assembly of Clitoria ternatea, we have identified 71 precursor genes of cyclotides. Out of 71 unique cyclotide precursor genes obtained, 51 sequences display unique cyclotide domains, of which 26 are novel cyclotide sequences, arising from four individual tissues. MALDI-TOF mass spectrometry analysis of fractions from different tissue extracts, coupled with precursor protein sequences obtained from transcriptomic data, established the cyclotide diversity in this plant species. Special focus in this study has also been on identifying possible enzymes responsible for proper folding and processing of cyclotides in the cell. Transcriptomic mining for oxidative folding enzymes such as protein-disulphide isomerases (PDI), ER oxidoreductin-1 (ERO1) and peptidylprolyl cis-trans isomerases (PPIases)/cyclophilins, and their levels of expression are also reported. In particular, it was observed that the CtPDI genes formed plant-specific clusters among PDI genes as compared to those from other plant species. Collectively, this work provides insights into the biogenesis of the medicinally important cyclotides and establishes the expression of certain key enzymes participating in peptide biosynthesis. Also, several novel cyclotide sequences are reported and precursor sequences are analysed in detail. In the absence of a published reference genome, a comprehensive transcriptomics approach was adopted to provide an overview of diverse properties and constituents of C. ternatea.
Collapse
Affiliation(s)
- Neha V Kalmankar
- National Centre for Biological Sciences (TIFR), GKVK Campus, Bangalore, Karnataka, 560065, India.,The University of Trans-Disciplinary Health Sciences and Technology (TDU), #74/2, Jarakabande Kaval, Post Attur, Via Yelahanka, Bangalore, Karnataka, 560064, India
| | - Radhika Venkatesan
- National Centre for Biological Sciences (TIFR), GKVK Campus, Bangalore, Karnataka, 560065, India.,Department of Biological Sciences, Indian Institute of Science, Education and Research, Kolkata, Mohanpur, West Bengal, 741246, India
| | - Padmanabhan Balaram
- National Centre for Biological Sciences (TIFR), GKVK Campus, Bangalore, Karnataka, 560065, India.,Molecular Biophysics Unit, Indian Institute of Science, Bangalore, Karnataka, 560012, India
| | - Ramanathan Sowdhamini
- National Centre for Biological Sciences (TIFR), GKVK Campus, Bangalore, Karnataka, 560065, India.
| |
Collapse
|
28
|
Endoplasmic reticulum stress, degeneration of pancreatic islet β-cells, and therapeutic modulation of the unfolded protein response in diabetes. Mol Metab 2020; 27S:S60-S68. [PMID: 31500832 PMCID: PMC6768499 DOI: 10.1016/j.molmet.2019.06.012] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Background Myriad challenges to the proper folding and structural maturation of secretory pathway client proteins in the endoplasmic reticulum (ER) — a condition referred to as “ER stress” — activate intracellular signaling pathways termed the unfolded protein response (UPR). Scope of review Through executing transcriptional and translational programs the UPR restores homeostasis in those cells experiencing manageable levels of ER stress. But the UPR also actively triggers cell degeneration and apoptosis in those cells that are encountering ER stress levels that exceed irremediable thresholds. Thus, UPR outputs are “double-edged”. In pancreatic islet β-cells, numerous genetic mutations affecting the balance between these opposing UPR functions cause diabetes mellitus in both rodents and humans, amply demonstrating the principle that the UPR is critical for the proper functioning and survival of the cell. Major conclusions Specifically, we have found that the UPR master regulator IRE1α kinase/endoribonuclease (RNase) triggers apoptosis, β-cell degeneration, and diabetes, when ER stress reaches critical levels. Based on these mechanistic findings, we find that novel small molecule compounds that inhibit IRE1α during such “terminal” UPR signaling can spare ER stressed β-cells from death, perhaps affording future opportunities to test new drug candidates for disease modification in patients suffering from diabetes.
Collapse
|
29
|
Aulestia FJ, Groeling J, Bomfim GHS, Costiniti V, Manikandan V, Chaloemtoem A, Concepcion AR, Li Y, Wagner LE, Idaghdour Y, Yule DI, Lacruz RS. Fluoride exposure alters Ca 2+ signaling and mitochondrial function in enamel cells. Sci Signal 2020; 13:eaay0086. [PMID: 32071168 PMCID: PMC7173621 DOI: 10.1126/scisignal.aay0086] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Fluoride ions are highly reactive, and their incorporation in forming dental enamel at low concentrations promotes mineralization. In contrast, excessive fluoride intake causes dental fluorosis, visually recognizable enamel defects that can increase the risk of caries. To investigate the molecular bases of dental fluorosis, we analyzed the effects of fluoride exposure in enamel cells to assess its impact on Ca2+ signaling. Primary enamel cells and an enamel cell line (LS8) exposed to fluoride showed decreased internal Ca2+ stores and store-operated Ca2+ entry (SOCE). RNA-sequencing analysis revealed changes in gene expression suggestive of endoplasmic reticulum (ER) stress in fluoride-treated LS8 cells. Fluoride exposure did not alter Ca2+ homeostasis or increase the expression of ER stress-associated genes in HEK-293 cells. In enamel cells, fluoride exposure affected the functioning of the ER-localized Ca2+ channel IP3R and the activity of the sarco-endoplasmic reticulum Ca2+-ATPase (SERCA) pump during Ca2+ refilling of the ER. Fluoride negatively affected mitochondrial respiration, elicited mitochondrial membrane depolarization, and disrupted mitochondrial morphology. Together, these data provide a potential mechanism underlying dental fluorosis.
