1
|
Cao W, Zhang M, Wu N, Zheng Y, Li X, Han H, Yu T, Wu Z, Qu P, Li B. Comparative Transcriptome Analysis Elucidates the Desiccation Stress Adaptation in Sargassum muticum. Genes (Basel) 2025; 16:587. [PMID: 40428409 PMCID: PMC12111738 DOI: 10.3390/genes16050587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2025] [Revised: 05/10/2025] [Accepted: 05/15/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND/OBJECTIVES Desiccation profoundly influences the distribution and abundance of intertidal seaweeds, necessitating robust molecular adaptations. Sargassum muticum is a brown seaweed inhabiting intertidal rocky substrates. During low tides, this species undergoes periodic aerial exposure. Such environmental conditions necessitate robust physiological mechanisms to mitigate desiccation stress. Yet, the molecular basis of this adaptation remains poorly understood. METHODS To investigate desiccation-responsive genes and elucidate the underlying mechanisms of adaptation, we exposed S. muticum to 6 h of controlled desiccation stress in sterilized ceramic trays, simulating natural tidal conditions, and performed comparative transcriptome analysis using RNA-seq on the Illumina NovaSeq 6000 platform. RESULTS High-quality sequencing identified 66,192 unigenes, with 1990 differentially expressed genes (1399 upregulated and 591 downregulated). These differentially expressed genes (DEGs) were categorized into regulatory genes-including mitogen-activated protein kinase (MAPK), calmodulin, elongation factor, and serine/threonine-protein kinase-and functional genes, such as heat shock protein family members (HSP20, HSP40, and HSP70), tubulin (TUBA and TUBB), and endoplasmic reticulum homeostasis-related genes (protein disulfide-isomerase A6, calreticulin, and calnexin). Gene Ontology (GO) enrichment highlighted upregulated DEGs in metabolic processes like glutathione metabolism, critical for oxidative stress mitigation, while downregulated genes were linked to transport functions, such as ammonium transport, suggesting reduced nutrient uptake during dehydration. KEGG pathway analysis revealed significant enrichment in "protein processing in endoplasmic reticulum" and "MAPK signaling pathway-plant", implicating endoplasmic reticulum stress response and conserved signaling cascades in desiccation adaptation. Validation via qRT-PCR confirmed consistent expression trends for key genes, reinforcing the reliability of transcriptomic data. CONCLUSIONS These findings suggest that S. muticum undergoes extensive biological adjustments to mitigate desiccation stress, highlighting candidate pathways for future investigations into recovery and tolerance mechanisms.
Collapse
Affiliation(s)
- Wei Cao
- Island and Reef Fishery Research Center, Changdao Enhancement and Experiment Station, Chinese Academy of Fishery Science, Yantai 264000, China; (W.C.); (M.Z.); (N.W.); (H.H.); (T.Y.)
| | - Mingyi Zhang
- Island and Reef Fishery Research Center, Changdao Enhancement and Experiment Station, Chinese Academy of Fishery Science, Yantai 264000, China; (W.C.); (M.Z.); (N.W.); (H.H.); (T.Y.)
| | - Nan Wu
- Island and Reef Fishery Research Center, Changdao Enhancement and Experiment Station, Chinese Academy of Fishery Science, Yantai 264000, China; (W.C.); (M.Z.); (N.W.); (H.H.); (T.Y.)
| | - Yanxin Zheng
- Island and Reef Fishery Research Center, Changdao Enhancement and Experiment Station, Chinese Academy of Fishery Science, Yantai 264000, China; (W.C.); (M.Z.); (N.W.); (H.H.); (T.Y.)
- Observation and Research Station of Bohai Strait Eco-Corridor, MNR, Qingdao 266000, China;
| | - Xiaodong Li
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture (CAS), Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266000, China;
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao 266000, China
| | - Haiying Han
- Island and Reef Fishery Research Center, Changdao Enhancement and Experiment Station, Chinese Academy of Fishery Science, Yantai 264000, China; (W.C.); (M.Z.); (N.W.); (H.H.); (T.Y.)
| | - Tao Yu
- Island and Reef Fishery Research Center, Changdao Enhancement and Experiment Station, Chinese Academy of Fishery Science, Yantai 264000, China; (W.C.); (M.Z.); (N.W.); (H.H.); (T.Y.)
| | - Zhongxun Wu
- National Ocean Park Administration Center of Changdao, Yantai 264000, China;
| | - Pei Qu
- Observation and Research Station of Bohai Strait Eco-Corridor, MNR, Qingdao 266000, China;
| | - Bo Li
- Island and Reef Fishery Research Center, Changdao Enhancement and Experiment Station, Chinese Academy of Fishery Science, Yantai 264000, China; (W.C.); (M.Z.); (N.W.); (H.H.); (T.Y.)
| |
Collapse
|
2
|
Kettel P, Karagöz GE. Endoplasmic reticulum: Monitoring and maintaining protein and membrane homeostasis in the endoplasmic reticulum by the unfolded protein response. Int J Biochem Cell Biol 2024; 172:106598. [PMID: 38768891 DOI: 10.1016/j.biocel.2024.106598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/01/2024] [Accepted: 05/14/2024] [Indexed: 05/22/2024]
Abstract
The endoplasmic reticulum (ER) regulates essential cellular processes, including protein folding, lipid synthesis, and calcium homeostasis. The ER homeostasis is maintained by a conserved set of signaling cascades called the Unfolded Protein Response (UPR). How the UPR senses perturbations in ER homeostasis has been the subject of active research for decades. In metazoans, the UPR consists of three ER-membrane embedded sensors: IRE1, PERK and ATF6. These sensors detect the accumulation of misfolded proteins in the ER lumen and adjust protein folding capacity according to cellular needs. Early work revealed that the ER-resident chaperone BiP binds to all three UPR sensors in higher eukaryotes and BiP binding was suggested to regulate their activity. More recent data have shown that in higher eukaryotes the interaction of the UPR sensors with a complex network of chaperones and misfolded proteins modulates their activation and deactivation dynamics. Furthermore, emerging evidence suggests that the UPR monitors ER membrane integrity beyond protein folding defects. However, the mechanistic and structural basis of UPR activation by proteotoxic and lipid bilayer stress in higher eukaryotes remains only partially understood. Here, we review the current understanding of novel protein interaction networks and the contribution of the lipid membrane environment to UPR activation.
Collapse
Affiliation(s)
- Paulina Kettel
- Max Perutz Laboratories Vienna, Vienna BioCenter, Vienna, Austria; Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, Vienna, Austria
| | - G Elif Karagöz
- Max Perutz Laboratories Vienna, Vienna BioCenter, Vienna, Austria; Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
3
|
Chen CC, Hsu LW, Chen KD, Chiu KW, Kung CP, Li SR, Chen CL, Huang KT. Calreticulin regulates hepatic stellate cell activation through modulating TGF-beta-induced Smad signaling. Cell Calcium 2024; 121:102895. [PMID: 38703416 DOI: 10.1016/j.ceca.2024.102895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 04/25/2024] [Accepted: 04/27/2024] [Indexed: 05/06/2024]
Abstract
Liver fibrosis is characterized by excessive deposition of extracellular matrix (ECM) as a wound healing process. Activated hepatic stellate cells (HpSCs) are the major producer of the ECM and play a central role in liver fibrogenesis. It has been widely accepted that elimination of activated HpSCs or reversion to a quiescent state can be a feasible strategy for resolving the disease, further highlighting the urgent need for novel therapeutic targets. Calreticulin (CRT) is a molecular chaperone that normally resides in the endoplasmic reticulum (ER), important in protein folding and trafficking through the secretory pathway. CRT also plays a critical role in calcium (Ca2+) homeostasis, with its Ca2+ storage capacity. In the current study, we aimed to demonstrate its function in directing HpSC activation. In a mouse liver injury model, CRT was up-regulated in HpSCs. In cellular experiments, we further showed that this activation was through modulating the canonical TGF-β signaling. As down-regulation of CRT in HpSCs elevated intracellular Ca2+ levels through a form of Ca2+ influx, named store-operated Ca2+ entry (SOCE), we examined whether moderating SOCE affected TGF-β signaling. Interestingly, blocking SOCE had little effect on TGF-β-induced gene expression. In contrast, inhibition of ER Ca2+ release using the inositol trisphosphate receptor inhibitor 2-APB increased TGF-β signaling. Treatment with 2-APB did not alter SOCE but decreased intracellular Ca2+ at the basal level. Indeed, adjusting Ca2+ concentrations by EGTA or BAPTA-AM chelation further enhanced TGF-β-induced signaling. Our results suggest a crucial role of CRT in the liver fibrogenic process through modulating Ca2+ concentrations and TGF-β signaling in HpSCs, which may provide new information and help advance the current discoveries for liver fibrosis.
Collapse
Affiliation(s)
- Chien-Chih Chen
- Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan; School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Li-Wen Hsu
- Liver Transplantation Center, Department of General Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Kuang-Den Chen
- Liver Transplantation Center, Department of General Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan; Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - King-Wah Chiu
- Liver Transplantation Center, Department of General Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan; Division of Hepato-Gastroenterology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Chao-Pin Kung
- Liver Transplantation Center, Department of General Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan; Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Shu-Rong Li
- Liver Transplantation Center, Department of General Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Chao-Long Chen
- Liver Transplantation Center, Department of General Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Kuang-Tzu Huang
- Liver Transplantation Center, Department of General Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan; Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan.
| |
Collapse
|
4
|
Karamali N, Daraei A, Rostamlou A, Mahdavi R, Akbari Jonoush Z, Ghadiri N, Mahmoudi Z, Mardi A, Javidan M, Sohrabi S, Baradaran B. Decoding contextual crosstalk: revealing distinct interactions between non-coding RNAs and unfolded protein response in breast cancer. Cancer Cell Int 2024; 24:104. [PMID: 38468244 PMCID: PMC10926595 DOI: 10.1186/s12935-024-03296-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 03/06/2024] [Indexed: 03/13/2024] Open
Abstract
Breast cancer is significantly influenced by endoplasmic reticulum (ER) stress, impacting both its initiation and progression. When cells experience an accumulation of misfolded or unfolded proteins, they activate the unfolded protein response (UPR) to restore cellular balance. In breast cancer, the UPR is frequently triggered due to challenging conditions within tumors. The UPR has a dual impact on breast cancer. On one hand, it can contribute to tumor growth by enhancing cell survival and resistance to programmed cell death in unfavorable environments. On the other hand, prolonged and severe ER stress can trigger cell death mechanisms, limiting tumor progression. Furthermore, ER stress has been linked to the regulation of non-coding RNAs (ncRNAs) in breast cancer cells. These ncRNAs, including microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), play essential roles in cancer development by influencing gene expression and cellular processes. An improved understanding of how ER stress and ncRNAs interact in breast cancer can potentially lead to new treatment approaches. Modifying specific ncRNAs involved in the ER stress response might interfere with cancer cell survival and induce cell death. Additionally, focusing on UPR-associated proteins that interact with ncRNAs could offer novel therapeutic possibilities. Therefore, this review provides a concise overview of the interconnection between ER stress and ncRNAs in breast cancer, elucidating the nuanced effects of the UPR on cell fate and emphasizing the regulatory roles of ncRNAs in breast cancer progression.
