51
|
OSMI-1 Enhances TRAIL-Induced Apoptosis through ER Stress and NF-κB Signaling in Colon Cancer Cells. Int J Mol Sci 2021; 22:ijms222011073. [PMID: 34681736 PMCID: PMC8539180 DOI: 10.3390/ijms222011073] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/09/2021] [Accepted: 10/09/2021] [Indexed: 12/12/2022] Open
Abstract
Levels of O-GlcNAc transferase (OGT) and hyper-O-GlcNAcylation expression levels are associated with cancer pathogenesis. This study aimed to find conditions that maximize the therapeutic effect of cancer and minimize tissue damage by combining an OGT inhibitor (OSMI-1) and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). We found that OSMI-1 treatment in HCT116 human colon cancer cells has a potent synergistic effect on TRAIL-induced apoptosis signaling. Interestingly, OSMI-1 significantly increased TRAIL-mediated apoptosis by increasing the expression of the cell surface receptor DR5. ROS-induced endoplasmic reticulum (ER) stress by OSMI-1 not only upregulated CHOP-DR5 signaling but also activated Jun-N-terminal kinase (JNK), resulting in a decrease in Bcl2 and the release of cytochrome c from mitochondria. TRAIL induced the activation of NF-κB and played a role in resistance as an antiapoptotic factor. During this process, O-GlcNAcylation of IκB kinase (IKK) and IκBα degradation occurred, followed by translocation of p65 into the nucleus. However, combination treatment with OSMI-1 counteracted the effect of TRAIL-mediated NF-κB signaling, resulting in a more synergistic effect on apoptosis. Therefore, the combined treatment of OSMI-1 and TRAIL synergistically increased TRAIL-induced apoptosis through caspase-8 activation. Conclusively, OSMI-1 potentially sensitizes TRAIL-induced cell death in HCT116 cells through the blockade of NF-κB signaling and activation of apoptosis through ER stress response.
Collapse
|
52
|
Chaudhary P, Sharma S, Singh R, Arya R. Elucidation of ER stress and UPR pathway in sialic acid-deficient cells: Pathological relevance to GNEM. J Cell Biochem 2021; 122:1886-1902. [PMID: 34555215 DOI: 10.1002/jcb.30148] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 09/01/2021] [Accepted: 09/07/2021] [Indexed: 12/31/2022]
Abstract
Accumulation of misfolded proteins in endoplasmic reticulum (ER) generates a stress condition in the cell. The cell combats ER stress by activating unfolded protein response (UPR) and ERAD (ER stress-associated degradation) pathway. Failure to restore favorable folding environment results in cell dysfunction and apoptosis. Various neurodegenerative disorders are characterized by the accumulation of misfolded protein, protein aggregates, and ER stress. GNE myopathy (GNEM) is a neuromuscular disorder pathologically characterized by rimmed vacuole formation due to the accumulation of protein aggregates. More than 200 mutations in key sialic acid biosynthetic enzyme UDP-N-acetylglucosamine 2-epimerase/N-acetylmannosamine kinase (GNE) have been identified worldwide in the muscle biopsies of GNE myopathy patients. However, the cellular and molecular pathomechanism leading to the disease ar poorly understood. In the present study, the phenomenon of ER stress has been elucidated in GNE mutant cells overexpressing GNE mutations of Indian origin. The effect of GNE mutations on activation of UPR signaling via inositol-requiring transmembrane kinase/endoribonuclease 1 (IRE-1), protein kinase RNA-like endoplasmic reticulum kinase (PERK), and activating transcription factor-6 (ATF6) were deciphered to understand the effect of GNE mutations on these proteins. GRP78 was upregulated with increased X-box-binding protein-1 (XBP-1) splicing and CCAAT/enhancer-binding protein (C/EBP) homologous protein (CHOP) upregulation leading to increased apoptosis of GNE mutant cells. Insulin-like growth factor 1 (IGF-1) ligand rescued the cells from apoptotic phenotype by supporting cell survival mechanism. Our study indicates a balance of cell death and survival that decides cell fate and offers potential therapeutic targets to combat ER stress in diseases associated with dysfunctional UPR pathway.
Collapse
Affiliation(s)
| | - Shweta Sharma
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Reema Singh
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Ranjana Arya
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India.,Special Centre for Systems Medicine, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
53
|
Methotrexate and theaflavin-3, 3'-digallate synergistically restore the balance between apoptosis and autophagy in synovial fibroblast of RA: an ex vivo approach with cultured human RA FLS. Inflammopharmacology 2021; 29:1427-1442. [PMID: 34350509 DOI: 10.1007/s10787-021-00857-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 07/26/2021] [Indexed: 02/05/2023]
Abstract
BACKGROUND Imbalance between apoptosis and autophagy in fibroblast-like synoviocytes (FLS) is one of the pathogenic mechanisms responsible for their abnormal proliferation in rheumatoid arthritis (RA). Methotrexate (MTX) demonstrated limited efficacy in amending this imbalance in fluid-derived (fd)-FLS. The active compound of black tea Theaflavin 3,3'-digallate (TF3) may be effective in restoring apoptosis-autophagy imbalance in (fd)-FLS. The combined effect of MTX + TF3 upon the same is yet to be elucidated. OBJECTIVE To evaluate the effect of MTX + TF3 on fd-FLS to induce apoptosis and inhibit autophagy through Endoplasmic Reticulum (ER) stress-mediated pathways. METHODS FLS from synovial fluid of 11 RA and 10 osteoarthritis patients were cultured after treatment with MTX/TF3 or a combination of MTX (125 nM) and TF3(10 µM) and the following parameters were evaluated. C-reactive protein, cytokines (TNF-α, IL-6), angiogenic markers were quantified by ELISA. fd-FLS viability was determined by MTT assay and apoptosis by flow cytometry. ER stress markers were estimated by RT-PCR (IRE1A, spliced-XBP-1) and immunoblotting (Grp78, Hsp70, CHOP, HIF-1α). Immunoblot studies were done to evaluate apoptotic (Bcl-2, Bax, Caspases) and autophagic (Beclin1, LC3b, p62) proteins. RESULTS MTX (IC25) and TF3 (IC50) both in single doses could down-regulate the levels of pro-inflammatory and angiogenic markers. Combinatorial treatment modulated autophagosomal proteins in fd-FLS and induced apoptosis by regulating ER stress response. CONCLUSION Disruption in homeostasis between apoptosis and autophagy in fd-FLS might be an underlying phenomenon in the progression of pathophysiology in RA. Co-administration of MTX + TF3 successfully restored the homeostasis by inducing apoptosis.