Collapse
Affiliation(s)
- Francisco J Aulestia
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY 10010, USA
| | - Johnny Groeling
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY 10010, USA
| | - Guilherme H S Bomfim
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY 10010, USA
| | - Veronica Costiniti
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY 10010, USA
| | - Vinu Manikandan
- Biology Program, Division of Science and Mathematics, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Ariya Chaloemtoem
- Biology Program, Division of Science and Mathematics, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Axel R Concepcion
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | - Yi Li
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY 10010, USA
| | - Larry E Wagner
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY 14526, USA
| | - Youssef Idaghdour
- Biology Program, Division of Science and Mathematics, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - David I Yule
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY 14526, USA
| | - Rodrigo S Lacruz
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY 10010, USA.
| |
Collapse
|
30
|
Ministro JH, Oliveira SS, Oliveira JG, Cardoso M, Aires-da-Silva F, Corte-Real S, Goncalves J. Synthetic antibody discovery against native antigens by CRISPR/Cas9-library generation and endoplasmic reticulum screening. Appl Microbiol Biotechnol 2020; 104:2501-2512. [PMID: 32020276 DOI: 10.1007/s00253-020-10423-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 01/18/2020] [Accepted: 01/26/2020] [Indexed: 01/03/2023]
Abstract
Despite the significant advances of antibodies as therapeutic agents, there is still much room for improvement concerning the discovery of these macromolecules. Here, we present a new synthetic cell-based strategy that takes advantage of eukaryotic cell biology to produce highly diverse antibody libraries and, simultaneously, link them to a high-throughput selection mechanism, replicating B cell diversification mechanisms. The interference of site-specific recognition by CRISPR/Cas9 with error-prone DNA repair mechanisms was explored for the generation of diversity, in a cell population containing a gene for a light chain antibody fragment. We achieved up to 93% of cells containing a mutated antibody gene after diversification mechanisms, specifically inside one of the antigen-binding sites. This targeted variability strategy was then integrated into an intracellular selection mechanism. By fusing the antibody with a KDEL retention signal, the interaction of antibodies and native membrane antigens occurs inside the endoplasmic reticulum during the process of protein secretion, enabling the detection of high-quality leads for expression and affinity by flow cytometry. We successfully obtained antibody lead candidates against CD3 as proof of concept. In summary, we developed a novel antibody discovery platform against native antigens by endoplasmic synthetic library generation using CRISPR/Cas9, which will contribute to a faster discovery of new biotherapeutic molecules, reducing the time-to-market.
Collapse
Affiliation(s)
- Joana H Ministro
- iMed.ULisboa - Research Institute for Medicines, Faculty of Pharmacy, University of Lisbon, Av. Professor Gama Pinto, 1649-019, Lisbon, Portugal.,Technophage SA, Av. Prof. Egas Moniz, Edifício Egas Moniz, 1649-028, Lisbon, Portugal
| | - Soraia S Oliveira
- iMed.ULisboa - Research Institute for Medicines, Faculty of Pharmacy, University of Lisbon, Av. Professor Gama Pinto, 1649-019, Lisbon, Portugal.,Technophage SA, Av. Prof. Egas Moniz, Edifício Egas Moniz, 1649-028, Lisbon, Portugal
| | - Joana G Oliveira
- iMed.ULisboa - Research Institute for Medicines, Faculty of Pharmacy, University of Lisbon, Av. Professor Gama Pinto, 1649-019, Lisbon, Portugal.,Technophage SA, Av. Prof. Egas Moniz, Edifício Egas Moniz, 1649-028, Lisbon, Portugal
| | - Miguel Cardoso
- iMed.ULisboa - Research Institute for Medicines, Faculty of Pharmacy, University of Lisbon, Av. Professor Gama Pinto, 1649-019, Lisbon, Portugal
| | - Frederico Aires-da-Silva
- CIISA - Faculdade de Medicina Veterinária, Universidade de Lisboa, Av. da Universidade Técnica, 1300-477, Lisbon, Portugal
| | - Sofia Corte-Real
- iMed.ULisboa - Research Institute for Medicines, Faculty of Pharmacy, University of Lisbon, Av. Professor Gama Pinto, 1649-019, Lisbon, Portugal.,Technophage SA, Av. Prof. Egas Moniz, Edifício Egas Moniz, 1649-028, Lisbon, Portugal
| | - Joao Goncalves
- iMed.ULisboa - Research Institute for Medicines, Faculty of Pharmacy, University of Lisbon, Av. Professor Gama Pinto, 1649-019, Lisbon, Portugal.
| |
Collapse
|
31
|
Di Martino R, Sticco L, Luini A. Regulation of cargo export and sorting at the trans‐Golgi network. FEBS Lett 2019; 593:2306-2318. [DOI: 10.1002/1873-3468.13572] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 08/02/2019] [Accepted: 08/06/2019] [Indexed: 12/28/2022]
Affiliation(s)
- Rosaria Di Martino
- Institute of Biochemistry and Cell Biology (IBBC) Italian National Research Council (CNR) Naples Italy
| | - Lucia Sticco
- Institute of Biochemistry and Cell Biology (IBBC) Italian National Research Council (CNR) Naples Italy
| | - Alberto Luini
- Institute of Biochemistry and Cell Biology (IBBC) Italian National Research Council (CNR) Naples Italy
| |
Collapse
|
32
|
Igbaria A, Merksamer PI, Trusina A, Tilahun F, Johnson JR, Brandman O, Krogan NJ, Weissman JS, Papa FR. Chaperone-mediated reflux of secretory proteins to the cytosol during endoplasmic reticulum stress. Proc Natl Acad Sci U S A 2019; 116:11291-11298. [PMID: 31101715 PMCID: PMC6561268 DOI: 10.1073/pnas.1904516116] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Diverse perturbations to endoplasmic reticulum (ER) functions compromise the proper folding and structural maturation of secretory proteins. To study secretory pathway physiology during such "ER stress," we employed an ER-targeted, redox-responsive, green fluorescent protein-eroGFP-that reports on ambient changes in oxidizing potential. Here we find that diverse ER stress regimes cause properly folded, ER-resident eroGFP (and other ER luminal proteins) to "reflux" back to the reducing environment of the cytosol as intact, folded proteins. By utilizing eroGFP in a comprehensive genetic screen in Saccharomyces cerevisiae, we show that ER protein reflux during ER stress requires specific chaperones and cochaperones residing in both the ER and the cytosol. Chaperone-mediated ER protein reflux does not require E3 ligase activity, and proceeds even more vigorously when these ER-associated degradation (ERAD) factors are crippled, suggesting that reflux may work in parallel with ERAD. In summary, chaperone-mediated ER protein reflux may be a conserved protein quality control process that evolved to maintain secretory pathway homeostasis during ER protein-folding stress.