Collapse
Affiliation(s)
- Negin Karamali
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Arshia Daraei
- Department of Immunology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Arman Rostamlou
- Department of Medical Biology, School of Medicine, University of EGE, Bornova, Izmir, Turkey
| | - Roya Mahdavi
- Student Research Committee, Ahvaz Jundishapur University of Medical Science, Ahvaz, Iran
- Department of Immunology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Zahra Akbari Jonoush
- Student Research Committee, Ahvaz Jundishapur University of Medical Science, Ahvaz, Iran
- Department of Immunology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Nooshin Ghadiri
- Student Research Committee, Ahvaz Jundishapur University of Medical Science, Ahvaz, Iran
- Department of Immunology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Zahra Mahmoudi
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amirhossein Mardi
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Moslem Javidan
- Student Research Committee, Ahvaz Jundishapur University of Medical Science, Ahvaz, Iran
- Department of Immunology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Sepideh Sohrabi
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
5
|
Cloots E, Guilbert P, Provost M, Neidhardt L, Van de Velde E, Fayazpour F, De Sutter D, Savvides SN, Eyckerman S, Janssens S. Activation of goblet-cell stress sensor IRE1β is controlled by the mucin chaperone AGR2. EMBO J 2024; 43:695-718. [PMID: 38177501 PMCID: PMC10907643 DOI: 10.1038/s44318-023-00015-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 11/13/2023] [Accepted: 11/15/2023] [Indexed: 01/06/2024] Open
Abstract
Intestinal goblet cells are secretory cells specialized in the production of mucins, and as such are challenged by the need for efficient protein folding. Goblet cells express Inositol-Requiring Enzyme-1β (IRE1β), a unique sensor in the unfolded protein response (UPR), which is part of an adaptive mechanism that regulates the demands of mucin production and secretion. However, how IRE1β activity is tuned to mucus folding load remains unknown. We identified the disulfide isomerase and mucin chaperone AGR2 as a goblet cell-specific protein that crucially regulates IRE1β-, but not IRE1α-mediated signaling. AGR2 binding to IRE1β disrupts IRE1β oligomerization, thereby blocking its downstream endonuclease activity. Depletion of endogenous AGR2 from goblet cells induces spontaneous IRE1β activation, suggesting that alterations in AGR2 availability in the endoplasmic reticulum set the threshold for IRE1β activation. We found that AGR2 mutants lacking their catalytic cysteine, or displaying the disease-associated mutation H117Y, were no longer able to dampen IRE1β activity. Collectively, these results demonstrate that AGR2 is a central chaperone regulating the goblet cell UPR by acting as a rheostat of IRE1β endonuclease activity.
Collapse
Affiliation(s)
- Eva Cloots
- Laboratory for ER stress and Inflammation, VIB Center for Inflammation Research, 9052, Ghent, Belgium
- Department of Pediatrics and Internal Medicine, Ghent University, 9052, Ghent, Belgium
| | - Phaedra Guilbert
- Laboratory for ER stress and Inflammation, VIB Center for Inflammation Research, 9052, Ghent, Belgium
- Department of Pediatrics and Internal Medicine, Ghent University, 9052, Ghent, Belgium
| | - Mathias Provost
- Unit for Structural Biology, VIB Center for Inflammation Research, 9052, Ghent, Belgium
- Unit for Structural Biology, Department of Biochemistry and Microbiology, 9052, Ghent, Belgium
| | - Lisa Neidhardt
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, CB2 0XY, UK
| | - Evelien Van de Velde
- Laboratory for ER stress and Inflammation, VIB Center for Inflammation Research, 9052, Ghent, Belgium
- Department of Pediatrics and Internal Medicine, Ghent University, 9052, Ghent, Belgium
| | - Farzaneh Fayazpour
- Laboratory for ER stress and Inflammation, VIB Center for Inflammation Research, 9052, Ghent, Belgium
- Department of Pediatrics and Internal Medicine, Ghent University, 9052, Ghent, Belgium
| | - Delphine De Sutter
- VIB Center for Medical Biotechnology, 9052, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, 9052, Ghent, Belgium
| | - Savvas N Savvides
- Unit for Structural Biology, VIB Center for Inflammation Research, 9052, Ghent, Belgium
- Unit for Structural Biology, Department of Biochemistry and Microbiology, 9052, Ghent, Belgium
| | - Sven Eyckerman
- VIB Center for Medical Biotechnology, 9052, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, 9052, Ghent, Belgium
| | - Sophie Janssens
- Laboratory for ER stress and Inflammation, VIB Center for Inflammation Research, 9052, Ghent, Belgium.
- Department of Pediatrics and Internal Medicine, Ghent University, 9052, Ghent, Belgium.
| |
Collapse
|
6
|
Wang G, Song S, Shen WB, Reece EA, Yang P. MicroRNA-322 overexpression reduces neural tube defects in diabetic pregnancies. Am J Obstet Gynecol 2024; 230:254.e1-254.e13. [PMID: 37531989 PMCID: PMC10828117 DOI: 10.1016/j.ajog.2023.07.048] [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: 03/28/2023] [Revised: 07/14/2023] [Accepted: 07/25/2023] [Indexed: 08/04/2023]
Abstract
BACKGROUND Hyperglycemia from pregestational diabetes mellitus induces neural tube defects in the developing fetus. Folate supplementation is the only effective way to prevent neural tube defects; however, some cases of neural tube defects are resistant to folate. Excess folate has been linked to higher maternal cancer risk and infant allergy. Therefore, additional interventions are needed. Understanding the mechanisms underlying maternal diabetes mellitus-induced neural tube defects can identify potential targets for preventing such defects. Despite not yet being in clinical use, growing evidence suggests that microRNAs are important intermediates in embryonic development and can serve as both biomarkers and drug targets for disease intervention. Our previous studies showed that maternal diabetes mellitus in vivo activates the inositol-requiring transmembrane kinase/endoribonuclease 1α (IRE1α) in the developing embryo and that a high glucose condition in vitro reduces microRNA-322 (miR-322) levels. IRE1α is an RNA endonuclease; however, it is unknown whether IRE1α targets and degrades miR-322 specifically or whether miR-322 degradation leads to neural tube defects via apoptosis. We hypothesize that IRE1α can inhibit miR-322 in maternal diabetes mellitus-induced neural tube defects and that restoring miR-322 expression in developing neuroepithelium ameliorates neural tube defects. OBJECTIVE This study aimed to identify potential targets for preventing maternal diabetes mellitus-induced neural tube defects and to investigate the roles and relationship of a microRNA and an RNA endonuclease in mouse embryos exposed to maternal diabetes mellitus. STUDY DESIGN To determine whether miR-322 reduction is necessary for neural tube defect formation in pregnancies complicated by diabetes mellitus, male mice carrying a transgene expressing miR-322 were mated with nondiabetic or diabetic wide-type female mice to generate embryos with or without miR-322 overexpression. At embryonic day 8.5 when the neural tube is not yet closed, embryos were harvested for the assessment of 3 miR-322 transcripts (primary, precursor, and mature miR-322), tumor necrosis factor receptor-associated factor 3 (TRAF3), and neuroepithelium cell survival. Neural tube defect incidences were determined in embryonic day 10.5 embryos when the neural tube should be closed if there is no neural tube defect formation. To identify which miR-322 transcript is affected by maternal diabetes mellitus and high glucose conditions, 3 miR-322 transcripts were assessed in embryos from dams with or without diabetes mellitus and in C17.2 mouse neural stem cells treated with different concentrations of glucose and at different time points. To determine whether the endonuclease IRE1α targets miR-322, small interfering RNA knockdown of IRE1α or overexpression of inositol-requiring transmembrane kinase/endoribonuclease 1α by DNA plasmid transfection was used to determine the effect of IRE1α deficiency or overexpression on miR-322 expression. RNA immunoprecipitation was performed to reveal the direct targets of inositol-requiring transmembrane kinase/endoribonuclease 1α. RESULTS Maternal diabetes mellitus suppressed miR-322 expression in the developing neuroepithelium. Restoring miR-322 expression in the neuroepithelium blocked maternal diabetes mellitus-induced caspase-3 and caspase-8 cleavage and cell apoptosis, leading to a neural tube defect reduction. Reversal of maternal diabetes mellitus-inhibited miR-322 via transgenic overexpression prevented TRAF3 up-regulation in embryos exposed to maternal diabetes mellitus. Activated IRE1α acted as an endonuclease and degraded precursor miR-322, resulting in mature miR-322 reduction. CONCLUSION This study supports the crucial role of the IRE1α-microRNA-TRAF3 circuit in the induction of neuroepithelial cell apoptosis and neural tube defect formation in pregnancies complicated by diabetes mellitus and identifies IRE1α and miR-322 as potential targets for preventing maternal diabetes mellitus-induced neural tube defects.