Collapse
|
54
|
Mutation in FBXO32 causes dilated cardiomyopathy through up-regulation of ER-stress mediated apoptosis. Commun Biol 2021; 4:884. [PMID: 34272480 PMCID: PMC8285540 DOI: 10.1038/s42003-021-02391-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 06/23/2021] [Indexed: 02/06/2023] Open
Abstract
Endoplasmic reticulum (ER) stress induction of cell death is implicated in cardiovascular diseases. Sustained activation of ER-stress induces the unfolded protein response (UPR) pathways, which in turn activate three major effector proteins. We previously reported a missense homozygous mutation in FBXO32 (MAFbx, Atrogin-1) causing advanced heart failure by impairing autophagy. In the present study, we performed transcriptional profiling and biochemical assays, which unexpectedly revealed a reduced activation of UPR effectors in patient mutant hearts, while a strong up-regulation of the CHOP transcription factor and of its target genes are observed. Expression of mutant FBXO32 in cells is sufficient to induce CHOP-associated apoptosis, to increase the ATF2 transcription factor and to impair ATF2 ubiquitination. ATF2 protein interacts with FBXO32 in the human heart and its expression is especially high in FBXO32 mutant hearts. These findings provide a new underlying mechanism for FBXO32-mediated cardiomyopathy, implicating abnormal activation of CHOP. These results suggest alternative non-canonical pathways of CHOP activation that could be considered to develop new therapeutic targets for the treatment of FBXO32-associated DCM. Al-Yacoub et al. investigate the consequences of FBXO32 mutation on dilated cardiomyopathy. ER stress, abnormal CHOP activation and CHOP-induced apoptosis with no UPR effector activation are found to underlie the FBXO32 mutation induced cardiomyopathy, suggesting an alternative pathway that can be considered to develop new therapeutic targets for its treatment.
Collapse
|
55
|
Moustardas P, Yamada-Fowler N, Apostolou E, Tzioufas AG, Turkina MV, Spyrou G. Deregulation of the Kallikrein Protease Family in the Salivary Glands of the Sjögren's Syndrome ERdj5 Knockout Mouse Model. Front Immunol 2021; 12:693911. [PMID: 34305928 PMCID: PMC8292930 DOI: 10.3389/fimmu.2021.693911] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 06/11/2021] [Indexed: 12/16/2022] Open
Abstract
Introduction The purpose of this study was to identify differentially expressed proteins in salivary glands of the ERdj5 knockout mouse model for Sjögren's syndrome and to elucidate possible mechanisms for the morbid phenotype development. At the same time, we describe for the first time the sexual dimorphism of the murine submandibular salivary gland at the proteome level. Methods We performed Liquid Chromatography/Mass Spectrometry in salivary gland tissues from both sexes of ERdj5 knockout and 129SV wildtype mice. The resulting list of proteins was evaluated with bioinformatic analysis and selected proteins were validated by western blot and immunohistochemistry and further analyzed at the transcription level by qRT-PCR. Results We identified 88 deregulated proteins in females, and 55 in males in wildtype vs knockout comparisons. In both sexes, Kallikrein 1b22 was highly upregulated (fold change>25, ANOVA p<0.0001), while all other proteases of this family were either downregulated or not significantly affected by the genotype. Bioinformatic analysis revealed a possible connection with the downregulated NGF that was further validated by independent methods. Concurrently, we identified 416 proteins that were significantly different in the salivary gland proteome of wildtype female vs male mice and highlighted pathways that could be driving the strong female bias of the pathology. Conclusion Our research provides a list of novel targets and supports the involvement of an NGF-mediating proteolytic deregulation pathway as a focus point towards the better understanding of the underlying mechanism of Sjögren's syndrome.
Collapse
Affiliation(s)
- Petros Moustardas
- Department of Biomedical and Clinical Sciences, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
- Center for Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Naomi Yamada-Fowler
- Department of Biomedical and Clinical Sciences, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
| | - Eirini Apostolou
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Athanasios G. Tzioufas
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria V. Turkina
- Department of Biomedical and Clinical Sciences, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
| | - Giannis Spyrou
- Department of Biomedical and Clinical Sciences, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
56
|
Abuaita BH, Sule GJ, Schultz TL, Gao F, Knight JS, O'Riordan MX. The IRE1α Stress Signaling Axis Is a Key Regulator of Neutrophil Antimicrobial Effector Function. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 207:210-220. [PMID: 34145058 DOI: 10.4049/jimmunol.2001321] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 04/26/2021] [Indexed: 12/12/2022]
Abstract
Activation of the endoplasmic reticulum stress sensor, IRE1α, is required for effective immune responses against bacterial infection and is associated with human inflammatory diseases in which neutrophils are a key immune component. However, the specific role of IRE1α in regulating neutrophil effector function has not been studied. In this study, we show that infection-induced IRE1α activation licenses neutrophil antimicrobial capacity, including IL-1β production, formation of neutrophil extracellular traps (NETs), and methicillin-resistant Staphylococcus aureus (MRSA) killing. Inhibition of IRE1α diminished production of mitochondrial reactive oxygen species and decreased CASPASE-2 activation, which both contributed to neutrophil antimicrobial activity. Mice deficient in CASPASE-2 or neutrophil IRE1α were highly susceptible to MRSA infection and failed to effectively form NETs in the s.c. abscess. IRE1α activation enhanced calcium influx and citrullination of histone H3 independently of mitochondrial reactive oxygen species production, suggesting that IRE1α coordinates multiple pathways required for NET formation. Our data demonstrate that the IRE1α-CASPASE-2 axis is a major driver of neutrophil activity against MRSA infection and highlight the importance of IRE1α in neutrophil antibacterial function.