Collapse
Affiliation(s)
- Aeid Igbaria
- Department of Medicine, University of California, San Francisco, CA 94143
- Diabetes Center, University of California, San Francisco, CA 94143
- Quantitative Biosciences Institute, University of California, San Francisco, CA 94143
| | - Philip I Merksamer
- Department of Medicine, University of California, San Francisco, CA 94143
- Diabetes Center, University of California, San Francisco, CA 94143
- Quantitative Biosciences Institute, University of California, San Francisco, CA 94143
- Gladstone Institute of Virology and Immunology, San Francisco, CA 94158
| | - Ala Trusina
- Center for Models of Life, Niels Bohr Institute, University of Copenhagen, DK 2100 Copenhagen, Denmark
| | - Firehiwot Tilahun
- Department of Medicine, University of California, San Francisco, CA 94143
- Diabetes Center, University of California, San Francisco, CA 94143
- Quantitative Biosciences Institute, University of California, San Francisco, CA 94143
| | - Jeffrey R Johnson
- Gladstone Institute of Virology and Immunology, San Francisco, CA 94158
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94143
| | - Onn Brandman
- Quantitative Biosciences Institute, University of California, San Francisco, CA 94143
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94143
- Howard Hughes Medical Institute, University of California, San Francisco, CA 94143
| | - Nevan J Krogan
- Gladstone Institute of Virology and Immunology, San Francisco, CA 94158
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94143
| | - Jonathan S Weissman
- Quantitative Biosciences Institute, University of California, San Francisco, CA 94143
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94143
- Howard Hughes Medical Institute, University of California, San Francisco, CA 94143
| | - Feroz R Papa
- Department of Medicine, University of California, San Francisco, CA 94143;
- Diabetes Center, University of California, San Francisco, CA 94143
- Quantitative Biosciences Institute, University of California, San Francisco, CA 94143
| |
Collapse
|
33
|
Lamandé SR, Bateman JF. Genetic Disorders of the Extracellular Matrix. Anat Rec (Hoboken) 2019; 303:1527-1542. [PMID: 30768852 PMCID: PMC7318566 DOI: 10.1002/ar.24086] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 10/26/2018] [Indexed: 12/11/2022]
Abstract
Mutations in the genes for extracellular matrix (ECM) components cause a wide range of genetic connective tissues disorders throughout the body. The elucidation of mutations and their correlation with pathology has been instrumental in understanding the roles of many ECM components. The pathological consequences of ECM protein mutations depend on its tissue distribution, tissue function, and on the nature of the mutation. The prevalent paradigm for the molecular pathology has been that there are two global mechanisms. First, mutations that reduce the production of ECM proteins impair matrix integrity largely due to quantitative ECM defects. Second, mutations altering protein structure may reduce protein secretion but also introduce dominant negative effects in ECM formation, structure and/or stability. Recent studies show that endoplasmic reticulum (ER) stress, caused by mutant misfolded ECM proteins, makes a significant contribution to the pathophysiology. This suggests that targeting ER‐stress may offer a new therapeutic strategy in a range of ECM disorders caused by protein misfolding mutations. Anat Rec, 2019. © 2019 The Authors. The Anatomical Record published by Wiley Periodicals, Inc. on behalf of American Association of Anatomists.
Collapse
Affiliation(s)
- Shireen R Lamandé
- Musculoskeletal Research, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville Victoria, Australia.,Department of Paediatrics, University of Melbourne, Parkville Victoria, Australia
| | - John F Bateman
- Musculoskeletal Research, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville Victoria, Australia.,Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville Victoria, Australia
| |
Collapse
|
34
|
Tao YX, Conn PM. Pharmacoperones as Novel Therapeutics for Diverse Protein Conformational Diseases. Physiol Rev 2018; 98:697-725. [PMID: 29442594 DOI: 10.1152/physrev.00029.2016] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
After synthesis, proteins are folded into their native conformations aided by molecular chaperones. Dysfunction in folding caused by genetic mutations in numerous genes causes protein conformational diseases. Membrane proteins are more prone to misfolding due to their more intricate folding than soluble proteins. Misfolded proteins are detected by the cellular quality control systems, especially in the endoplasmic reticulum, and proteins may be retained there for eventual degradation by the ubiquitin-proteasome system or through autophagy. Some misfolded proteins aggregate, leading to pathologies in numerous neurological diseases. In vitro, modulating mutant protein folding by altering molecular chaperone expression can ameliorate some misfolding. Some small molecules known as chemical chaperones also correct mutant protein misfolding in vitro and in vivo. However, due to their lack of specificity, their potential as therapeutics is limited. Another class of compounds, known as pharmacological chaperones (pharmacoperones), binds with high specificity to misfolded proteins, either as enzyme substrates or receptor ligands, leading to decreased folding energy barriers and correction of the misfolding. Because many of the misfolded proteins are misrouted but do not have defects in function per se, pharmacoperones have promising potential in advancing to the clinic as therapeutics, since correcting routing may ameliorate the underlying mechanism of disease. This review will comprehensively summarize this exciting area of research, surveying the literature from in vitro studies in cell lines to transgenic animal models and clinical trials in several protein misfolding diseases.
Collapse
Affiliation(s)
- Ya-Xiong Tao
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University , Auburn, Alabama ; and Departments of Internal Medicine and Cell Biology, Texas Tech University Health Science Center , Lubbock, Texas
| | - P Michael Conn
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University , Auburn, Alabama ; and Departments of Internal Medicine and Cell Biology, Texas Tech University Health Science Center , Lubbock, Texas
| |
Collapse
|
35
|
Sundaram A, Appathurai S, Plumb R, Mariappan M. Dynamic changes in complexes of IRE1α, PERK, and ATF6α during endoplasmic reticulum stress. Mol Biol Cell 2018; 29:1376-1388. [PMID: 29851562 PMCID: PMC5994896 DOI: 10.1091/mbc.e17-10-0594] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The endoplasmic reticulum (ER) localized unfolded protein response (UPR) sensors, IRE1α, PERK, and ATF6α, are activated by the accumulation of misfolded proteins in the ER. It is unclear how the endogenous UPR sensors are regulated by both ER stress and the ER luminal chaperone BiP, which is a negative regulator of UPR sensors. Here we simultaneously examined the changes in the endogenous complexes of UPR sensors by blue native PAGE immunoblotting in unstressed and stressed cells. We found that all three UPR sensors exist as preformed complexes even in unstressed cells. While PERK complexes shift to large complexes, ATF6α complexes are reduced to smaller complexes on ER stress. In contrast, IRE1α complexes were not significantly increased in size on ER stress, unless IRE1α is overexpressed. Surprisingly, depletion of BiP had little impact on the endogenous complexes of UPR sensors. In addition, overexpression of BiP did not significantly affect UPR complexes, but suppressed ER stress mediated activation of IRE1α, ATF6α and, to a lesser extent, PERK. Furthermore, we captured the interaction between IRE1α and misfolded secretory proteins in cells, which suggests that the binding of unfolded proteins to preformed complexes of UPR sensors may be crucial for activation.