Collapse
Affiliation(s)
- Guanglei Wang
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD
| | - Shicong Song
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD
| | - Wei-Bin Shen
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD
| | - E Albert Reece
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD; Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD
| | - Peixin Yang
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD; Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD.
| |
Collapse
|
7
|
Bracchi-Ricard V, Nguyen K, Ricci D, Gaudette B, Henao-Meija J, Brambilla R, Martynyuk T, Gidalevitz T, Allman D, Bethea JR, Argon Y. Increased activity of IRE1 improves the clinical presentation of EAE. FASEB J 2023; 37:e23283. [PMID: 37983957 PMCID: PMC10662669 DOI: 10.1096/fj.202300769rr] [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: 04/19/2023] [Revised: 10/03/2023] [Accepted: 10/17/2023] [Indexed: 11/22/2023]
Abstract
Activation of the endoplasmic reticulum (ER) stress sensor inositol-requiring enzyme-1α (IRE1α) contributes to neuronal development and is known to induce neuronal remodeling in vitro and in vivo. On the contrary, excessive IRE1 activity is often detrimental and may contribute to neurodegeneration. To determine the consequences of increased activation of IRE1α, we used a mouse model expressing a C148S variant of IRE1α with increased and sustained activation. Surprisingly, the mutation did not affect the differentiation of highly secretory antibody-producing cells but exhibited a beneficial effect in a mouse model of experimental autoimmune encephalomyelitis (EAE). Although mechanical allodynia was unaffected, significant improvement in motor function was found in IRE1C148S mice with EAE relative to wild type (WT) mice. Coincident with this improvement, there was reduced microgliosis in the spinal cord of IRE1C148S mice, with reduced expression of proinflammatory cytokine genes. This was accompanied by reduced axonal degeneration and enhanced 2',3'-cyclic nucleotide 3'-phosphodiesterase (CNPase) levels, suggesting improved myelin integrity. Interestingly, while the IRE1C148S mutation is expressed in all cells, the reduction in proinflammatory cytokines and in the microglial activation marker ionized calcium-binding adapter molecule (IBA1), along with preservation of phagocytic gene expression, all point to microglia as the cell type contributing to the clinical improvement in IRE1C148S animals. Our data suggest that sustained increase in IRE1α activity can be beneficial in vivo, and that this protection is cell type and context dependent. Considering the overwhelming but conflicting evidence for the role of ER stress in neurological diseases, a better understanding of the function of ER stress sensors in physiological contexts is clearly needed.
Collapse
Affiliation(s)
| | - Kayla Nguyen
- Department of Biology, Drexel University, Philadelphia, PA
| | - Daniela Ricci
- Department of Pathology and Lab Medicine, The Children’s Hospital of Philadelphia and the University of Pennsylvania, Philadelphia, PA, USA
| | - Brian Gaudette
- Department of Pathology and Lab Medicine, the University of Pennsylvania, Philadelphia, PA, USA
| | - Jorge Henao-Meija
- Department of Pathology and Lab Medicine, The Children’s Hospital of Philadelphia and the University of Pennsylvania, Philadelphia, PA, USA
| | - Roberta Brambilla
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark
- BRIDGE - Brain Research - Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, 5000 Odense, Denmark
| | | | | | - David Allman
- Department of Pathology and Lab Medicine, the University of Pennsylvania, Philadelphia, PA, USA
| | - John R. Bethea
- Department of Biology, Drexel University, Philadelphia, PA
| | - Yair Argon
- Department of Pathology and Lab Medicine, The Children’s Hospital of Philadelphia and the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
8
|
Fujii S, Ushioda R, Nagata K. Redox states in the endoplasmic reticulum directly regulate the activity of calcium channel, inositol 1,4,5-trisphosphate receptors. Proc Natl Acad Sci U S A 2023; 120:e2216857120. [PMID: 37216546 PMCID: PMC10235943 DOI: 10.1073/pnas.2216857120] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 03/14/2023] [Indexed: 05/24/2023] Open
Abstract
Inositol 1,4,5-trisphosphate receptors (IP3Rs) are one of the two types of tetrameric ion channels that release calcium ion (Ca2+) from the endoplasmic reticulum (ER) into the cytosol. Ca2+ released via IP3Rs is a fundamental second messenger for numerous cell functions. Disturbances in the intracellular redox environment resulting from various diseases and aging interfere with proper calcium signaling, however, the details are unclear. Here, we elucidated the regulatory mechanisms of IP3Rs by protein disulfide isomerase family proteins localized in the ER by focusing on four cysteine residues residing in the ER lumen of IP3Rs. First, we revealed that two of the cysteine residues are essential for functional tetramer formation of IP3Rs. Two other cysteine residues, on the contrary, were revealed to be involved in the regulation of IP3Rs activity; its oxidation by ERp46 and the reduction by ERdj5 caused the activation and the inactivation of IP3Rs activity, respectively. We previously reported that ERdj5 can activate the sarco/endoplasmic reticulum Ca2+-ATPase isoform 2b (SERCA2b) using its reducing activity [Ushioda et al., Proc. Natl. Acad. Sci. U.S.A. 113, E6055-E6063 (2016)]. Thus, we here established that ERdj5 exerts the reciprocal regulatory function for IP3Rs and SERCA2b by sensing the ER luminal Ca2+ concentration, which contributes to the calcium homeostasis in the ER.
Collapse
Affiliation(s)
- Shohei Fujii
- Laboratory of Molecular and Cellular Biology, Department of Frontier Life Sciences, Faculty of Life Science, Kyoto Sangyo University, Kyoto603-8555, Japan
| | - Ryo Ushioda
- Laboratory of Molecular and Cellular Biology, Department of Frontier Life Sciences, Faculty of Life Science, Kyoto Sangyo University, Kyoto603-8555, Japan
- Institute for Protein Dynamics, Kyoto Sangyo University, Kyoto603-8555, Japan
| | - Kazuhiro Nagata
- Laboratory of Molecular and Cellular Biology, Department of Frontier Life Sciences, Faculty of Life Science, Kyoto Sangyo University, Kyoto603-8555, Japan
- Institute for Protein Dynamics, Kyoto Sangyo University, Kyoto603-8555, Japan
- JT Biohistory Research Hall, Takatsuki City, Osaka569-1125, Japan
| |
Collapse
|
9
|
Groenendyk J, Michalak M. Interplay between calcium and endoplasmic reticulum stress. Cell Calcium 2023; 113:102753. [PMID: 37209448 DOI: 10.1016/j.ceca.2023.102753] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/05/2023] [Accepted: 05/06/2023] [Indexed: 05/22/2023]
Abstract
Cellular homeostasis is crucial for the healthy functioning of the organism. Disruption of cellular homeostasis activates endoplasmic reticulum (ER) stress coping responses including the unfolded protein response (UPR). There are three ER resident stress sensors responsible for UPR activation - IRE1α, PERK and ATF6. Ca2+ signaling plays an important role in stress responses including the UPR and the ER is the main Ca2+ storage organelle and a source of Ca2+ for cell signaling. The ER contains many proteins involved in Ca2+ import/export/ storage, Ca2+ movement between different cellular organelles and ER Ca2+ stores refilling. Here we focus on selected aspects of ER Ca2+ homeostasis and its role in activation of the ER stress coping responses.
Collapse
Affiliation(s)
- Jody Groenendyk
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada.
| | - Marek Michalak
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada.
| |
Collapse
|
10
|
Bracchi-Ricard V, Nguyen K, Ricci D, Gaudette B, Henao-Meija J, Brambilla R, Martynyuk T, Gidalevitz T, Allman D, Bethea JR, Argon Y. Increased activity of IRE1 improves the clinical presentation of EAE. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.19.537391. [PMID: 37131811 PMCID: PMC10153167 DOI: 10.1101/2023.04.19.537391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Activation of the ER stress sensor IRE1α contributes to neuronal development and is known to induce neuronal remodeling in vitro and in vivo. On the other hand, excessive IRE1 activity is often detrimental and may contribute to neurodegeneration. To determine the consequences of increased activation of IRE1α, we used a mouse model expressing a C148S variant of IRE1α with increased and sustained activation. Surprisingly, the mutation did not affect the differentiation of highly secretory antibody-producing cells, but exhibited a strong protective effect in a mouse model of experimental autoimmune encephalomyelitis (EAE). Significant improvement in motor function was found in IRE1C148S mice with EAE relative to WT mice. Coincident with this improvement, there was reduced microgliosis in the spinal cord of IRE1C148S mice, with reduced expression of pro-inflammatory cytokine genes. This was accompanied by reduced axonal degeneration and enhanced CNPase levels, suggestiing improved myelin integrity. Interestingly, while the IRE1C148S mutation is expressed in all cells, the reduction in proinflammatory cytokines and in the activation of microglial activation marker IBA1, along with preservation of phagocytic gene expression, all point to microglia as the cell type contributing to the clinical improvement in IRE1C148S animals. Our data suggest that sustained increase in IRE1α activity can be protective in vivo, and that this protection is cell type and context dependent. Considering the overwhelming but conflicting evidence for the role of the ER stress in neurological diseases, a better understanding of the function of ER stress sensors in physiological contexts is clearly needed.
Collapse
Affiliation(s)
| | - Kayla Nguyen
- Department of Biology, Drexel University, Philadelphia, PA
| | - Daniela Ricci
- Department of Pathology and Lab Medicine, The Children's Hospital of Philadelphia and the University of Pennsylvania, Philadelphia, PA, USA
| | - Brian Gaudette
- Department of Pathology and Lab Medicine, the University of Pennsylvania, Philadelphia, PA, USA
| | - Jorge Henao-Meija
- Department of Pathology and Lab Medicine, The Children's Hospital of Philadelphia and the University of Pennsylvania, Philadelphia, PA, USA
| | - Roberta Brambilla
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark
- BRIDGE - Brain Research - Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, 5000 Odense, Denmark
| | | | | | - David Allman
- Department of Pathology and Lab Medicine, the University of Pennsylvania, Philadelphia, PA, USA
| | - John R Bethea
- Department of Biology, Drexel University, Philadelphia, PA
| | - Yair Argon
- Department of Pathology and Lab Medicine, The Children's Hospital of Philadelphia and the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
11
|
Li K, Li Y, Ding H, Chen J, Zhang X. Metal-Binding Proteins Cross-Linking with Endoplasmic Reticulum Stress in Cardiovascular Diseases. J Cardiovasc Dev Dis 2023; 10:jcdd10040171. [PMID: 37103050 PMCID: PMC10143100 DOI: 10.3390/jcdd10040171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 04/11/2023] [Accepted: 04/15/2023] [Indexed: 04/28/2023] Open
Abstract
The endoplasmic reticulum (ER), an essential organelle in eukaryotic cells, is widely distributed in myocardial cells. The ER is where secreted protein synthesis, folding, post-translational modification, and transport are all carried out. It is also where calcium homeostasis, lipid synthesis, and other processes that are crucial for normal biological cell functioning are regulated. We are concerned that ER stress (ERS) is widespread in various damaged cells. To protect cells' function, ERS reduces the accumulation of misfolded proteins by activating the unfolded protein response (UPR) pathway in response to numerous stimulating factors, such as ischemia or hypoxia, metabolic disorders, and inflammation. If these stimulatory factors are not eliminated for a long time, resulting in the persistence of the UPR, it will aggravate cell damage through a series of mechanisms. In the cardiovascular system, it will cause related cardiovascular diseases and seriously endanger human health. Furthermore, there has been a growing number of studies on the antioxidative stress role of metal-binding proteins. We observed that a variety of metal-binding proteins can inhibit ERS and, hence, mitigate myocardial damage.