Collapse
Affiliation(s)
- Basel H Abuaita
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI; and
| | - Gautam J Sule
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI
| | - Tracey L Schultz
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI; and
| | - Fushan Gao
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI; and
| | - Jason S Knight
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI
| | - Mary X O'Riordan
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI; and
| |
Collapse
|
57
|
Rahman S, Kumar V, Kumar A, Abdullah TS, Rather IA, Jan AT. Molecular Perspective of Nanoparticle Mediated Therapeutic Targeting in Breast Cancer: An Odyssey of Endoplasmic Reticulum Unfolded Protein Response (UPR ER) and Beyond. Biomedicines 2021; 9:biomedicines9060635. [PMID: 34199484 PMCID: PMC8229605 DOI: 10.3390/biomedicines9060635] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/26/2021] [Accepted: 05/28/2021] [Indexed: 12/21/2022] Open
Abstract
Breast cancer (BC) is the second most frequent cause of death among women. Representing a complex and heterogeneous type of cancer, its occurrence is attributed by both genetic (gene mutations, e.g., BRCA1, BRCA2) and non-genetic (race, ethnicity, etc.) risk factors. The effectiveness of available treatment regimens (small molecules, cytotoxic agents, and inhibitors) decreased due to their poor penetration across biological barriers, limited targeting, and rapid body clearance along with their effect on normal resident cells of bone marrow, gastrointestinal tract, and hair follicles. This significantly reduced their clinical outcomes, which led to an unprecedented increase in the number of cases worldwide. Nanomedicine, a nano-formulation of therapeutics, emerged as a versatile delivering module for employment in achieving the effective and target specific delivery of pharmaceutical payloads. Adoption of nanotechnological approaches in delivering therapeutic molecules to target cells ensures not only reduced immune response and toxicity, but increases the stability of therapeutic entities in the systemic circulation that averts their degradation and as such increased extravasations and accumulation via enhanced permeation and the retention (EPR) effect in target tissues. Additionally, nanoparticle (NP)-induced ER stress, which enhances apoptosis and autophagy, has been utilized as a combative strategy in the treatment of cancerous cells. As nanoparticles-based avenues have been capitalized to achieve better efficacy of the new genera of therapeutics with enhanced specificity and safety, the present study is aimed at providing the fundamentals of BC, nanotechnological modules (organic, inorganic, and hybrid) employed in delivering different therapeutic molecules, and mechanistic insights of nano-ER stress induced apoptosis and autophagy with a perspective of exploring this avenue for use in the nano-toxicological studies. Furthermore, the current scenario of USA FDA approved nano-formulations and the future perspective of nanotechnological based interventions to overcome the existing challenges are also discussed.
Collapse
Affiliation(s)
- Safikur Rahman
- Department of Botany, Munshi Singh College, BR Ambedkar Bihar University, Muzaffarpur 845401, India;
| | - Vijay Kumar
- Department of Biotechnology, Yeungnam University, Gyeongsan 38541, Korea;
| | - Anuj Kumar
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Korea;
| | - Tasduq S. Abdullah
- Council of Scientific and Industrial Research–Indian Institute of Integrative Medicine (CSIR–IIIM), Jammu 180001, India;
| | - Irfan A. Rather
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University (KAU), P.O. Box 80141, Jeddah 21589, Saudi Arabia
- Correspondence: (I.A.R.); (A.T.J.)
| | - Arif Tasleem Jan
- School of Biosciences and Biotechnology, Baba Ghulam Shah Badshah University, Rajouri 185234, India
- Correspondence: (I.A.R.); (A.T.J.)
| |
Collapse
|
58
|
Effects of Long-Term DHA Supplementation and Physical Exercise on Non-Alcoholic Fatty Liver Development in Obese Aged Female Mice. Nutrients 2021; 13:nu13020501. [PMID: 33546405 PMCID: PMC7913512 DOI: 10.3390/nu13020501] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/22/2021] [Accepted: 01/26/2021] [Indexed: 02/06/2023] Open
Abstract
Obesity and aging are associated to non-alcoholic fatty liver disease (NAFLD) development. Here, we investigate whether long-term feeding with a docosahexaenoic acid (DHA)-enriched diet and aerobic exercise, alone or in combination, are effective in ameliorating NAFLD in aged obese mice. Two-month-old female C57BL/6J mice received control or high fat diet (HFD) for 4 months. Then, the diet-induced obese (DIO) mice were distributed into four groups: DIO, DIO + DHA (15% dietary lipids replaced by a DHA-rich concentrate), DIO + EX (treadmill running), and DIO + DHA + EX up to 18 months. The DHA-rich diet reduced liver steatosis in DIO mice, decreasing lipogenic genes (Dgat2, Scd1, Srebp1c), and upregulated lipid catabolism genes (Hsl/Acox) expression. A similar pattern was observed in the DIO + EX group. The combination of DHA + exercise potentiated an increase in Cpt1a and Ppara genes, and AMPK activation, key regulators of fatty acid oxidation. Exercise, alone or in combination with DHA, significantly reversed the induction of proinflammatory genes (Mcp1, Il6, Tnfα, Tlr4) in DIO mice. DHA supplementation was effective in preventing the alterations induced by the HFD in endoplasmic reticulum stress-related genes (Ern1/Xbp1) and autophagy markers (LC3II/I ratio, p62, Atg7). In summary, long-term DHA supplementation and/or exercise could be helpful to delay NAFLD progression during aging in obesity.
Collapse
|
59
|
Kumar V, Maity S. ER Stress-Sensor Proteins and ER-Mitochondrial Crosstalk-Signaling Beyond (ER) Stress Response. Biomolecules 2021; 11:173. [PMID: 33525374 PMCID: PMC7911976 DOI: 10.3390/biom11020173] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/14/2021] [Accepted: 01/19/2021] [Indexed: 02/07/2023] Open
Abstract
Recent studies undoubtedly show the importance of inter organellar connections to maintain cellular homeostasis. In normal physiological conditions or in the presence of cellular and environmental stress, each organelle responds alone or in coordination to maintain cellular function. The Endoplasmic reticulum (ER) and mitochondria are two important organelles with very specialized structural and functional properties. These two organelles are physically connected through very specialized proteins in the region called the mitochondria-associated ER membrane (MAM). The molecular foundation of this relationship is complex and involves not only ion homeostasis through the shuttling of calcium but also many structural and apoptotic proteins. IRE1alpha and PERK are known for their canonical function as an ER stress sensor controlling unfolded protein response during ER stress. The presence of these transmembrane proteins at the MAM indicates its potential involvement in other biological functions beyond ER stress signaling. Many recent studies have now focused on the non-canonical function of these sensors. In this review, we will focus on ER mitochondrial interdependence with special emphasis on the non-canonical role of ER stress sensors beyond ER stress.
Collapse
|
60
|
Silwal P, Paik S, Kim JK, Yoshimori T, Jo EK. Regulatory Mechanisms of Autophagy-Targeted Antimicrobial Therapeutics Against Mycobacterial Infection. Front Cell Infect Microbiol 2021; 11:633360. [PMID: 33828998 PMCID: PMC8019938 DOI: 10.3389/fcimb.2021.633360] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 01/08/2021] [Indexed: 01/25/2023] Open
Abstract
Mycobacterium tuberculosis (Mtb) is an intracellular pathogen causing human tuberculosis, an infectious disease that still remains as a global health problem. Autophagy, a lysosomal degradative process, has emerged as a critical pathway to restrict intracellular Mtb growth through enhancement of phagosomal maturation. Indeed, several autophagy-modulating agents show promise as host-directed therapeutics for Mtb infection. In this Review, we discuss recent progress in our understanding the molecular mechanisms underlying the action of autophagy-modulating agents to overcome the immune escape strategies mediated by Mtb. The factors and pathways that govern such mechanisms include adenosine 5'-monophosphate-activated protein kinase, Akt/mammalian TOR kinase, Wnt signaling, transcription factor EB, cathelicidins, inflammation, endoplasmic reticulum stress, and autophagy-related genes. A further understanding of these mechanisms will facilitate the development of host-directed therapies against tuberculosis as well as infections with other intracellular bacteria targeted by autophagic degradation.