Collapse
Affiliation(s)
- Arunkumar Sundaram
- Department of Cell Biology, Nanobiology Institute, Yale School of Medicine, West Haven, CT 06516
| | - Suhila Appathurai
- Department of Cell Biology, Nanobiology Institute, Yale School of Medicine, West Haven, CT 06516
| | - Rachel Plumb
- Department of Cell Biology, Nanobiology Institute, Yale School of Medicine, West Haven, CT 06516
| | - Malaiyalam Mariappan
- Department of Cell Biology, Nanobiology Institute, Yale School of Medicine, West Haven, CT 06516
| |
Collapse
|
36
|
Berner N, Reutter KR, Wolf DH. Protein Quality Control of the Endoplasmic Reticulum and Ubiquitin-Proteasome-Triggered Degradation of Aberrant Proteins: Yeast Pioneers the Path. Annu Rev Biochem 2018; 87:751-782. [PMID: 29394096 DOI: 10.1146/annurev-biochem-062917-012749] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Cells must constantly monitor the integrity of their macromolecular constituents. Proteins are the most versatile class of macromolecules but are sensitive to structural alterations. Misfolded or otherwise aberrant protein structures lead to dysfunction and finally aggregation. Their presence is linked to aging and a plethora of severe human diseases. Thus, misfolded proteins have to be rapidly eliminated. Secretory proteins constitute more than one-third of the eukaryotic proteome. They are imported into the endoplasmic reticulum (ER), where they are folded and modified. A highly elaborated machinery controls their folding, recognizes aberrant folding states, and retrotranslocates permanently misfolded proteins from the ER back to the cytosol. In the cytosol, they are degraded by the highly selective ubiquitin-proteasome system. This process of protein quality control followed by proteasomal elimination of the misfolded protein is termed ER-associated degradation (ERAD), and it depends on an intricate interplay between the ER and the cytosol.
Collapse
Affiliation(s)
- Nicole Berner
- Institute of Biochemistry and Technical Biochemistry, University of Stuttgart, 70569 Stuttgart, Germany; , ,
| | - Karl-Richard Reutter
- Institute of Biochemistry and Technical Biochemistry, University of Stuttgart, 70569 Stuttgart, Germany; , ,
| | - Dieter H Wolf
- Institute of Biochemistry and Technical Biochemistry, University of Stuttgart, 70569 Stuttgart, Germany; , ,
| |
Collapse
|
37
|
Inflammation induced ER stress affects absorptive intestinal epithelial cells function and integrity. Int Immunopharmacol 2018; 55:336-344. [DOI: 10.1016/j.intimp.2017.12.016] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Revised: 12/11/2017] [Accepted: 12/12/2017] [Indexed: 02/07/2023]
|
38
|
Hetz C, Papa FR. The Unfolded Protein Response and Cell Fate Control. Mol Cell 2018; 69:169-181. [DOI: 10.1016/j.molcel.2017.06.017] [Citation(s) in RCA: 744] [Impact Index Per Article: 106.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 06/08/2017] [Accepted: 06/15/2017] [Indexed: 12/12/2022]
|
39
|
Bakunts A, Orsi A, Vitale M, Cattaneo A, Lari F, Tadè L, Sitia R, Raimondi A, Bachi A, van Anken E. Ratiometric sensing of BiP-client versus BiP levels by the unfolded protein response determines its signaling amplitude. eLife 2017; 6:27518. [PMID: 29251598 PMCID: PMC5792092 DOI: 10.7554/elife.27518] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 12/15/2017] [Indexed: 01/03/2023] Open
Abstract
Insufficient folding capacity of the endoplasmic reticulum (ER) activates the unfolded protein response (UPR) to restore homeostasis. Yet, how the UPR achieves ER homeostatic readjustment is poorly investigated, as in most studies the ER stress that is elicited cannot be overcome. Here we show that a proteostatic insult, provoked by persistent expression of the secretory heavy chain of immunoglobulin M (µs), is well-tolerated in HeLa cells. Upon µs expression, its levels temporarily eclipse those of the ER chaperone BiP, leading to acute, full-geared UPR activation. Once BiP is in excess again, the UPR transitions to chronic, submaximal activation, indicating that the UPR senses ER stress in a ratiometric fashion. In this process, the ER expands about three-fold and becomes dominated by BiP. As the UPR is essential for successful ER homeostatic readjustment in the HeLa-µs model, it provides an ideal system for dissecting the intricacies of how the UPR evaluates and alleviates ER stress.
Collapse
Affiliation(s)
- Anush Bakunts
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Andrea Orsi
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy.,Università Vita-Salute San Raffaele, Milan, Italy
| | - Milena Vitale
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy.,Università Vita-Salute San Raffaele, Milan, Italy
| | | | - Federica Lari
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Laura Tadè
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Roberto Sitia
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy.,Università Vita-Salute San Raffaele, Milan, Italy
| | - Andrea Raimondi
- Experimental Imaging Center, San Raffaele Scientific Institute, Milan, Italy
| | - Angela Bachi
- IFOM, FIRC Institute of Molecular Oncology, Milan, Italy
| | - Eelco van Anken
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy.,Università Vita-Salute San Raffaele, Milan, Italy
| |
Collapse
|
40
|
Cao Z, Xiao Q, Dai X, Zhou Z, Jiang R, Cheng Y, Yang X, Guo H, Wang J, Xi Z, Yao H, Chao J. circHIPK2-mediated σ-1R promotes endoplasmic reticulum stress in human pulmonary fibroblasts exposed to silica. Cell Death Dis 2017; 8:3212. [PMID: 29238093 PMCID: PMC5870587 DOI: 10.1038/s41419-017-0017-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 09/16/2017] [Accepted: 10/02/2017] [Indexed: 12/16/2022]
Abstract
Silicosis is characterized by fibroblast accumulation and excessive deposition of extracellular matrix. Although the roles of SiO2-induced chemokines and cytokines released from alveolar macrophages have received significant attention, the direct effects of SiO2 on protein production and functional changes in pulmonary fibroblasts have been less extensively studied. Sigma-1 receptor, which has been associated with cell proliferation and migration in the central nervous system, is expressed in the lung, but its role in silicosis remains unknown. To elucidate the role of sigma-1 receptor in fibrosis induced by silica, both the upstream molecular mechanisms and the functional effects on cell proliferation and migration were investigated. Both molecular biological assays and pharmacological techniques, combined with functional experiments, such as migration and proliferation, were applied in human pulmonary fibroblasts from adults to analyze the molecular and functional changes induced by SiO2. SiO2 induced endoplasmic reticulum stress in association with enhanced expression of sigma-1 receptor. Endoplasmic reticulum stress promoted migration and proliferation of human pulmonary fibroblasts-adult exposed to SiO2, inducing the development of silicosis. Inhibition of sigma-1 receptor ameliorated endoplasmic reticulum stress and fibroblast functional changes induced by SiO2. circHIPK2 is involved in the regulation of sigma-1 receptor in human pulmonary fibroblasts-adult exposed to SiO2. Our study elucidated a link between SiO2-induced fibrosis and sigma-1 receptor signaling, thereby providing novel insight into the potential use of sigma-1 receptor/endoplasmic reticulum stress in the development of novel therapeutic strategies for silicosis treatment.