Collapse
Affiliation(s)
- Kejuan Li
- Department of Cardiology, Lanzhou University Second Hospital, Lanzhou University, Lanzhou 730031, China
| | - Yongnan Li
- Department of Cardiac Surgery, Lanzhou University Second Hospital, Lanzhou University, Lanzhou 730031, China
| | - Hong Ding
- Department of Cardiology, Lanzhou University Second Hospital, Lanzhou University, Lanzhou 730031, China
| | - Jianshu Chen
- Department of Cardiology, Lanzhou University Second Hospital, Lanzhou University, Lanzhou 730031, China
| | - Xiaowei Zhang
- Department of Cardiology, Lanzhou University Second Hospital, Lanzhou University, Lanzhou 730031, China
| |
Collapse
|
12
|
Matai L, Slack FJ. MicroRNAs in Age-Related Proteostasis and Stress Responses. Noncoding RNA 2023; 9:26. [PMID: 37104008 PMCID: PMC10143298 DOI: 10.3390/ncrna9020026] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/27/2023] [Accepted: 03/30/2023] [Indexed: 04/28/2023] Open
Abstract
Aging is associated with the accumulation of damaged and misfolded proteins through a decline in the protein homeostasis (proteostasis) machinery, leading to various age-associated protein misfolding diseases such as Huntington's or Parkinson's. The efficiency of cellular stress response pathways also weakens with age, further contributing to the failure to maintain proteostasis. MicroRNAs (miRNAs or miRs) are a class of small, non-coding RNAs (ncRNAs) that bind target messenger RNAs at their 3'UTR, resulting in the post-transcriptional repression of gene expression. From the discovery of aging roles for lin-4 in C. elegans, the role of numerous miRNAs in controlling the aging process has been uncovered in different organisms. Recent studies have also shown that miRNAs regulate different components of proteostasis machinery as well as cellular response pathways to proteotoxic stress, some of which are very important during aging or in age-related pathologies. Here, we present a review of these findings, highlighting the role of individual miRNAs in age-associated protein folding and degradation across different organisms. We also broadly summarize the relationships between miRNAs and organelle-specific stress response pathways during aging and in various age-associated diseases.
Collapse
Affiliation(s)
| | - Frank J. Slack
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
13
|
Groenendyk J, Stoletov K, Paskevicius T, Li W, Dai N, Pujol M, Busaan E, Ng HH, Boukouris AE, Saleme B, Haromy A, Cui K, Hu M, Yan Y, Zhang R, Michelakis E, Chen XZ, Lewis JD, Tang J, Agellon LB, Michalak M. Loss of the fructose transporter SLC2A5 inhibits cancer cell migration. Front Cell Dev Biol 2022; 10:896297. [PMID: 36268513 PMCID: PMC9578049 DOI: 10.3389/fcell.2022.896297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 09/09/2022] [Indexed: 11/13/2022] Open
Abstract
Metastasis is the primary cause of cancer patient death and the elevation of SLC2A5 gene expression is often observed in metastatic cancer cells. Here we evaluated the importance of SLC2A5 in cancer cell motility by silencing its gene. We discovered that CRISPR/Cas9-mediated inactivation of the SLC2A5 gene inhibited cancer cell proliferation and migration in vitro as well as metastases in vivo in several animal models. Moreover, SLC2A5-attenuated cancer cells exhibited dramatic alterations in mitochondrial architecture and localization, uncovering the importance of SLC2A5 in directing mitochondrial function for cancer cell motility and migration. The direct association of increased abundance of SLC2A5 in cancer cells with metastatic risk in several types of cancers identifies SLC2A5 as an important therapeutic target to reduce or prevent cancer metastasis.
Collapse
Affiliation(s)
- Jody Groenendyk
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | | | | | - Wenjuan Li
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Ning Dai
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Myriam Pujol
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Erin Busaan
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Hoi Hei Ng
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | | | - Bruno Saleme
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Alois Haromy
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Kaisa Cui
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Miao Hu
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, China
| | - Yanan Yan
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, China
| | - Rui Zhang
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, China
| | | | - Xing-Zhen Chen
- Department of Physiology, University of Alberta, Edmonton, AB, Canada
| | - John D. Lewis
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
| | - Jingfeng Tang
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, China
| | - Luis B. Agellon
- School of Human Nutrition, McGill University, Montreal, QC, Canada
- *Correspondence: Luis B. Agellon, ; Marek Michalak,
| | - Marek Michalak
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
- *Correspondence: Luis B. Agellon, ; Marek Michalak,
| |
Collapse
|
14
|
Carvalho-Gontijo R, Han C, Zhang L, Zhang V, Hosseini M, Mekeel K, Schnabl B, Loomba R, Karin M, Brenner DA, Kisseleva T. Metabolic Injury of Hepatocytes Promotes Progression of NAFLD and AALD. Semin Liver Dis 2022; 42:233-249. [PMID: 36001995 PMCID: PMC9662188 DOI: 10.1055/s-0042-1755316] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Nonalcoholic liver disease is a component of metabolic syndrome associated with obesity, insulin resistance, and hyperlipidemia. Excessive alcohol consumption may accelerate the progression of steatosis, steatohepatitis, and fibrosis. While simple steatosis is considered a benign condition, nonalcoholic steatohepatitis with inflammation and fibrosis may progress to cirrhosis, liver failure, and hepatocellular cancer. Studies in rodent experimental models and primary cell cultures have demonstrated several common cellular and molecular mechanisms in the pathogenesis and regression of liver fibrosis. Chronic injury and death of hepatocytes cause the recruitment of myeloid cells, secretion of inflammatory and fibrogenic cytokines, and activation of myofibroblasts, resulting in liver fibrosis. In this review, we discuss the role of metabolically injured hepatocytes in the pathogenesis of nonalcoholic steatohepatitis and alcohol-associated liver disease. Specifically, the role of chemokine production and de novo lipogenesis in the development of steatotic hepatocytes and the pathways of steatosis regulation are discussed.
Collapse
Affiliation(s)
- Raquel Carvalho-Gontijo
- Department of Medicine, University of California, San Diego School of Medicine, La Jolla
- Department of Surgery, University of California, San Diego School of Medicine, La Jolla
| | - Cuijuan Han
- Department of Medicine, University of California, San Diego School of Medicine, La Jolla
- Department of Surgery, University of California, San Diego School of Medicine, La Jolla
| | - Lei Zhang
- Department of Medicine, University of California, San Diego School of Medicine, La Jolla
- Department of Surgery, University of California, San Diego School of Medicine, La Jolla
| | - Vivian Zhang
- Department of Medicine, University of California, San Diego School of Medicine, La Jolla
- Department of Surgery, University of California, San Diego School of Medicine, La Jolla
| | - Mojgan Hosseini
- Department of Pathology, University of California, San Diego School of Medicine, La Jolla
| | - Kristin Mekeel
- Department of Surgery, University of California, San Diego School of Medicine, La Jolla
| | - Bernd Schnabl
- Department of Medicine, University of California, San Diego School of Medicine, La Jolla
| | - Rohit Loomba
- Department of Medicine, University of California, San Diego School of Medicine, La Jolla
| | - Michael Karin
- Department of Pharmacology, University of California, San Diego School of Medicine, La Jolla
| | - David A. Brenner
- Department of Medicine, University of California, San Diego School of Medicine, La Jolla
| | - Tatiana Kisseleva
- Department of Surgery, University of California, San Diego School of Medicine, La Jolla
| |
Collapse
|
15
|
Mestre-Farràs N, Guerrero S, Bley N, Rivero E, Coll O, Borràs E, Sabidó E, Indacochea A, Casillas-Serra C, Järvelin AI, Oliva B, Castello A, Hüttelmaier S, Gebauer F. Melanoma RBPome identification reveals PDIA6 as an unconventional RNA-binding protein involved in metastasis. Nucleic Acids Res 2022; 50:8207-8225. [PMID: 35848924 PMCID: PMC9371929 DOI: 10.1093/nar/gkac605] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 06/10/2022] [Accepted: 07/01/2022] [Indexed: 11/13/2022] Open
Abstract
RNA-binding proteins (RBPs) have been relatively overlooked in cancer research despite their contribution to virtually every cancer hallmark. Here, we use RNA interactome capture (RIC) to characterize the melanoma RBPome and uncover novel RBPs involved in melanoma progression. Comparison of RIC profiles of a non-tumoral versus a metastatic cell line revealed prevalent changes in RNA-binding capacities that were not associated with changes in RBP levels. Extensive functional validation of a selected group of 24 RBPs using five different in vitro assays unveiled unanticipated roles of RBPs in melanoma malignancy. As proof-of-principle we focused on PDIA6, an ER-lumen chaperone that displayed a novel RNA-binding activity. We show that PDIA6 is involved in metastatic progression, map its RNA-binding domain, and find that RNA binding is required for PDIA6 tumorigenic properties. These results exemplify how RIC technologies can be harnessed to uncover novel vulnerabilities of cancer cells.