Collapse
Affiliation(s)
- Prashanta Silwal
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, South Korea
- Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Seungwha Paik
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, South Korea
- Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Jin Kyung Kim
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, South Korea
- Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Tamotsu Yoshimori
- Department of Genetics, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Intracellular Membrane Dynamics, Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Eun-Kyeong Jo
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, South Korea
- Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, South Korea
- *Correspondence: Eun-Kyeong Jo,
| |
Collapse
|
61
|
Cistanches deserticola PhG-RE through Inhibiting ERS Apoptosis Mechanism to Protect Myocardial Cell Apoptosis from H 2O 2-Induced Endoplasmic Reticulum Stress. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:8219296. [PMID: 33062023 PMCID: PMC7533003 DOI: 10.1155/2020/8219296] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/15/2020] [Accepted: 07/24/2020] [Indexed: 02/06/2023]
Abstract
The herb Cistanche deserticola has some myocardial protective effects. This study attempted to explain the mechanism by which PhG-RE protects myocardial cells and verify if this protection occurs through regulating the apoptosis mechanism associated with endoplasmic reticulum stress (ERS). Rat myocardial cells were exposed to 150 μg·mL−1 PhG-RE for 24 h and then to 100 μmol·mL−1 H2O2 for 18 h to induce ERS and establish a cell damage model. Thapsigargin (TG), a specific ERS activator, and 4-phenylbutyric acid (4-PBA), an ERS inhibitor, were used to validate the accuracy of the experiment. Our results demonstrated that PhG-RE significantly improved cell viability, protected cells, and reduced cell damage and apoptosis. PhG-RE played a role similar to that of the ERS inhibitor 4-PBA in protecting myocardial cells against apoptosis and damage induced by ER stress. Furthermore, PhG-RE significantly attenuated the mRNA expression of the ERS-associated apoptotic factors GRP78, CHOP, and Caspase-12 and the protein expression of the ERS-associated apoptotic factors GRP78, CHOP, Caspase-12, and p-JNK. Taken together, these findings suggest that PhG-RE can effectively protect myocardial cells and reduce cell apoptosis and damage, which may be related to the regulation of ERS-associated apoptosis.
Collapse
|
62
|
Li W, Cao T, Luo C, Cai J, Zhou X, Xiao X, Liu S. Crosstalk between ER stress, NLRP3 inflammasome, and inflammation. Appl Microbiol Biotechnol 2020; 104:6129-6140. [PMID: 32447438 DOI: 10.1007/s00253-020-10614-y] [Citation(s) in RCA: 177] [Impact Index Per Article: 35.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 03/31/2020] [Accepted: 04/05/2020] [Indexed: 12/17/2022]
Abstract
Endoplasmic reticulum stress (ERS) is a protective response to restore protein homeostasis by activating the unfolded protein response (UPR). However, UPR can trigger cell death under severe and/or persistently high ERS. The NLRP3 inflammasome is a complex of multiple proteins that activates the secretion of the proinflammatory cytokine IL-1β in a caspase-1-dependent manner to participate in the regulation of inflammation. The NLRP3 inflammasome involvement in ERS-induced inflammation has not been completely described. The intersection of ERS with multiple inflammatory pathways can initiate and aggravate chronic diseases. Accumulating evidence suggests that ERS-induced activation of NLRP3 inflammasome is the pathological basis of various inflammatory diseases. In this review, we have discussed the networks between ERS and NLRP3 inflammasome, with the view to identifying novel therapeutic targets in inflammatory diseases. KEY POINTS: • Endoplasmic reticulum stress (ERS) is an important factor for the activation of the NLRP3 inflammasomes that results in pathological processes. • ERS can activate the NLRP3 inflammasome to induce inflammatory responses via oxidative stress, calcium homeostasis, and NF-κB activation. • The interactions between ERS and NLRP3 inflammasome are associated with inflammation, which represent a potential therapeutic opportunity of inflammatory diseases.
Collapse
Affiliation(s)
- Wei Li
- Department of Clinical Laboratory, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Ting Cao
- Department of Clinical Laboratory, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Chunyi Luo
- Department of Clinical Laboratory, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Jialun Cai
- Department of Clinical Laboratory, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Xiangping Zhou
- Department of Clinical Laboratory, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Xinhua Xiao
- Department of Endocrinology, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China.
| | - Shuangquan Liu
- Department of Clinical Laboratory, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China.
| |
Collapse
|
63
|
Kuss-Duerkop SK, Keestra-Gounder AM. NOD1 and NOD2 Activation by Diverse Stimuli: a Possible Role for Sensing Pathogen-Induced Endoplasmic Reticulum Stress. Infect Immun 2020; 88:e00898-19. [PMID: 32229616 PMCID: PMC7309630 DOI: 10.1128/iai.00898-19] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Prompt recognition of microbes by cells is critical to eliminate invading pathogens. Some cell-associated pattern recognition receptors (PRRs) recognize and respond to microbial ligands. However, others can respond to cellular perturbations, such as damage-associated molecular patterns (DAMPs). Nucleotide oligomerization domains 1 and 2 (NOD1/2) are PRRs that recognize and respond to multiple stimuli of microbial and cellular origin, such as bacterial peptidoglycan, viral infections, parasitic infections, activated Rho GTPases, and endoplasmic reticulum (ER) stress. How NOD1/2 are stimulated by such diverse stimuli is not fully understood but may partly rely on cellular changes during infection that result in ER stress. NOD1/2 are ER stress sensors that facilitate proinflammatory responses for pathogen clearance; thus, NOD1/2 may help mount broad antimicrobial responses through detection of ER stress, which is often induced during a variety of infections. Some pathogens may subvert this response to promote infection through manipulation of NOD1/2 responses to ER stress that lead to apoptosis. Here, we review NOD1/2 stimuli and cellular responses. Furthermore, we discuss pathogen-induced ER stress and how it might potentiate NOD1/2 signaling.