Collapse
Affiliation(s)
- Zhouli Cao
- Department of Physiology, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China
- Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, 210096, China
- Department of Respiration, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Qingling Xiao
- Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Xiaoniu Dai
- Department of Physiology, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Zewei Zhou
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Rong Jiang
- Department of Physiology, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China
- Department of Respiration, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Yusi Cheng
- Department of Physiology, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Xiyue Yang
- Department of Physiology, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China
- Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, 210096, China
| | - Huifang Guo
- Department of Physiology, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China
- Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, 210096, China
| | - Jing Wang
- Department of Physiology, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Zhaoqing Xi
- Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Honghong Yao
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China
- Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, 210096, China
| | - Jie Chao
- Department of Physiology, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China.
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China.
- Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, 210096, China.
- Department of Respiration, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China.
| |
Collapse
|
41
|
Luarte A, Cornejo VH, Bertin F, Gallardo J, Couve A. The axonal endoplasmic reticulum: One organelle-many functions in development, maintenance, and plasticity. Dev Neurobiol 2017; 78:181-208. [PMID: 29134778 DOI: 10.1002/dneu.22560] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 11/02/2017] [Accepted: 11/07/2017] [Indexed: 12/11/2022]
Abstract
The endoplasmic reticulum (ER) is highly conserved in eukaryotes and neurons. Indeed, the localization of the organelle in axons has been known for nearly half a century. However, the relevance of the axonal ER is only beginning to emerge. In this review, we discuss the structure of the ER in axons, examining the role of ER-shaping proteins and highlighting reticulons. We analyze the multiple functions of the ER and their potential contribution to axonal physiology. First, we examine the emerging roles of the axonal ER in lipid synthesis, protein translation, processing, quality control, and secretory trafficking of transmembrane proteins. We also review the impact of the ER on calcium dynamics, focusing on intracellular mechanisms and functions. We describe the interactions between the ER and endosomes, mitochondria, and synaptic vesicles. Finally, we analyze available proteomic data of axonal preparations to reveal the dynamic functionality of the ER in axons during development. We suggest that the dynamic proteome and a validated axonal interactome, together with state-of-the-art methodologies, may provide interesting research avenues in axon physiology that may extend to pathology and regeneration. © 2017 Wiley Periodicals, Inc. Develop Neurobiol 78: 181-208, 2018.
Collapse
Affiliation(s)
- Alejandro Luarte
- Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Víctor Hugo Cornejo
- Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Francisca Bertin
- Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Javiera Gallardo
- Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Andrés Couve
- Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| |
Collapse
|
42
|
Kumar R, Kumari B, Kumar M. Prediction of endoplasmic reticulum resident proteins using fragmented amino acid composition and support vector machine. PeerJ 2017; 5:e3561. [PMID: 28890846 PMCID: PMC5588793 DOI: 10.7717/peerj.3561] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 06/20/2017] [Indexed: 12/15/2022] Open
Abstract
Background The endoplasmic reticulum plays an important role in many cellular processes, which includes protein synthesis, folding and post-translational processing of newly synthesized proteins. It is also the site for quality control of misfolded proteins and entry point of extracellular proteins to the secretory pathway. Hence at any given point of time, endoplasmic reticulum contains two different cohorts of proteins, (i) proteins involved in endoplasmic reticulum-specific function, which reside in the lumen of the endoplasmic reticulum, called as endoplasmic reticulum resident proteins and (ii) proteins which are in process of moving to the extracellular space. Thus, endoplasmic reticulum resident proteins must somehow be distinguished from newly synthesized secretory proteins, which pass through the endoplasmic reticulum on their way out of the cell. Approximately only 50% of the proteins used in this study as training data had endoplasmic reticulum retention signal, which shows that these signals are not essentially present in all endoplasmic reticulum resident proteins. This also strongly indicates the role of additional factors in retention of endoplasmic reticulum-specific proteins inside the endoplasmic reticulum. Methods This is a support vector machine based method, where we had used different forms of protein features as inputs for support vector machine to develop the prediction models. During training leave-one-out approach of cross-validation was used. Maximum performance was obtained with a combination of amino acid compositions of different part of proteins. Results In this study, we have reported a novel support vector machine based method for predicting endoplasmic reticulum resident proteins, named as ERPred. During training we achieved a maximum accuracy of 81.42% with leave-one-out approach of cross-validation. When evaluated on independent dataset, ERPred did prediction with sensitivity of 72.31% and specificity of 83.69%. We have also annotated six different proteomes to predict the candidate endoplasmic reticulum resident proteins in them. A webserver, ERPred, was developed to make the method available to the scientific community, which can be accessed at http://proteininformatics.org/mkumar/erpred/index.html. Discussion We found that out of 124 proteins of the training dataset, only 66 proteins had endoplasmic reticulum retention signals, which shows that these signals are not an absolute necessity for endoplasmic reticulum resident proteins to remain inside the endoplasmic reticulum. This observation also strongly indicates the role of additional factors in retention of proteins inside the endoplasmic reticulum. Our proposed predictor, ERPred, is a signal independent tool. It is tuned for the prediction of endoplasmic reticulum resident proteins, even if the query protein does not contain specific ER-retention signal.