Collapse
Affiliation(s)
- Neus Mestre-Farràs
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, 08003 Barcelona, Spain
| | - Santiago Guerrero
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, 08003 Barcelona, Spain
| | - Nadine Bley
- Institute of Molecular Medicine, Section for Molecular Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, 06120 Halle, Germany
| | - Ezequiel Rivero
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, 08003 Barcelona, Spain
| | - Olga Coll
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, 08003 Barcelona, Spain
| | - Eva Borràs
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, 08003 Barcelona, Spain.,Department of Health and Experimental Sciences, Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain
| | - Eduard Sabidó
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, 08003 Barcelona, Spain.,Department of Health and Experimental Sciences, Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain
| | - Alberto Indacochea
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, 08003 Barcelona, Spain
| | - Carlos Casillas-Serra
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, 08003 Barcelona, Spain
| | - Aino I Järvelin
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Baldomero Oliva
- Department of Health and Experimental Sciences, Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain
| | - Alfredo Castello
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Stefan Hüttelmaier
- Institute of Molecular Medicine, Section for Molecular Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, 06120 Halle, Germany
| | - Fátima Gebauer
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, 08003 Barcelona, Spain.,Department of Health and Experimental Sciences, Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain
| |
Collapse
|
16
|
Shimada M, Maeda H, Nanashima N, Yamada K, Nakajima A. Anthocyanin‐rich blackcurrant extract improves long‐term memory impairment and emotional abnormality in senescence‐accelerated mice. J Food Biochem 2022; 46:e14295. [DOI: 10.1111/jfbc.14295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/16/2022] [Accepted: 06/10/2022] [Indexed: 11/28/2022]
Affiliation(s)
- Minori Shimada
- Department of Applied Biology and Food Sciences, Faculty of Agriculture and Life Science Hirosaki University Hirosaki Japan
| | - Hayato Maeda
- Department of Applied Biology and Food Sciences, Faculty of Agriculture and Life Science Hirosaki University Hirosaki Japan
| | - Naoki Nanashima
- Department of Biomedical Science and Laboratory Medicine Hirosaki University Graduate School of Health Sciences Hirosaki Japan
| | - Kiyofumi Yamada
- Department of Neuropsychopharmacology and Hospital Pharmacy Nagoya University Graduate School of Medicine Nagoya Japan
| | - Akira Nakajima
- Department of Applied Biology and Food Sciences, Faculty of Agriculture and Life Science Hirosaki University Hirosaki Japan
- Department of Industry Development Sciences Hirosaki University Graduate School of Sustainable Community Studies Hirosaki Japan
| |
Collapse
|
17
|
Critical roles of protein disulfide isomerases in balancing proteostasis in the nervous system. J Biol Chem 2022; 298:102087. [PMID: 35654139 PMCID: PMC9253707 DOI: 10.1016/j.jbc.2022.102087] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 05/05/2022] [Accepted: 05/08/2022] [Indexed: 02/08/2023] Open
Abstract
Protein disulfide isomerases (PDIs) constitute a family of oxidoreductases promoting redox protein folding and quality control in the endoplasmic reticulum. PDIs catalyze disulfide bond formation, isomerization, and reduction, operating in concert with molecular chaperones to fold secretory cargoes in addition to directing misfolded proteins to be refolded or degraded. Importantly, PDIs are emerging as key components of the proteostasis network, integrating protein folding status with central surveillance mechanisms to balance proteome stability according to cellular needs. Recent advances in the field driven by the generation of new mouse models, human genetic studies, and omics methodologies, in addition to interventions using small molecules and gene therapy, have revealed the significance of PDIs to the physiology of the nervous system. PDIs are also implicated in diverse pathologies, ranging from neurodevelopmental conditions to neurodegenerative diseases and traumatic injuries. Here, we review the principles of redox protein folding in the ER with a focus on current evidence linking genetic mutations and biochemical alterations to PDIs in the etiology of neurological conditions.
Collapse
|
18
|
Yang L, Li Z, Ouyang Y. Taurine attenuates ER stress‑associated apoptosis and catabolism in nucleus pulposus cells. Mol Med Rep 2022; 25:172. [PMID: 35315493 PMCID: PMC8971911 DOI: 10.3892/mmr.2022.12688] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 02/10/2022] [Indexed: 11/20/2022] Open
Abstract
Nucleus pulposus (NP) apoptosis and subsequent excessive degradation of the extracellular matrix (ECM) are key pathological characteristics of intervertebral disc degeneration (IDD). The present study aims to examine the signaling processes underlying the effects of taurine on IDD, with specific focus on endoplasmic reticulum (ER) stress-mediated apoptosis and ECM degradation, in NP cells. To clarify the role of taurine in IDD, NP cells were treated with various concentrations of taurine and IL-1β or thapsigargin (TG). Cell Counting Kit-8, western blotting, TUNEL, immunofluorescence assays and reverse transcription-quantitative PCR were applied to measure cell viability, the expression of ER stress-associated proteins (GRP78, CHOP and caspase-12), apoptosis and the levels of metabolic factors associated with ECM (MMP-1, 3, 9, ADAMTS-4, 5 and collagen II), respectively. Taurine was found to attenuate ER stress and prevent apoptosis in NP cells induced by IL-1β treatment. Additionally, taurine significantly decreased the expression of ER stress-activated glucose regulatory protein 78, C/EBP homologous protein and caspase-12. TUNEL results revealed that taurine decreased the number of apoptotic TG-treated NP cells. TG-treated NP cells also exhibited characteristics of increased ECM degradation, supported by observations of increased MMP-1, MMP-3, MMP-9 and A disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS)-4 and ADAMTS-5 expression in addition to decreased collagen-II expression. However, taurine treatment significantly reversed all indicators of excessive ECM catabolism aforementioned. These data suggest that taurine can mediate protection against apoptosis and ECM degradation in NP cells by inhibiting ER stress, implicating therapeutic potential for the treatment of IDD.
Collapse
Affiliation(s)
- Liuxie Yang
- Department of Orthopedics, Shanghai Jing'an District Zhabei Central Hospital, Shanghai 200040, P.R. China
| | - Zhenhuan Li
- Department of Orthopedics, Shanghai Jing'an District Zhabei Central Hospital, Shanghai 200040, P.R. China
| | - Yueping Ouyang
- Department of Orthopedics, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| |
Collapse
|
19
|
Demirel-Yalciner T, Sozen E, Ozer NK. Endoplasmic Reticulum Stress and miRNA Impairment in Aging and Age-Related Diseases. FRONTIERS IN AGING 2022; 2:790702. [PMID: 35822008 PMCID: PMC9261320 DOI: 10.3389/fragi.2021.790702] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 12/14/2021] [Indexed: 11/20/2022]
Abstract
Aging is a physiological process defined by decreased cellular and tissue functions. Reduced capacity of protein degradation is one of the important hallmarks of aging that may lead to misfolded protein accumulation and progressive loss of function in organ systems. Recognition of unfolded/misfolded protein aggregates via endoplasmic reticulum (ER) stress sensors activates an adaptive mechanism, the unfolded protein response (UPR). The initial step of UPR is defined by chaperone enhancement, ribosomal translation suppression, and misfolded protein degradation, while prolonged ER stress triggers apoptosis. MicroRNAs (miRNAs) are non-coding RNAs affecting various signaling pathways through degradation or translational inhibition of targeted mRNAs. Therefore, UPR and miRNA impairment in aging and age-related diseases is implicated in various studies. This review will highlight the recent insights in ER stress–miRNAs alterations during aging and age-related diseases, including metabolic, cardiovascular, and neurodegenerative diseases and several cancers.
Collapse
Affiliation(s)
| | - Erdi Sozen
- Department of Biochemistry, Faculty of Medicine, Marmara University, Maltepe, Turkey
- Genetic and Metabolic Diseases Research and Investigation Center (GEMHAM), Marmara University, Maltepe, Turkey
| | - Nesrin Kartal Ozer
- Department of Biochemistry, Faculty of Medicine, Marmara University, Maltepe, Turkey
- *Correspondence: Nesrin Kartal Ozer,
| |
Collapse
|
20
|
Zhou Z, Wang Q, Michalak M. Inositol Requiring Enzyme (IRE), a multiplayer in sensing endoplasmic reticulum stress. Anim Cells Syst (Seoul) 2022; 25:347-357. [PMID: 35059134 PMCID: PMC8765250 DOI: 10.1080/19768354.2021.2020901] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Affiliation(s)
- Zhixin Zhou
- Department of Biochemistry, University of Alberta, Edmonton, Canada
| | - Qian Wang
- Department of Biochemistry, University of Alberta, Edmonton, Canada
| | - Marek Michalak
- Department of Biochemistry, University of Alberta, Edmonton, Canada
| |
Collapse
|
21
|
Moraga P, Aravena R, Urra H, Hetz C. Assays to Study IRE1 Activation and Signaling. Methods Mol Biol 2022; 2378:141-168. [PMID: 34985699 DOI: 10.1007/978-1-0716-1732-8_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The endoplasmic reticulum (ER) stress sensor IRE1 is a a major player of the unfolded protein response (UPR), the main pathway driving adaptation processes to restore proteostasis. In addition, overactivation of IRE1 signaling contributes to a variety of pathologies including diabetes, neurodegenerative diseases, and cancer. Under ER stress, IRE1 auto-transphosphorylates and oligomerizes, triggering the activation of its endoribonuclease domain located in the cytosolic region. Active IRE1 catalyzes the splicing of the mRNA encoding for the XBP1 transcription factor, in addition to degrade several RNAs through a process known as regulated IRE1-dependent decay of mRNA (RIDD). Besides its role as an UPR transducer, several posttranslational modifications and protein-protein interactions can regulate IRE1 activity and modulate its signaling in the absence of stress. Thus, investigating the function of IRE1 in physiology and disease requires the use of complementary approaches. Here, we provide detailed protocols to perform four different assays to study IRE1 activation and signaling: (i) Phos-tag gels to evaluate the phosphorylation status of IRE1, (ii) microscopy using TREX-IRE1-GFP cells to measure IRE1 oligomerization, (iii) conventional RT-PCR to assess XBP1 mRNA processing, and (iv) quantitative PCR to determine the levels of canonical UPR target genes and the degradation of several mRNAs that are target of RIDD. We propose to use these experimental strategies as "gold standards" to study IRE1 signaling.