Collapse
Affiliation(s)
- Sharon K Kuss-Duerkop
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - A Marijke Keestra-Gounder
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| |
Collapse
|
64
|
Batista A, Rodvold JJ, Xian S, Searles SC, Lew A, Iwawaki T, Almanza G, Waller TC, Lin J, Jepsen K, Carter H, Zanetti M. IRE1α regulates macrophage polarization, PD-L1 expression, and tumor survival. PLoS Biol 2020; 18:e3000687. [PMID: 32520957 PMCID: PMC7307794 DOI: 10.1371/journal.pbio.3000687] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 06/22/2020] [Accepted: 05/20/2020] [Indexed: 12/14/2022] Open
Abstract
In the tumor microenvironment, local immune dysregulation is driven in part by macrophages and dendritic cells that are polarized to a mixed proinflammatory/immune-suppressive phenotype. The unfolded protein response (UPR) is emerging as the possible origin of these events. Here we report that the inositol-requiring enzyme 1 (IRE1α) branch of the UPR is directly involved in the polarization of macrophages in vitro and in vivo, including the up-regulation of interleukin 6 (IL-6), IL-23, Arginase1, as well as surface expression of CD86 and programmed death ligand 1 (PD-L1). Macrophages in which the IRE1α/X-box binding protein 1 (Xbp1) axis is blocked pharmacologically or deleted genetically have significantly reduced polarization and CD86 and PD-L1 expression, which was induced independent of IFNγ signaling, suggesting a novel mechanism in PD-L1 regulation in macrophages. Mice with IRE1α- but not Xbp1-deficient macrophages showed greater survival than controls when implanted with B16.F10 melanoma cells. Remarkably, we found a significant association between the IRE1α gene signature and CD274 gene expression in tumor-infiltrating macrophages in humans. RNA sequencing (RNASeq) analysis showed that bone marrow-derived macrophages with IRE1α deletion lose the integrity of the gene connectivity characteristic of regulated IRE1α-dependent decay (RIDD) and the ability to activate CD274 gene expression. Thus, the IRE1α/Xbp1 axis drives the polarization of macrophages in the tumor microenvironment initiating a complex immune dysregulation leading to failure of local immune surveillance.
Collapse
Affiliation(s)
- Alyssa Batista
- The Laboratory of Immunology, Department of Medicine and Moores Cancer Center, University of California, San Diego, La Jolla, California, United States of America
| | - Jeffrey J. Rodvold
- The Laboratory of Immunology, Department of Medicine and Moores Cancer Center, University of California, San Diego, La Jolla, California, United States of America
| | - Su Xian
- Division of Medical Genetics; Department of Medicine, and Bioinformatics and Systems Biology Program, University of California San Diego, La Jolla, California, United States of America
| | - Stephen C. Searles
- The Laboratory of Immunology, Department of Medicine and Moores Cancer Center, University of California, San Diego, La Jolla, California, United States of America
| | - Alyssa Lew
- The Laboratory of Immunology, Department of Medicine and Moores Cancer Center, University of California, San Diego, La Jolla, California, United States of America
| | - Takao Iwawaki
- Laboratory for Cell Recovery Mechanisms, Brain Science Institute, RIKEN, Hirosawa, Japan
| | - Gonzalo Almanza
- The Laboratory of Immunology, Department of Medicine and Moores Cancer Center, University of California, San Diego, La Jolla, California, United States of America
| | - T. Cameron Waller
- Division of Medical Genetics; Department of Medicine, and Bioinformatics and Systems Biology Program, University of California San Diego, La Jolla, California, United States of America
| | - Jonathan Lin
- Department of Pathology, Stanford University, Palo Alto, California, United States of America
| | - Kristen Jepsen
- IGM Genomics Center, University of California, San Diego, La Jolla, California, United States of America
| | - Hannah Carter
- Division of Medical Genetics; Department of Medicine, and Bioinformatics and Systems Biology Program, University of California San Diego, La Jolla, California, United States of America
| | - Maurizio Zanetti
- The Laboratory of Immunology, Department of Medicine and Moores Cancer Center, University of California, San Diego, La Jolla, California, United States of America
| |
Collapse
|
65
|
Tong Y, Yang L, Shao F, Yan X, Li X, Huang G, Xiao Y, Zhou Z. Distinct secretion pattern of serum proinsulin in different types of diabetes. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:452. [PMID: 32395496 PMCID: PMC7210169 DOI: 10.21037/atm.2020.03.189] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Background Latent autoimmune diabetes in adults (LADA) is characterized by autoimmunity, late-onset and intermediate beta-cell deprivation rate between type 2 diabetes mellitus (T2DM) and type 1 diabetes mellitus (T1DM). Herein, we investigated proinsulin (PI) secretion patterns and the endoplasmic reticulum (ER) dysfunction biomarker, PI-to-C-peptide (PI:CP) ratio, to elucidate beta-cell intrinsic pathogenesis mechanisms in different types of diabetes. Methods Total serum fasting PI (FPI) were measured in adult-onset and newly-diagnosed diabetes patients, including 60 T1DM, 60 LADA and 60 T2DM. Thirty of each type underwent mixed meal tolerance tests (MMTTs), and hence 120 min postprandial PI (PPI) were detected. PI:CP ratio = PI (pmol/L) ÷ CP (pmol/L) × 100%. PI-related measurements among types of diabetes were compared. Correlation between PI-related measurements and beta-cell autoimmunity were analyzed. The possibility of discriminating LADA from T1DM and T2DM with PI-related measurements were tested. Results FPI and PPI were significantly higher in LADA than T1DM (P<0.001 for both comparisons), but lower than those in T2DM (P<0.001 and P=0.026, respectively). Fasting PI:CP ratio was significantly higher in T1DM than both LADA and T2DM (median 3.25% vs. 2.13% and 2.32%, P=0.011 and P=0.017, respectively). In LADA, positive autoantibody numbers increased by both fasting and postprandial PI:CP ratio (P=0.007 and P=0.034, respectively). Areas under receiver operation characteristic curves (AUCROC) of FPI and PPI for discriminating LADA from adult-onset T1DM were 0.751 (P<0.001) and 0.838 (P<0.001), respectively. Between LADA and T2DM, AUCROC of FPI and PPI were 0.685 (P<0.001) and 0.741 (P=0.001), respectively. Conclusions In the development of autoimmune diabetes, interplays between ER stress and beta-cell autoimmunity are potentially responsible for severer beta-cell destruction. PI-related measurements could help in differentiating LADA from adult-onset T1DM and T2DM.