Collapse
Affiliation(s)
- Ravindra Kumar
- Department of Biophysics, University of Delhi South Campus, New Delhi, India.,Current affiliation: Newe-Ya'ar Research Center, Agricultural Research Organization, Ramat Yishay, Israel
| | - Bandana Kumari
- Department of Biophysics, University of Delhi South Campus, New Delhi, India
| | - Manish Kumar
- Department of Biophysics, University of Delhi South Campus, New Delhi, India
| |
Collapse
|
43
|
Böldicke T. Single domain antibodies for the knockdown of cytosolic and nuclear proteins. Protein Sci 2017; 26:925-945. [PMID: 28271570 PMCID: PMC5405437 DOI: 10.1002/pro.3154] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 03/03/2017] [Indexed: 12/12/2022]
Abstract
Single domain antibodies (sdAbs) from camels or sharks comprise only the variable heavy chain domain. Human sdAbs comprise the variable domain of the heavy chain (VH) or light chain (VL) and can be selected from human antibodies. SdAbs are stable, nonaggregating molecules in vitro and in vivo compared to complete antibodies and scFv fragments. They are excellent novel inhibitors of cytosolic/nuclear proteins because they are correctly folded inside the cytosol in contrast to scFv fragments. SdAbs are unique because of their excellent specificity and possibility to target posttranslational modifications such as phosphorylation sites, conformers or interaction regions of proteins that cannot be targeted with genetic knockout techniques and are impossible to knockdown with RNAi. The number of inhibiting cytosolic/nuclear sdAbs is increasing and usage of synthetic single pot single domain antibody libraries will boost the generation of these fascinating molecules without the need of immunization. The most frequently selected antigenic epitopes belong to viral and oncogenic proteins, followed by toxins, proteins of the nervous system as well as plant- and drosophila proteins. It is now possible to select functional sdAbs against virtually every cytosolic/nuclear protein and desired epitope. The development of new endosomal escape protein domains and cell-penetrating peptides for efficient transfection broaden the application of inhibiting sdAbs. Last but not least, the generation of relatively new cell-specific nanoparticles such as polymersomes and polyplexes carrying cytosolic/nuclear sdAb-DNA or -protein will pave the way to apply cytosolic/nuclear sdAbs for inhibition of viral infection and cancer in the clinic.
Collapse
Affiliation(s)
- Thomas Böldicke
- Helmholtz Centre for Infection Research, Structure and Function of ProteinsInhoffenstraße 7, D‐38124BraunschweigGermany
| |
Collapse
|
44
|
Ye ZW, Zhang J, Ancrum T, Manevich Y, Townsend DM, Tew KD. Glutathione S-Transferase P-Mediated Protein S-Glutathionylation of Resident Endoplasmic Reticulum Proteins Influences Sensitivity to Drug-Induced Unfolded Protein Response. Antioxid Redox Signal 2017; 26:247-261. [PMID: 26838680 PMCID: PMC5312626 DOI: 10.1089/ars.2015.6486] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
AIMS S-glutathionylation of cysteine residues, catalyzed by glutathione S-transferase Pi (GSTP), alters structure/function characteristics of certain targeted proteins. Our goal is to characterize how S-glutathionylation of proteins within the endoplasmic reticulum (ER) impact cell sensitivity to ER-stress inducing drugs. RESULTS We identify GSTP to be an ER-resident protein where it demonstrates both chaperone and catalytic functions. Redox based proteomic analyses identified a cluster of proteins cooperatively involved in the regulation of ER stress (immunoglobulin heavy chain-binding protein [BiP], protein disulfide isomerase [PDI], calnexin, calreticulin, endoplasmin, sarco/endoplasmic reticulum Ca2+-ATPase [SERCA]) that individually co-immunoprecipitated with GSTP (implying protein complex formation) and were subject to reactive oxygen species (ROS) induced S-glutathionylation. S-glutathionylation of each of these six proteins was attenuated in cells (liver, embryo fibroblasts or bone marrow dendritic) from mice lacking GSTP (Gstp1/p2-/-) compared to wild type (Gstp1/p2+/+). Moreover, Gstp1/p2-/- cells were significantly more sensitive to the cytotoxic effects of the ER-stress inducing drugs, thapsigargin (7-fold) and tunicamycin (2-fold). INNOVATION Within the family of GST isozymes, GSTP has been ascribed the broadest range of catalytic and chaperone functions. Now, for the first time, we identify it as an ER resident protein that catalyzes S-glutathionylation of critical ER proteins within this organelle. Of note, this can provide a nexus for linkage of redox based signaling and pathways that regulate the unfolded protein response (UPR). This has novel importance in determining how some drugs kill cancer cells. CONCLUSIONS Contextually, these results provide mechanistic evidence that GSTP can exert redox regulation in the oxidative ER environment and indicate that, within the ER, GSTP influences the cellular consequences of the UPR through S-glutathionylation of a series of key interrelated proteins. Antioxid. Redox Signal. 26, 247-261.
Collapse
Affiliation(s)
- Zhi-Wei Ye
- 1 Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina , Charleston, South Carolina
| | - Jie Zhang
- 1 Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina , Charleston, South Carolina
| | - Tiffany Ancrum
- 1 Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina , Charleston, South Carolina
| | - Yefim Manevich
- 1 Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina , Charleston, South Carolina
| | - Danyelle M Townsend
- 2 Department of Pharmaceutical and Biomedical Sciences, Medical University of South Carolina , Charleston, South Carolina
| | - Kenneth D Tew
- 1 Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina , Charleston, South Carolina
| |
Collapse
|
45
|
Peng RH, Qiu J, Tian YS, Gao JJ, Han HJ, Fu XY, Zhu B, Xu J, Wang B, Li ZJ, Wang LJ, Yao QH. Disulfide isomerase-like protein AtPDIL1-2 is a good candidate for trichlorophenol phytodetoxification. Sci Rep 2017; 7:40130. [PMID: 28059139 PMCID: PMC5216352 DOI: 10.1038/srep40130] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 12/02/2016] [Indexed: 12/29/2022] Open
Abstract
Trichlorophenol (TCP) is a widely used and persistent environmentally toxic compound that poses a carcinogenic risk to humans. Phytoremediation is a proficient cleanup technology for organic pollutants. In this study, we found that the disulfide isomerase-like protein AtPDIL1-2 in plants is a good candidate for enhancing 2,4,6-TCP phytoremediation. The expression of AtPDIL1-2 in Arabidopsis was induced by 2,4,6-TCP. The heterologously expressed AtPDIL1-2 in Escherichia coli exhibited both oxidase and isomerase activities as protein disulfide isomerase and improved bacteria tolerance to 2,4,6-TCP. Further research revealed that transgenic tobacco overexpressing AtPDIL1-2 was more tolerant to high concentrations of 2,4,6-TCP and removed the toxic compound at far greater rates than the control plants. To elucidate the mechanism of action of AtPDIL1-2, we investigated the chemical interaction of AtPDIL1-2 with 2,4,6-TCP for the first time. HPLC analysis implied that AtPDIL1-2 exerts a TCP-binding activity. A suitable configuration of AtPDIL1-2-TCP binding was obtained by molecular docking studies using the AutoDock program. It predicted that the TCP binding site is located in the b-b' domain of AtPDIL1-2 and that His254 of the protein is critical for the binding interaction. These findings imply that AtPDIL1-2 can be used for TCP detoxification by the way of overexpression in plants.