Collapse
Affiliation(s)
- Paloma Moraga
- Faculty of Medicine, Biomedical Neuroscience Institute (BNI), University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health and Metabolism (GERO), Santiago, Chile
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), University of Chile, Santiago, Chile
| | - Raul Aravena
- Faculty of Medicine, Biomedical Neuroscience Institute (BNI), University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health and Metabolism (GERO), Santiago, Chile
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), University of Chile, Santiago, Chile
| | - Hery Urra
- Faculty of Medicine, Biomedical Neuroscience Institute (BNI), University of Chile, Santiago, Chile.
- Center for Geroscience, Brain Health and Metabolism (GERO), Santiago, Chile.
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), University of Chile, Santiago, Chile.
| | - Claudio Hetz
- Faculty of Medicine, Biomedical Neuroscience Institute (BNI), University of Chile, Santiago, Chile.
- Center for Geroscience, Brain Health and Metabolism (GERO), Santiago, Chile.
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), University of Chile, Santiago, Chile.
- The Buck Institute for Research in Aging, Novato, CA, USA.
| |
Collapse
|
22
|
Characterization of the AGR2 interactome uncovers new players of Protein Disulfide Isomerase network in cancer cells. Mol Cell Proteomics 2021; 21:100188. [PMID: 34929376 PMCID: PMC8816719 DOI: 10.1016/j.mcpro.2021.100188] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 12/03/2021] [Accepted: 12/14/2021] [Indexed: 12/03/2022] Open
Abstract
Anterior gradient 2 (AGR2) is an endoplasmic reticulum (ER)-resident protein disulfide isomerase (PDI) known to be overexpressed in many human epithelial cancers and is involved in cell migration, cellular transformation, angiogenesis, and metastasis. This protein inhibits the activity of the tumor suppressor p53, and its expression levels can be used to predict cancer patient outcome. However, the precise network of AGR2-interacting partners and clients remains to be fully characterized. Herein, we used label-free quantification and also stable isotope labeling with amino acids in cell culture–based LC–MS/MS analyses to identify proteins interacting with AGR2. Functional annotation confirmed that AGR2 and its interaction partners are associated with processes in the ER that maintain intracellular metabolic homeostasis and participate in the unfolded protein response, including those associated with changes in cellular metabolism, energy, and redox states in response to ER stress. As a proof of concept, the interaction between AGR2 and PDIA3, another ER-resident PDI, was studied in more detail. Pathway analysis revealed that AGR2 and PDIA3 play roles in protein folding in ER, including post-translational modification and in cellular response to stress. We confirmed the AGR2–PDIA3 complex formation in cancer cells, which was enhanced in response to ER stress. Accordingly, molecular docking characterized potential quaternary structure of this complex; however, it remains to be elucidated whether AGR2 rather contributes to PDIA3 maturation in ER, the complex directly acts in cellular signaling, or mediates AGR2 secretion. Our study provides a comprehensive insight into the protein–protein interaction network of AGR2 by identifying functionally relevant proteins and related cellular and biochemical pathways associated with the role of AGR2 in cancer cells. LC–MS/MS analysis of AGR2-interacting proteins in T47D and H1299 cells. About 15 overlapping AGR2 interactors, including PDIA3 and PDIA6, were identified in both cell lines. PDI family members represent the key part of the network. AGR2–PDIA3 interaction is even stronger under ER stress. AGR2–PDIA3 complex formation supports extracellular secretion of AGR2.
Collapse
|
23
|
Matsusaki M, Okada R, Tanikawa Y, Kanemura S, Ito D, Lin Y, Watabe M, Yamaguchi H, Saio T, Lee YH, Inaba K, Okumura M. Functional Interplay between P5 and PDI/ERp72 to Drive Protein Folding. BIOLOGY 2021; 10:biology10111112. [PMID: 34827105 PMCID: PMC8615271 DOI: 10.3390/biology10111112] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/21/2021] [Accepted: 10/27/2021] [Indexed: 11/16/2022]
Abstract
P5 is one of protein disulfide isomerase family proteins (PDIs) involved in endoplasmic reticulum (ER) protein quality control that assists oxidative folding, inhibits protein aggregation, and regulates the unfolded protein response. P5 reportedly interacts with other PDIs via intermolecular disulfide bonds in cultured cells, but it remains unclear whether complex formation between P5 and other PDIs is involved in regulating enzymatic and chaperone functions. Herein, we established the far-western blot method to detect non-covalent interactions between P5 and other PDIs and found that PDI and ERp72 are partner proteins of P5. The enzymatic activity of P5-mediated oxidative folding is up-regulated by PDI, while the chaperone activity of P5 is stimulated by ERp72. These findings shed light on the mechanism by which the complex formations among PDIs drive to synergistically accelerate protein folding and prevents aggregation. This knowledge has implications for understanding misfolding-related pathology.
Collapse
Affiliation(s)
- Motonori Matsusaki
- Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, 6-3, Aramakiaza Aoba, Aoba-ku, Sendai 980-8578, Japan; (M.M.); (S.K.); (M.W.)
- Institute of Advanced Medical Sciences, Tokushima University, 3-18-15, Kuramoto-cho, Tokushima 770-8503, Japan;
| | - Rina Okada
- School of Science and Technology, Kwansei Gakuin University, 2-1, Gakuen, Sanda 669-1337, Japan; (R.O.); (Y.T.); (H.Y.)
| | - Yuya Tanikawa
- School of Science and Technology, Kwansei Gakuin University, 2-1, Gakuen, Sanda 669-1337, Japan; (R.O.); (Y.T.); (H.Y.)
| | - Shingo Kanemura
- Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, 6-3, Aramakiaza Aoba, Aoba-ku, Sendai 980-8578, Japan; (M.M.); (S.K.); (M.W.)
- School of Science and Technology, Kwansei Gakuin University, 2-1, Gakuen, Sanda 669-1337, Japan; (R.O.); (Y.T.); (H.Y.)
| | - Dai Ito
- Department of Brain and Cognitive Science, Daegu Gyeongbuk Institute of Science and Technology, 333, Techno Jungang Daero, Daegu 42988, Korea;
| | - Yuxi Lin
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, 162, Yeongudanji-ro, Ochang-eup, Cheongwon-gu, Cheongju-si 28119, Korea; (Y.L.); (Y.-H.L.)
| | - Mai Watabe
- Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, 6-3, Aramakiaza Aoba, Aoba-ku, Sendai 980-8578, Japan; (M.M.); (S.K.); (M.W.)
| | - Hiroshi Yamaguchi
- School of Science and Technology, Kwansei Gakuin University, 2-1, Gakuen, Sanda 669-1337, Japan; (R.O.); (Y.T.); (H.Y.)
| | - Tomohide Saio
- Institute of Advanced Medical Sciences, Tokushima University, 3-18-15, Kuramoto-cho, Tokushima 770-8503, Japan;
| | - Young-Ho Lee
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, 162, Yeongudanji-ro, Ochang-eup, Cheongwon-gu, Cheongju-si 28119, Korea; (Y.L.); (Y.-H.L.)
- Bio-Analytical Science, University of Science and Technology, 217, Gajeong-ro, Yuseong-gu, Daejeon 34113, Korea
- Graduate School of Analytical Science and Technology, Chungnam National University, 99, Daehak-ro, Yuseong-gu, Daejeon 34134, Korea
- Research Headquarters, Korea Brain Research Institute, 61, Cheomdan-ro, Dong-gu, Daegu 41068, Korea
| | - Kenji Inaba
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai 980-8577, Japan;
| | - Masaki Okumura
- Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, 6-3, Aramakiaza Aoba, Aoba-ku, Sendai 980-8578, Japan; (M.M.); (S.K.); (M.W.)
- Correspondence: ; Tel.: +81-22-795-5764
| |
Collapse
|
24
|
Kim T, Croce CM. MicroRNA and ER stress in cancer. Semin Cancer Biol 2021; 75:3-14. [PMID: 33422566 DOI: 10.1016/j.semcancer.2020.12.025] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 12/24/2020] [Accepted: 12/30/2020] [Indexed: 12/12/2022]
Abstract
The development of biological technologies in genomics, proteomics, and bioinformatics has led to the identification and characterization of the complete set of coding genes and their roles in various cellular pathways in cancer. Nevertheless, the cellular pathways have not been fully figured out like a jigsaw puzzle with missing pieces. The discovery of noncoding RNAs including microRNAs (miRNAs) has provided the missing pieces of the cellular pathways. Likewise, miRNAs have settled many questions of inexplicable patches in the endoplasmic reticulum (ER) stress pathways. The ER stress-caused pathways typified by the unfolded protein response (UPR) are pivotal processes for cellular homeostasis and survival, rectifying uncontrolled proteostasis and determining the cell fate. Although various factors and pathways have been studied and characterized, the understanding of the ER stress requires more wedges to fill the cracks of knowledge about the ER stress pathways. Moreover, the roles of the ER stress and UPR are still controversial in cancer despite their strong potential to promote cancer. The noncoding RNAs, in particular, miRNAs aid in a better understanding of the ER stress and its role in cancer. In this review, miRNAs that are the more-investigated subtype of noncoding RNAs are focused on the interpretation of the ER stress in cancer, following the introduction of miRNA and ER stress.