Collapse
Affiliation(s)
- Yue Tong
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha 410011, China.,National Clinical Research Center for Metabolic Diseases, Changsha 410011, China.,Key Laboratory of Diabetes Immunology, Central South University, Ministry of Education, Changsha 410011, China
| | - Lin Yang
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha 410011, China.,National Clinical Research Center for Metabolic Diseases, Changsha 410011, China.,Key Laboratory of Diabetes Immunology, Central South University, Ministry of Education, Changsha 410011, China
| | - Feng Shao
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha 410011, China.,National Clinical Research Center for Metabolic Diseases, Changsha 410011, China.,Key Laboratory of Diabetes Immunology, Central South University, Ministry of Education, Changsha 410011, China
| | - Xiang Yan
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha 410011, China.,National Clinical Research Center for Metabolic Diseases, Changsha 410011, China.,Key Laboratory of Diabetes Immunology, Central South University, Ministry of Education, Changsha 410011, China
| | - Xia Li
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha 410011, China.,National Clinical Research Center for Metabolic Diseases, Changsha 410011, China.,Key Laboratory of Diabetes Immunology, Central South University, Ministry of Education, Changsha 410011, China
| | - Gan Huang
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha 410011, China.,National Clinical Research Center for Metabolic Diseases, Changsha 410011, China.,Key Laboratory of Diabetes Immunology, Central South University, Ministry of Education, Changsha 410011, China
| | - Yang Xiao
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha 410011, China.,National Clinical Research Center for Metabolic Diseases, Changsha 410011, China.,Key Laboratory of Diabetes Immunology, Central South University, Ministry of Education, Changsha 410011, China
| | - Zhiguang Zhou
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha 410011, China.,National Clinical Research Center for Metabolic Diseases, Changsha 410011, China.,Key Laboratory of Diabetes Immunology, Central South University, Ministry of Education, Changsha 410011, China
| |
Collapse
|
66
|
Li A, Song NJ, Riesenberg BP, Li Z. The Emerging Roles of Endoplasmic Reticulum Stress in Balancing Immunity and Tolerance in Health and Diseases: Mechanisms and Opportunities. Front Immunol 2020; 10:3154. [PMID: 32117210 PMCID: PMC7026265 DOI: 10.3389/fimmu.2019.03154] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 12/30/2019] [Indexed: 12/14/2022] Open
Abstract
The endoplasmic reticulum (ER) is an organelle equipped with mechanisms for proper protein folding, trafficking, and degradation to maintain protein homeostasis in the secretory pathway. As a defense mechanism, perturbation of ER proteostasis by ER stress agents activates a cascade of signaling pathways from the ER to the nucleus known as unfolded protein response (UPR). The primary goal of UPR is to induce transcriptional and translational programs to restore ER homeostasis for cell survival. As such, defects in UPR signaling have been implicated as a key contributor to multiple diseases including metabolic diseases, degenerative diseases, inflammatory disorders, and cancer. Growing evidence support the critical role of ER stress in regulating the fate as well as the magnitude of the immune response. Moreover, the availability of multiple UPR pharmacological inhibitors raises the hope that targeting UPR can be a new strategy for immune modulation and immunotherapy of diseases. This paper reviews the principal mechanisms by which ER stress affects immune cell biology and function, with a focus of discussion on UPR-associated immunopathology and the development of potential ER stress-targeted therapeutics.
Collapse
Affiliation(s)
- Anqi Li
- College of Medicine, The Ohio State University, Columbus, OH, United States.,The James Comprehensive Cancer Center, Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, OH, United States
| | - No-Joon Song
- The James Comprehensive Cancer Center, Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, OH, United States
| | - Brian P Riesenberg
- The James Comprehensive Cancer Center, Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, OH, United States
| | - Zihai Li
- College of Medicine, The Ohio State University, Columbus, OH, United States.,The James Comprehensive Cancer Center, Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, OH, United States.,Division of Medical Oncology, Department of Medicine, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
67
|
Ding C, Dou M, Wang Y, Li Y, Wang Y, Zheng J, Li X, Xue W, Ding X, Tian P. miR-124/IRE-1α affects renal ischemia/reperfusion injury by regulating endoplasmic reticulum stress in renal tubular epithelial cells. Acta Biochim Biophys Sin (Shanghai) 2020; 52:160-167. [PMID: 31965139 DOI: 10.1093/abbs/gmz150] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 10/24/2019] [Accepted: 11/25/2019] [Indexed: 12/11/2022] Open
Abstract
Acute kidney injury (AKI) refers to a clinical syndrome that occurs as a result of a rapid decline in renal function caused by multiple factors. Renal ischemia/reperfusion (I/R) injury is one of the main causes of AKI and has a high incidence and mortality. However, the specific pathogenesis of renal I/R injury is still unclear. In recent years, a major breakthrough has been made in the study of endoplasmic reticulum stress (ERS)-mediated apoptosis in I/R injury. It has been reported that miRNAs play protective roles in ischemic/reperfused organs, but the molecular mechanisms have not been investigated deeply. In this study, the renal I/R mouse model was used to explore the roles of miR-124 in ERS and in renal I/R injury. The western blot results showed that the expression levels of ERS-related proteins IRE-1α, XBP-1, and glucose-regulated protein 78 (GRP78) were significantly increased in the I/R model group when compared with those in the control group. Meanwhile, qPCR results showed that miR-124 expression was decreased in the I/R injury model, and overexpression of miR-124 using miR-124 mimics effectively reduced the expression of ERS-related proteins and alleviated renal I/R injury. In addition, luciferase reporter assay was performed, and the results showed that IRE-1α and miR-124 may have direct interaction. In conclusion, our data indicated that miR-124 was a negative regulator of ERS via binding to IRE-1α, ultimately conferring its protective effect on the kidney, which demonstrates the regulatory mechanism of miR-124 in renal I/R injury and provides new ideas and methods for the prevention and treatment of renal I/R injury.
Collapse
Affiliation(s)
- Chenguang Ding
- Department of Kidney Transplantation, Nephropathy Hospital, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
- Institute of Organ Transplantation, Xi’an Jiaotong University, Xi’an 710061, China
| | - Meng Dou
- Department of Kidney Transplantation, Nephropathy Hospital, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
- Institute of Organ Transplantation, Xi’an Jiaotong University, Xi’an 710061, China
| | - Yuxiang Wang
- Department of Kidney Transplantation, Nephropathy Hospital, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
- Institute of Organ Transplantation, Xi’an Jiaotong University, Xi’an 710061, China
| | - Yang Li
- Department of Kidney Transplantation, Nephropathy Hospital, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
- Institute of Organ Transplantation, Xi’an Jiaotong University, Xi’an 710061, China
| | - Ying Wang
- Department of Kidney Transplantation, Nephropathy Hospital, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
- Institute of Organ Transplantation, Xi’an Jiaotong University, Xi’an 710061, China
| | - Jin Zheng
- Department of Kidney Transplantation, Nephropathy Hospital, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
- Institute of Organ Transplantation, Xi’an Jiaotong University, Xi’an 710061, China
| | - Xiao Li
- Department of Kidney Transplantation, Nephropathy Hospital, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
- Institute of Organ Transplantation, Xi’an Jiaotong University, Xi’an 710061, China
| | - Wujun Xue
- Department of Kidney Transplantation, Nephropathy Hospital, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
- Institute of Organ Transplantation, Xi’an Jiaotong University, Xi’an 710061, China
| | - Xiaoming Ding
- Department of Kidney Transplantation, Nephropathy Hospital, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
- Institute of Organ Transplantation, Xi’an Jiaotong University, Xi’an 710061, China
| | - Puxun Tian
- Department of Kidney Transplantation, Nephropathy Hospital, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
- Institute of Organ Transplantation, Xi’an Jiaotong University, Xi’an 710061, China
| |
Collapse
|
68
|
Cao ZH, Wu Z, Hu C, Zhang M, Wang WZ, Hu XB. Endoplasmic reticulum stress and destruction of pancreatic β cells in type 1 diabetes. Chin Med J (Engl) 2020; 133:68-73. [PMID: 31923106 PMCID: PMC7028193 DOI: 10.1097/cm9.0000000000000583] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Indexed: 02/07/2023] Open
Abstract
Type 1 diabetes (T1D) results from dysfunction of pancreatic islets β cells. Recent studies supported that endoplasmic reticulum (ER) stress takes an important role in pancreatic β cell excessive loss, resulting in T1D. Here, we aimed to review the relationship between ER stress and T1D. Additionally, we also reviewed the potential mechanisms underlying ER stress mediated T1D. Studies have shown that severe ER stress is directly involved in the pancreatic β cells destruction and pathogenesis of T1D. ER stress plays a key part in pancreatic β cells and T1D, which will help in developing new effective therapeutics for T1D.