Collapse
Affiliation(s)
- Ri-He Peng
- Agro-Biotechnology Research Institute, Shanghai Academy of Agricultural Sciences; Shanghai Key Laboratory of Agricultural Genetics and Breeding, 2901 Beidi Rd., Shanghai, People’s Republic of China
| | - Jin Qiu
- Agro-Biotechnology Research Institute, Shanghai Academy of Agricultural Sciences; Shanghai Key Laboratory of Agricultural Genetics and Breeding, 2901 Beidi Rd., Shanghai, People’s Republic of China
| | - Yong-Sheng Tian
- Agro-Biotechnology Research Institute, Shanghai Academy of Agricultural Sciences; Shanghai Key Laboratory of Agricultural Genetics and Breeding, 2901 Beidi Rd., Shanghai, People’s Republic of China
| | - Jian-jie Gao
- Agro-Biotechnology Research Institute, Shanghai Academy of Agricultural Sciences; Shanghai Key Laboratory of Agricultural Genetics and Breeding, 2901 Beidi Rd., Shanghai, People’s Republic of China
| | - Hong-juan Han
- Agro-Biotechnology Research Institute, Shanghai Academy of Agricultural Sciences; Shanghai Key Laboratory of Agricultural Genetics and Breeding, 2901 Beidi Rd., Shanghai, People’s Republic of China
| | - Xiao-Yan Fu
- Agro-Biotechnology Research Institute, Shanghai Academy of Agricultural Sciences; Shanghai Key Laboratory of Agricultural Genetics and Breeding, 2901 Beidi Rd., Shanghai, People’s Republic of China
| | - Bo Zhu
- Agro-Biotechnology Research Institute, Shanghai Academy of Agricultural Sciences; Shanghai Key Laboratory of Agricultural Genetics and Breeding, 2901 Beidi Rd., Shanghai, People’s Republic of China
| | - Jing Xu
- Agro-Biotechnology Research Institute, Shanghai Academy of Agricultural Sciences; Shanghai Key Laboratory of Agricultural Genetics and Breeding, 2901 Beidi Rd., Shanghai, People’s Republic of China
| | - Bo Wang
- Agro-Biotechnology Research Institute, Shanghai Academy of Agricultural Sciences; Shanghai Key Laboratory of Agricultural Genetics and Breeding, 2901 Beidi Rd., Shanghai, People’s Republic of China
| | - Zhen-jun Li
- Agro-Biotechnology Research Institute, Shanghai Academy of Agricultural Sciences; Shanghai Key Laboratory of Agricultural Genetics and Breeding, 2901 Beidi Rd., Shanghai, People’s Republic of China
| | - Li-juan Wang
- Agro-Biotechnology Research Institute, Shanghai Academy of Agricultural Sciences; Shanghai Key Laboratory of Agricultural Genetics and Breeding, 2901 Beidi Rd., Shanghai, People’s Republic of China
| | - Quan-Hong Yao
- Agro-Biotechnology Research Institute, Shanghai Academy of Agricultural Sciences; Shanghai Key Laboratory of Agricultural Genetics and Breeding, 2901 Beidi Rd., Shanghai, People’s Republic of China
| |
Collapse
|
46
|
Bhoola NH, Kramvis A. Hepatitis B e Antigen Expression by Hepatitis B Virus Subgenotype A1 Relative to Subgenotypes A2 and D3 in Cultured Hepatocellular Carcinoma (Huh7) Cells. Intervirology 2016; 59:48-59. [PMID: 27553619 DOI: 10.1159/000446240] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 04/16/2016] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Hepatitis B virus (HBV) is hyperendemic in southern Africa, with subgenotype A1 prevailing. The precore/core (preC/C) region of A1, encoding for hepatitis B e antigen (HBeAg), has unique sequence characteristics, differentiating it from subgenotypes A2 and D3. Our aim was to follow the expression of HBeAg in vitro by the three subgenotypes. METHODS Huh7 cells were transfected with plasmids belonging to subgenotypes A1, A2, and D3. Using indirect immunofluorescence, the expression of HBeAg was followed, as was the activation of the unfolded protein response (UPR) and subsequent activation of apoptosis. RESULTS AND CONCLUSIONS Following transfection with D3, HBeAg passed through the secretory pathway earlier than cells transfected with genotype A. Cells transfected with A1 showed a lower expression of the preC/C precursor in the secretory pathway and a higher co-localization in the nucleus. Cells transfected with A1 showed greater endoplasmic reticulum (ER) stress and an earlier, prolonged activation of the UPR seen by the higher activity of three ER-localized transmembrane transducers (double-stranded RNA-dependent protein kinase-like ER kinase, activating transcription factor 6, and inositol-requiring enzyme 1), on day 3 compared to day 5. Moreover, our study also found that cells transfected with A1 had increased apoptosis.
Collapse
Affiliation(s)
- Nimisha Harshadrai Bhoola
- Hepatitis Virus Diversity Research Unit, Department of Internal Medicine, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | | |
Collapse
|
47
|
Feldman HC, Tong M, Wang L, Meza-Acevedo R, Gobillot TA, Lebedev I, Gliedt MJ, Hari SB, Mitra AK, Backes BJ, Papa FR, Seeliger MA, Maly DJ. Structural and Functional Analysis of the Allosteric Inhibition of IRE1α with ATP-Competitive Ligands. ACS Chem Biol 2016; 11:2195-205. [PMID: 27227314 DOI: 10.1021/acschembio.5b00940] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The accumulation of unfolded proteins under endoplasmic reticulum (ER) stress leads to the activation of the multidomain protein sensor IRE1α as part of the unfolded protein response (UPR). Clustering of IRE1α lumenal domains in the presence of unfolded proteins promotes kinase trans-autophosphorylation in the cytosol and subsequent RNase domain activation. Interestingly, there is an allosteric relationship between the kinase and RNase domains of IRE1α, which allows ATP-competitive inhibitors to modulate the activity of the RNase domain. Here, we use kinase inhibitors to study how ATP-binding site conformation affects the activity of the RNase domain of IRE1α. We find that diverse ATP-competitive inhibitors of IRE1α promote dimerization and activation of RNase activity despite blocking kinase autophosphorylation. In contrast, a subset of ATP-competitive ligands, which we call KIRAs, allosterically inactivate the RNase domain through the kinase domain by stabilizing monomeric IRE1α. Further insight into how ATP-competitive inhibitors are able to divergently modulate the RNase domain through the kinase domain was gained by obtaining the first structure of apo human IRE1α in the RNase active back-to-back dimer conformation. Comparison of this structure with other existing structures of IRE1α and integration of our extensive structure activity relationship (SAR) data has led us to formulate a model to rationalize how ATP-binding site ligands are able to control the IRE1α oligomeric state and subsequent RNase domain activity.