Collapse
Affiliation(s)
- Taewan Kim
- Department of Anatomy, Histology & Developmental Biology, Base for International Science and Technology Cooperation, Carson Cancer Stem Cell Vaccines R&D Center, International Cancer Center, Shenzhen University Health Science Center, Shenzhen 518055, China; The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA.
| | - Carlo M Croce
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
25
|
A point mutation in the Pdia6 gene results in loss of pancreatic β-cell identity causing overt diabetes. Mol Metab 2021; 54:101334. [PMID: 34487921 PMCID: PMC8515296 DOI: 10.1016/j.molmet.2021.101334] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 08/06/2021] [Accepted: 08/31/2021] [Indexed: 11/22/2022] Open
Abstract
OBJECTIVE Protein disulfide isomerases (PDIs) are oxidoreductases that are involved in catalyzing the formation and rearrangement of disulfide bonds during protein folding. One of the PDI members is the PDI-associated 6 (PDIA6) protein, which has been shown to play a vital role in β-cell dysfunction and diabetes. However, very little is known about the function of this protein in β-cells in vivo. This study aimed to describe the consequences of a point mutation in Pdia6 on β-cell development and function. METHODS We generated an ENU mouse model carrying a missense mutation (Phe175Ser) in the second thioredoxin domain of the Pdia6 gene. Using biochemical and molecular tools, we determined the effects of the mutation on the β-cell development at embryonic day (E)18.5 and β-cell identity as well as function at postnatal stages. RESULTS Mice homozygous for the Phe175Ser (F175S) mutation were mildly hyperglycemic at weaning and subsequently became hypoinsulinemic and overtly diabetic at the adult stage. Although no developmental phenotype was detected during embryogenesis, mutant mice displayed reduced insulin-expressing β-cells at P14 and P21 without any changes in the rate of cell death and proliferation. Further analysis revealed an increase in BiP and the PDI family member PDIA4, but without any concomitant apoptosis and cell death. Instead, the expression of prominent markers of β-cell maturation and function, such as Ins2, Mafa, and Slc2a2, along with increased expression of α-cell markers, Mafb, and glucagon was observed in adult mice, suggesting loss of β-cell identity. CONCLUSIONS The results demonstrate that a global Pdia6 mutation renders mice hypoinsulinemic and hyperglycemic. This occurs due to the loss of pancreatic β-cell function and identity, suggesting a critical role of PDIA6 specifically for β-cells.
Collapse
|
26
|
Han Y, Yuan M, Guo YS, Shen XY, Gao ZK, Bi X. Mechanism of Endoplasmic Reticulum Stress in Cerebral Ischemia. Front Cell Neurosci 2021; 15:704334. [PMID: 34408630 PMCID: PMC8365026 DOI: 10.3389/fncel.2021.704334] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 07/09/2021] [Indexed: 12/25/2022] Open
Abstract
Endoplasmic reticulum (ER) is the main organelle for protein synthesis, trafficking and maintaining intracellular Ca2+ homeostasis. The stress response of ER results from the disruption of ER homeostasis in neurological disorders. Among these disorders, cerebral ischemia is a prevalent reason of death and disability in the world. ER stress stemed from ischemic injury initiates unfolded protein response (UPR) regarded as a protection mechanism. Important, disruption of Ca2+ homeostasis resulted from cytosolic Ca2+ overload and depletion of Ca2+ in the lumen of the ER could be a trigger of ER stress and the misfolded protein synthesis. Brain cells including neurons, glial cells and endothelial cells are involved in the complex pathophysiology of ischemic stroke. This is generally important for protein underfolding, but even more for cytosolic Ca2+ overload. Mild ER stress promotes cells to break away from danger signals and enter the adaptive procedure with the activation of pro-survival mechanism to rescue ischemic injury, while chronic ER stress generally serves as a detrimental role on nerve cells via triggering diverse pro-apoptotic mechanism. What’s more, the determination of some proteins in UPR during cerebral ischemia to cell fate may have two diametrically opposed results which involves in a specialized set of inflammatory and apoptotic signaling pathways. A reasonable understanding and exploration of the underlying molecular mechanism related to ER stress and cerebral ischemia is a prerequisite for a major breakthrough in stroke treatment in the future. This review focuses on recent findings of the ER stress as well as the progress research of mechanism in ischemic stroke prognosis provide a new treatment idea for recovery of cerebral ischemia.
Collapse
Affiliation(s)
- Yu Han
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, China.,Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Mei Yuan
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, China.,Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Yi-Sha Guo
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, China.,Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Xin-Ya Shen
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China.,Shanghai University of Medicine and Health Sciences Affiliated Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhen-Kun Gao
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China.,Shanghai University of Medicine and Health Sciences Affiliated Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xia Bi
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| |
Collapse
|
27
|
Groenendyk J, Agellon LB, Michalak M. Calcium signaling and endoplasmic reticulum stress. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021; 363:1-20. [PMID: 34392927 DOI: 10.1016/bs.ircmb.2021.03.003] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cellular homeostasis is essential for healthy functioning of cells and tissues as well as proper organ development and maintenance. A disruption in cellular homeostasis triggers stress responses including the unfolded protein response (UPR), an endoplasmic reticulum (ER) stress coping response. There is increasing evidence that Ca2+ signaling plays a pivotal role in stress responses, as Ca2+ is involved many cellular activities. The ER is the main Ca2+ storage organelle and the source of Ca2+ for intracellular signaling. The ER is equipped with a variety of stress sensors and contains many Ca2+ handling proteins that support a role for Ca2+ in stress sensing and in coordinating strategies required to cope with cellular stress. Maintenance of ER Ca2+ homeostasis is therefore vital in sustaining cellular functions especially during times of cellular stress. Here we focus on selected aspects of ER Ca2+ homeostasis, its links to ER stress, and activation of the ER stress coping response.
Collapse
Affiliation(s)
- Jody Groenendyk
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada.
| | - Luis B Agellon
- School of Human Nutrition, McGill University, Ste. Anne de Bellevue, QC, Canada.
| | - Marek Michalak
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
28
|
Panda S, Behera S, Alam MF, Syed GH. Endoplasmic reticulum & mitochondrial calcium homeostasis: The interplay with viruses. Mitochondrion 2021; 58:227-242. [PMID: 33775873 DOI: 10.1016/j.mito.2021.03.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 03/08/2021] [Accepted: 03/22/2021] [Indexed: 02/08/2023]
Abstract
Calcium ions (Ca2+) act as secondary messengers in a plethora of cellular processes and play crucial role in cellular organelle function and homeostasis. The average resting concentration of Ca2+ is nearly 100 nM and in certain cells it can reach up to 1 µM. The high range of Ca2+ concentration across the plasma membrane and intracellular Ca2+ stores demands a well-coordinated maintenance of free Ca2+ via influx, efflux, buffering and storage. Endoplasmic Reticulum (ER) and Mitochondria depend on Ca2+ for their function and also serve as major players in intracellular Ca2+ homeostasis. The ER-mitochondria interplay helps in orchestrating cellular calcium homeostasis to avoid any detrimental effect resulting from Ca2+ overload or depletion. Since Ca2+ plays a central role in many biological processes it is an essential component of the virus-host interactions. The large gradient across membranes enable the viruses to easily modulate this buffered environment to meet their needs. Viruses exploit Ca2+ signaling to establish productive infection and evade the host immune defense. In this review we will detail the interplay between the viruses and cellular & ER-mitochondrial calcium signaling and the significance of these events on viral life cycle and disease pathogenesis.
Collapse
Affiliation(s)
- Swagatika Panda
- Institute of Life Sciences, Bhubaneswar, Virus-Host Interaction Lab, Institute of Life Sciences, Bhubaneswar, India
| | - Suchismita Behera
- Institute of Life Sciences, Bhubaneswar, Clinical Proteomics Laboratory, Institute of Life Sciences, Bhubaneswar, India
| | - Mohd Faraz Alam
- Institute of Life Sciences, Bhubaneswar, Virus-Host Interaction Lab, Institute of Life Sciences, Bhubaneswar, India
| | - Gulam Hussain Syed
- Institute of Life Sciences, Bhubaneswar, Virus-Host Interaction Lab, Institute of Life Sciences, Bhubaneswar, India.
| |
Collapse
|
29
|
Evolution and function of the epithelial cell-specific ER stress sensor IRE1β. Mucosal Immunol 2021; 14:1235-1246. [PMID: 34075183 PMCID: PMC8528705 DOI: 10.1038/s41385-021-00412-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 04/30/2021] [Accepted: 05/01/2021] [Indexed: 02/04/2023]
Abstract
Barrier epithelial cells lining the mucosal surfaces of the gastrointestinal and respiratory tracts interface directly with the environment. As such, these tissues are continuously challenged to maintain a healthy equilibrium between immunity and tolerance against environmental toxins, food components, and microbes. An extracellular mucus barrier, produced and secreted by the underlying epithelium plays a central role in this host defense response. Several dedicated molecules with a unique tissue-specific expression in mucosal epithelia govern mucosal homeostasis. Here, we review the biology of Inositol-requiring enzyme 1β (IRE1β), an ER-resident endonuclease and paralogue of the most evolutionarily conserved ER stress sensor IRE1α. IRE1β arose through gene duplication in early vertebrates and adopted functions unique from IRE1α which appear to underlie the basic development and physiology of mucosal tissues.
Collapse
|
30
|
Dastghaib S, Kumar PS, Aftabi S, Damera G, Dalvand A, Sepanjnia A, Kiumarsi M, Aghanoori MR, Sohal SS, Ande SR, Alizadeh J, Mokarram P, Ghavami S, Sharma P, Zeki AA. Mechanisms Targeting the Unfolded Protein Response in Asthma. Am J Respir Cell Mol Biol 2021; 64:29-38. [PMID: 32915643 PMCID: PMC12042654 DOI: 10.1165/rcmb.2019-0235tr] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Accepted: 09/11/2020] [Indexed: 12/16/2022] Open
Abstract
Lung cells are constantly exposed to various internal and external stressors that disrupt protein homeostasis. To cope with these stimuli, cells evoke a highly conserved adaptive mechanism called the unfolded protein response (UPR). UPR stressors can impose greater protein secretory demands on the endoplasmic reticulum (ER), resulting in the development, differentiation, and survival of these cell types to meet these increasing functional needs. Dysregulation of the UPR leads to the development of the disease. The UPR and ER stress are involved in several human conditions, such as chronic inflammation, neurodegeneration, metabolic syndrome, and cancer. Furthermore, potent and specific compounds that target the UPR pathway are under development as future therapies. The focus of this review is to thoroughly describe the effects of both internal and external stressors on the ER in asthma. Furthermore, we discuss how the UPR signaling pathway is activated in the lungs to overcome cellular damage. We also present an overview of the pathogenic mechanisms, with a brief focus on potential strategies for pharmacological interventions.