Collapse
Affiliation(s)
- Zhao-Hui Cao
- Department of Biochemistry and Molecular Biology, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | | | | | | | | | | |
Collapse
|
69
|
Liu Q, Körner H, Wu H, Wei W. Endoplasmic reticulum stress in autoimmune diseases. Immunobiology 2019; 225:151881. [PMID: 31879042 DOI: 10.1016/j.imbio.2019.11.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 11/26/2019] [Indexed: 12/26/2022]
Abstract
If the body's immune system is disordered and begins to attack "self" and therefore, its own tissues this is considered to be an autoimmune pathology. The specific mechanisms vary between the different diseases and have not always been elucidated but chronic, non-resolving inflammation is a common theme in the pathogenesis of autoimmune diseases. Interestingly, it has been shown that development and occurrence of various inflammatory responses are closely correlated to endoplasmic reticulum stress. Therefore, this review discusses the current progress of research about the relationship between autoimmune diseases and endoplasmic reticulum stress, specifically the unfolded protein response (UPR).
Collapse
Affiliation(s)
- Qi Liu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation, China; Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, Anhui Province, China
| | - Heinrich Körner
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation, China; Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, Anhui Province, China
| | - Huaxun Wu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation, China; Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, Anhui Province, China.
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation, China; Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, Anhui Province, China.
| |
Collapse
|
70
|
Go D, Lee J, Choi JA, Cho SN, Kim SH, Son SH, Song CH. Reactive oxygen species-mediated endoplasmic reticulum stress response induces apoptosis of Mycobacterium avium-infected macrophages by activating regulated IRE1-dependent decay pathway. Cell Microbiol 2019; 21:e13094. [PMID: 31386788 PMCID: PMC6899680 DOI: 10.1111/cmi.13094] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 07/15/2019] [Accepted: 07/30/2019] [Indexed: 12/12/2022]
Abstract
Mycobacterium avium, a slow‐growing nontuberculous mycobacterium, causes fever, diarrhoea, loss of appetite, and weight loss in immunocompromised people. We have proposed that endoplasmic reticulum (ER) stress‐mediated apoptosis plays a critical role in removing intracellular mycobacteria. In the present study, we investigated the role of the regulated IRE1‐dependent decay (RIDD) pathway in macrophages during M. avium infection based on its role in the regulation of gene expression. The inositol‐requiring enzyme 1 (IRE1)/apoptosis signal‐regulating kinase 1 (ASK1)/c‐Jun N‐terminal kinase (JNK) signalling pathway was activated in macrophages after infection with M. avium. The expression of RIDD‐associated genes, such as Bloc1s1 and St3gal5, was decreased in M. avium‐infected macrophages. Interestingly, M. avium‐induced apoptosis was significantly suppressed by pretreatment with irestatin (inhibitor of IRE1α) and 4μ8c (RIDD blocker). Macrophages pretreated with N‐acetyl cysteine (NAC) showed decreased levels of reactive oxygen species (ROS), IRE1α, and apoptosis after M. avium infection. The expression of Bloc1s1 and St3gal5 was increased in NAC‐pretreated macrophages following infection with M. avium. Growth of M. avium was significantly increased in irestatin‐, 4μ8c‐, and NAC‐treated macrophages compared with the control. The data indicate that the ROS‐mediated ER stress response induces apoptosis of M. avium‐infected macrophages by activating IRE1α‐RIDD. Thus, activation of IRE1α suppresses the intracellular survival of M. avium in macrophages.
Collapse
Affiliation(s)
- Dam Go
- Department of Medical Science, Chungnam National University, Daejeon, South Korea.,Department of Microbiology, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Junghwan Lee
- Department of Medical Science, Chungnam National University, Daejeon, South Korea.,Department of Microbiology, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Ji-Ae Choi
- Department of Medical Science, Chungnam National University, Daejeon, South Korea.,Department of Microbiology, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Soo-Na Cho
- Department of Medical Science, Chungnam National University, Daejeon, South Korea.,Department of Microbiology, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Seon-Hwa Kim
- Department of Medical Science, Chungnam National University, Daejeon, South Korea.,Department of Microbiology, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Sang-Hun Son
- Department of Medical Science, Chungnam National University, Daejeon, South Korea.,Department of Microbiology, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Chang-Hwa Song
- Department of Medical Science, Chungnam National University, Daejeon, South Korea.,Department of Microbiology, College of Medicine, Chungnam National University, Daejeon, South Korea.,Research Institute for Medical Sciences, College of Medicine, Chungnam National University, Daejeon, South Korea
| |
Collapse
|
71
|
Ahmadiany M, Alavi-Samani M, Hashemi Z, Moosavi MA, Rahmati M. The Increased RNase Activity of IRE1α in PBMCs from Patients with Rheumatoid Arthritis. Adv Pharm Bull 2019; 9:505-509. [PMID: 31592115 PMCID: PMC6773928 DOI: 10.15171/apb.2019.060] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Revised: 05/30/2019] [Accepted: 06/01/2019] [Indexed: 12/17/2022] Open
Abstract
Purpose: Despite recent advances in the diagnosis and treatment of rheumatoid arthritis (RA), this
inflammatory disease remains a challenge to patients and physicians. Recent evidence highlights
the contribution of endoplasmic reticulum (ER) stress in the pathogenesis and treatment of RA.