Collapse
Affiliation(s)
- Hannah C. Feldman
- Department
of Chemistry, University of Washington, Seattle, Washington, United States
| | - Michael Tong
- Department
of Pharmacological Sciences, Stony Brook University Medical School, Stony
Brook, New York, United States
| | - Likun Wang
- Department
of Medicine, University of California−San Francisco, San Francisco, California, United States
- Diabetes
Center, University of California−San Francisco, San Francisco, California, United States
- Lung
Biology Center, University of California−San Francisco, San Francisco, California, United States
- California
Institute for Quantitative Biosciences, University of California−San Francisco, San Francisco, California, United States
| | - Rosa Meza-Acevedo
- Department
of Medicine, University of California−San Francisco, San Francisco, California, United States
- Diabetes
Center, University of California−San Francisco, San Francisco, California, United States
- Lung
Biology Center, University of California−San Francisco, San Francisco, California, United States
- California
Institute for Quantitative Biosciences, University of California−San Francisco, San Francisco, California, United States
| | - Theodore A. Gobillot
- Department
of Chemistry, University of Washington, Seattle, Washington, United States
| | - Ivan Lebedev
- Department
of Pharmacological Sciences, Stony Brook University Medical School, Stony
Brook, New York, United States
| | - Micah J. Gliedt
- Department
of Medicine, University of California−San Francisco, San Francisco, California, United States
- Lung
Biology Center, University of California−San Francisco, San Francisco, California, United States
| | - Sanjay B. Hari
- Department
of Chemistry, University of Washington, Seattle, Washington, United States
| | - Arinjay K. Mitra
- Department
of Chemistry, University of Washington, Seattle, Washington, United States
| | - Bradley J. Backes
- Department
of Medicine, University of California−San Francisco, San Francisco, California, United States
- Lung
Biology Center, University of California−San Francisco, San Francisco, California, United States
| | - Feroz R. Papa
- Department
of Medicine, University of California−San Francisco, San Francisco, California, United States
- Diabetes
Center, University of California−San Francisco, San Francisco, California, United States
- Lung
Biology Center, University of California−San Francisco, San Francisco, California, United States
- California
Institute for Quantitative Biosciences, University of California−San Francisco, San Francisco, California, United States
| | - Markus A. Seeliger
- Department
of Pharmacological Sciences, Stony Brook University Medical School, Stony
Brook, New York, United States
| | - Dustin J. Maly
- Department
of Chemistry, University of Washington, Seattle, Washington, United States
- Department
of Biochemistry, University of Washington, Seattle, Washington, United States
| |
Collapse
|
48
|
de Ruijter JC, Koskela EV, Frey AD. Enhancing antibody folding and secretion by tailoring the Saccharomyces cerevisiae endoplasmic reticulum. Microb Cell Fact 2016; 15:87. [PMID: 27216259 PMCID: PMC4878073 DOI: 10.1186/s12934-016-0488-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 05/11/2016] [Indexed: 01/20/2023] Open
Abstract
Background The yeast Saccharomyces cerevisiae provides intriguing possibilities for synthetic biology and bioprocess applications, but its use is still constrained by cellular characteristics that limit the product yields. Considering the production of advanced biopharmaceuticals, a major hindrance lies in the yeast endoplasmic reticulum (ER), as it is not equipped for efficient and large scale folding of complex proteins, such as human antibodies. Results Following the example of professional secretory cells, we show that inducing an ER expansion in yeast by deleting the lipid-regulator gene OPI1 can improve the secretion capacity of full-length antibodies up to fourfold. Based on wild-type and ER-enlarged yeast strains, we conducted a screening of a folding factor overexpression library to identify proteins and their expression levels that enhance the secretion of antibodies. Out of six genes tested, addition of the peptidyl-prolyl isomerase CPR5 provided the most beneficial effect on specific product yield while PDI1, ERO1, KAR2, LHS1 and SIL1 had a mild or even negative effect to antibody secretion efficiency. Combining genes for ER enhancement did not induce any significant additional effect compared to addition of just one element. By combining the Δopi1 strain, with the enlarged ER, with CPR5 overexpression, we were able to boost the specific antibody product yield by a factor of 10 relative to the non-engineered strain. Conclusions Engineering protein folding in vivo is a major task for biopharmaceuticals production in yeast and needs to be optimized at several levels. By rational strain design and high-throughput screening applications we were able to increase the specific secreted antibody yields of S. cerevisiae up to 10-fold, providing a promising strain for further process optimization and platform development for antibody production. Electronic supplementary material The online version of this article (doi:10.1186/s12934-016-0488-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jorg C de Ruijter
- Department of Biotechnology and Chemical Technology, Aalto University, Kemistintie 1, 02150, Espoo, Finland
| | - Essi V Koskela
- Department of Biotechnology and Chemical Technology, Aalto University, Kemistintie 1, 02150, Espoo, Finland
| | - Alexander D Frey
- Department of Biotechnology and Chemical Technology, Aalto University, Kemistintie 1, 02150, Espoo, Finland.
| |
Collapse
|
49
|
|
50
|
Abstract
Aging involves defined genetic, biochemical and cellular pathways that regulate lifespan. These pathways are called longevity pathways and they have relevance for many age-related diseases. In the eye, longevity pathways are involved in the major blinding diseases, cataract, glaucoma, age-related macular degeneration (AMD) and diabetic retinopathy. Pharmaceutical targeting of longevity pathways can extend healthy lifespan in laboratory model systems. This offers the possibility of therapeutic interventions to also delay onset or slow the progression of age-related eye diseases. I suggest that retinal degeneration may be viewed as accelerated aging of photoreceptors and that interventions extending healthy lifespan may also slow the pace of photoreceptor loss.
Collapse
|