Herein, we study the expression of the ER stress sensor inositol-requiring enzyme 1α (IRE1α),
as well as XBP1 splicing and the regulated IRE1-dependent decay (RIDD), in peripheral blood
mononuclear cells (PBMCs) from patients with RA compared with healthy controls.
Methods: The PBMCs from blood samples of RA patients and healthy volunteers were isolated
by a density gradient centrifugation method using Ficoll. The gene expression levels of
GRP78/
Bip, IRE1, XBP1s, micro-RNAs (miRNAs)
were evaluated by real-time PCR.
Results: The expression of GRP78, IRE1, and XBP1s were increased in PBMCs of RA patients
compared with healthy controls. We further show that the RIDD targets (miRNA-17, -34a, -96,
and -125b) were downregulated in RA samples.
Conclusion: This study can expand our knowledge on the importance of RNase activity of
IRE1α in RA and may offer new potentials for developing novel diagnostic and/or therapeutic
biomarkers.
Collapse
Affiliation(s)
- Mahdieh Ahmadiany
- Cancer Biology Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Biochemistry, Faculty of Advanced Sciences & Technology, Pharmaceutical Sciences Branch, Islamic Azad University, Tehran, Iran (IAUPS)
| | - Mahshid Alavi-Samani
- Cancer Biology Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Biochemistry, Faculty of Advanced Sciences & Technology, Pharmaceutical Sciences Branch, Islamic Azad University, Tehran, Iran (IAUPS)
| | - Zahra Hashemi
- Department of Rheumatology, Imam Hossein Teaching Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Amin Moosavi
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology, Tehran P.O Box 14965/161, Iran
| | - Marveh Rahmati
- Cancer Biology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
72
|
Shi W, Chen Z, Li L, Liu H, Zhang R, Cheng Q, Xu D, Wu L. Unravel the molecular mechanism of XBP1 in regulating the biology of cancer cells. J Cancer 2019; 10:2035-2046. [PMID: 31205564 PMCID: PMC6548171 DOI: 10.7150/jca.29421] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 04/02/2019] [Indexed: 02/07/2023] Open
Abstract
Cancer cells are usually exposed to stressful environments, such as hypoxia, nutrient deprivation, and other metabolic dysfunctional regulation, leading to continuous endoplasmic reticulum (ER) stress. As the most conserved branch among the three un-folded protein response (UPR) pathways, Inositol-requiring enzyme 1α (IRE1α)-X-box-binding protein 1 (XBP1) signaling has been implicated in cancer development and progression. Active XBP1 with transactivation domain functions as a transcription factor to regulate the expression of downstream target genes, including many oncogenic factors. The regulatory activity of XBP1 in cell proliferation, apoptosis, metastasis, and drug resistance promotes cell survival, leading to tumorigenesis and tumor progression. In addition, the XBP1 peptides-based vaccination and/or combination with immune-modulatory drug administration have been developed for effective management for several cancers. Potentially, XBP1 is the biomarker of cancer development and progression and the strategy for clinical cancer management.
Collapse
Affiliation(s)
- Weimei Shi
- College of Pharmacy, Gannan Medical University, Ganzhou China, 341000
| | - Zhixi Chen
- College of Pharmacy, Gannan Medical University, Ganzhou China, 341000
| | - Linfu Li
- College of Pharmacy, Gannan Medical University, Ganzhou China, 341000
| | - Hai Liu
- College of Pharmacy, Gannan Medical University, Ganzhou China, 341000
| | - Rui Zhang
- College of Pharmacy, Gannan Medical University, Ganzhou China, 341000
| | - Qilai Cheng
- College of Pharmacy, Gannan Medical University, Ganzhou China, 341000
| | - Daohua Xu
- Department of Pharmacology, Guangdong Medical University, Dongguan China, 523808
| | - Longhuo Wu
- College of Pharmacy, Gannan Medical University, Ganzhou China, 341000
| |
Collapse
|
73
|
Rahman S, Archana A, Jan AT, Dutta D, Shankar A, Kim J, Minakshi R. Molecular Insights Into the Relationship Between Autoimmune Thyroid Diseases and Breast Cancer: A Critical Perspective on Autoimmunity and ER Stress. Front Immunol 2019; 10:344. [PMID: 30881358 PMCID: PMC6405522 DOI: 10.3389/fimmu.2019.00344] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Accepted: 02/11/2019] [Indexed: 12/12/2022] Open
Abstract
The etiopathologies behind autoimmune thyroid diseases (AITDs) unravel misbehavior of immune components leading to the corruption of immune homeostasis where thyroid autoantigens turn foe to the self. In AITDs lymphocytic infiltration in the thyroid shows up a deranged immune system charging the follicular cells of the thyroid gland (thyrocytes) leading to the condition of either hyperthyroidism or hypothyroidism. The inflammation in AITDs consistently associate with ER function due to which disturbances in the ER protein homeostasis leads to unfolded protein response (UPR) that promotes pathogenesis of autoimmunity. The roles of ER stress in the instantaneous downregulation of MHC class I molecules on thyrocytes and the relevance of IFN γ in the pathogenesis of AITD has been well-documented. Thyroglobulin being the major target of autoantibodies in most of the AITDs is because of its unusual processing in the ER. Autoimmune disorders display a conglomeration of ER stress-induced UPR activated molecules. Several epidemiological data highlight the preponderance of AITDs in women as well as its concurrence with breast cancer. Both being an active glandular system displaying endocrine activity, thyroid as well as breast tissue show various commonalities in the expression pattern of heterogenous molecules that not only participate in the normal functioning but at the same time share the blame during disease establishment. Studies on the development and progression of breast carcinoma display a deranged and uncontrolled immune response, which is meticulously exploited during tumor metastasis. The molecular crosstalks between AITDs and breast tumor microenvironment rely on active participation of immune cells. The induction of ER stress by Tunicamycin advocates to provide a model for cancer therapy by intervening glycosylation. Therefore, this review attempts to showcase the molecules that are involved in feeding up the relationship between breast carcinoma and AITDs.
Collapse
Affiliation(s)
- Safikur Rahman
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| | - Ayyagari Archana
- Department of Microbiology, Swami Shraddhanand College, University of Delhi, New Delhi, India
| | - Arif Tasleem Jan
- School of Biosciences and Biotechnology, Baba Ghulam Shah Badshah University, Rajouri, India
| | - Durgashree Dutta
- Department of Biochemistry, Jan Nayak Chaudhary Devilal Dental College, Sirsa, India
| | - Abhishek Shankar
- Department of Preventive Oncology, Dr. B. R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Jihoe Kim
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| | - Rinki Minakshi
- Department of Microbiology, Swami Shraddhanand College, University of Delhi, New Delhi, India
| |
Collapse
|