Collapse
Affiliation(s)
- Sanaz Dastghaib
- Department of Clinical Biochemistry and
- Autophagy Research Center, Health Policy Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| | - P Sravan Kumar
- National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Sajjad Aftabi
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine
- Medical Physics Department and
| | - Gautam Damera
- Personalized and Predictive Medicine (Respiratory), Global Research and Development, Teva Pharmaceuticals, Malvern, Pennsylvania
| | - Azadeh Dalvand
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine
| | - Adel Sepanjnia
- Department of Immunology, School of Medicine, Jiroft University of Medical Science, Jiroft, Iran
| | - Mohammad Kiumarsi
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine
| | - Mohamad-Reza Aghanoori
- Department of Human Genetics, School of Medicine, and
- Department of Pharmacology and Therapeutics
- Division of Neurodegenerative Disorders, Albrechtsen Research Centre, St. Boniface Hospital, Winnipeg, Manitoba, Canada
| | - Sukhwinder Singh Sohal
- Respiratory Translational Research Group, Department of Laboratory Medicine, College of Health and Medicine, University of Tasmania, Launceston, Tasmania, Australia
| | | | - Javad Alizadeh
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine
- Research Institute of Oncology and Hematology, Cancer Care Manitoba-University of Manitoba, Winnipeg, Manitoba, Canada
| | - Pooneh Mokarram
- Department of Clinical Biochemistry and
- Autophagy Research Center, Health Policy Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saeid Ghavami
- Autophagy Research Center, Health Policy Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine
- Department of Internal Medicine, and
- Biology of Breathing Theme, Children Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Pawan Sharma
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Amir A Zeki
- Lung Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, Davis, California; and
- Veterans Affairs Medical Center, Mather, California
| |
Collapse
|
31
|
Lee SK. Endoplasmic Reticulum Homeostasis and Stress Responses in Caenorhabditis elegans. PROGRESS IN MOLECULAR AND SUBCELLULAR BIOLOGY 2021; 59:279-303. [PMID: 34050871 DOI: 10.1007/978-3-030-67696-4_13] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The unfolded protein response (UPR) is an evolutionarily conserved adaptive regulatory pathway that alleviates protein-folding defects in the endoplasmic reticulum (ER). Physiological demands, environmental perturbations and pathological conditions can cause accumulation of unfolded proteins in the ER and the stress signal is transmitted to the nucleus to turn on a series of genes to respond the challenge. In metazoan, the UPR pathways consisted of IRE1/XBP1, PEK-1 and ATF6, which function in parallel and downstream transcriptional activation triggers the proteostasis networks consisting of molecular chaperones, protein degradation machinery and other stress response pathways ((Labbadia J, Morimoto RI, F1000Prime Rep 6:7, 2014); (Shen X, Ellis RE, Lee K, Annu Rev Biochem 28:893-903, 2014)). The integrated responses act on to resolve the ER stress by increasing protein folding capacity, attenuating ER-loading translation, activating ER-associated proteasomal degradation (ERAD), and regulating IRE1-dependent decay of mRNA (RIDD). Therefore, the effective UPR to internal and external causes is linked to the multiple pathophysiological conditions such as aging, immunity, and neurodegenerative diseases. Recent development in the research of the UPR includes cell-nonautonomous features of the UPR, interplay between the UPR and other stress response pathways, unconventional UPR inducers, and noncanonical UPR independent of the three major branches, originated from multiple cellular and molecular machineries in addition to ER. Caenorhabditis elegans model system has critically contributed to these unprecedented aspects of the ER UPR and broadens the possible therapeutic targets to treat the ER-stress associated human disorders and time-dependent physiological deterioration of aging.
Collapse
Affiliation(s)
- Sun-Kyung Lee
- Department of Life Science, Research Institute for Natural Sciences, College of Natural Sciences, Hanyang University, Seoul, Republic of Korea.
| |
Collapse
|
32
|
Liu Y, Tan Z, Yang Y. Negative feedback and modern anti-cancer strategies targeting the ER stress response. FEBS Lett 2020; 594:4247-4265. [PMID: 33206409 DOI: 10.1002/1873-3468.14000] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 09/18/2020] [Accepted: 10/25/2020] [Indexed: 12/13/2022]
Abstract
Endoplasmic reticulum (ER) stress is a cell state in which misfolded or unfolded proteins are aberrantly accumulated in the ER. ER stress induces an evolutionarily conserved adaptive response, named the ER stress response, that deploys a self-regulated machinery to maintain cellular proteostasis. However, compared to its well-established canonical activation mechanism, the negative feedback mechanisms regulating the ER stress response remain unclear and no accepted methods or markers have been established. Several studies have documented that both endogenous and exogenous insults can induce ER stress in cancer. Based on this evidence, small molecule inhibitors targeting ER stress response have been designed to kill cancer cells, with some of them showing excellent curative effects. Here, we review recent advances in our understanding of negative feedback of the ER stress response and compare the markers used to date. We also summarize therapeutic inhibitors targeting ER stress response and highlight the promises and challenges ahead.
Collapse
Affiliation(s)
- Yaofu Liu
- Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, China
| | - Zhenzhi Tan
- Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, China
| | - Yili Yang
- Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, China
| |
Collapse
|
33
|
Preissler S, Rato C, Yan Y, Perera LA, Czako A, Ron D. Calcium depletion challenges endoplasmic reticulum proteostasis by destabilising BiP-substrate complexes. eLife 2020; 9:62601. [PMID: 33295873 PMCID: PMC7758071 DOI: 10.7554/elife.62601] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 12/08/2020] [Indexed: 12/14/2022] Open
Abstract
The metazoan endoplasmic reticulum (ER) serves both as a hub for maturation of secreted proteins and as an intracellular calcium storage compartment, facilitating calcium-release-dependent cellular processes. ER calcium depletion robustly activates the unfolded protein response (UPR). However, it is unclear how fluctuations in ER calcium impact organellar proteostasis. Here, we report that calcium selectively affects the dynamics of the abundant metazoan ER Hsp70 chaperone BiP, by enhancing its affinity for ADP. In the calcium-replete ER, ADP rebinding to post-ATP hydrolysis BiP-substrate complexes competes with ATP binding during both spontaneous and co-chaperone-assisted nucleotide exchange, favouring substrate retention. Conversely, in the calcium-depleted ER, relative acceleration of ADP-to-ATP exchange favours substrate release. These findings explain the rapid dissociation of certain substrates from BiP observed in the calcium-depleted ER and suggest a mechanism for tuning ER quality control and coupling UPR activity to signals that mobilise ER calcium in secretory cells.
Collapse
Affiliation(s)
- Steffen Preissler
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Claudia Rato
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Yahui Yan
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Luke A Perera
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Aron Czako
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - David Ron
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
34
|
Urra H, Pihán P, Hetz C. The UPRosome - decoding novel biological outputs of IRE1α function. J Cell Sci 2020; 133:133/15/jcs218107. [PMID: 32788208 DOI: 10.1242/jcs.218107] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Different perturbations alter the function of the endoplasmic reticulum (ER), resulting in the accumulation of misfolded proteins in its lumen, a condition termed ER stress. To restore ER proteostasis, a highly conserved pathway is engaged, known as the unfolded protein response (UPR), triggering adaptive programs or apoptosis of terminally damaged cells. IRE1α (also known as ERN1), the most conserved UPR sensor, mediates the activation of responses to determine cell fate under ER stress. The complexity of IRE1α regulation and its signaling outputs is mediated in part by the assembly of a dynamic multi-protein complex, named the UPRosome, that regulates IRE1α activity and the crosstalk with other pathways. We discuss several studies identifying components of the UPRosome that have illuminated novel functions in cell death, autophagy, DNA damage, energy metabolism and cytoskeleton dynamics. Here, we provide a theoretical analysis to assess the biological significance of the UPRosome and present the results of a systematic bioinformatics analysis of the available IRE1α interactome data sets followed by functional enrichment clustering. This in silico approach decoded that IRE1α also interacts with proteins involved in the cell cycle, transport, differentiation, response to viral infection and immune response. Thus, defining the spectrum of IRE1α-binding partners will reveal novel signaling outputs and the relevance of the pathway to human diseases.
Collapse
Affiliation(s)
- Hery Urra
- Biomedical Neuroscience Institute (BNI), Faculty of Medicine, University of Chile, Santiago 8380453, Chile .,Center for Geroscience, Brain Health and Metabolism (GERO), Santiago 7800003, Chile.,Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), University of Chile, Santiago 8380453, Chile
| | - Philippe Pihán
- Biomedical Neuroscience Institute (BNI), Faculty of Medicine, University of Chile, Santiago 8380453, Chile.,Center for Geroscience, Brain Health and Metabolism (GERO), Santiago 7800003, Chile.,Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), University of Chile, Santiago 8380453, Chile
| | - Claudio Hetz
- Biomedical Neuroscience Institute (BNI), Faculty of Medicine, University of Chile, Santiago 8380453, Chile .,Center for Geroscience, Brain Health and Metabolism (GERO), Santiago 7800003, Chile.,Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), University of Chile, Santiago 8380453, Chile.,The Buck Institute for Research in Aging, Novato, CA 94945, USA
| |
Collapse
|
35
|
Hetz C, Zhang K, Kaufman RJ. Mechanisms, regulation and functions of the unfolded protein response. Nat Rev Mol Cell Biol 2020; 21:421-438. [PMID: 32457508 DOI: 10.1038/s41580-020-0250-z] [Citation(s) in RCA: 1546] [Impact Index Per Article: 309.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/14/2020] [Indexed: 12/21/2022]
Abstract
Cellular stress induced by the abnormal accumulation of unfolded or misfolded proteins at the endoplasmic reticulum (ER) is emerging as a possible driver of human diseases, including cancer, diabetes, obesity and neurodegeneration. ER proteostasis surveillance is mediated by the unfolded protein response (UPR), a signal transduction pathway that senses the fidelity of protein folding in the ER lumen. The UPR transmits information about protein folding status to the nucleus and cytosol to adjust the protein folding capacity of the cell or, in the event of chronic damage, induce apoptotic cell death. Recent advances in the understanding of the regulation of UPR signalling and its implications in the pathophysiology of disease might open new therapeutic avenues.
Collapse
Affiliation(s)
- Claudio Hetz
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile. .,FONDAP Center for Geroscience Brain Health and Metabolism (GERO), Santiago, Chile. .,Program of Cellular and Molecular Biology, Institute of Biomedical Science, University of Chile, Santiago, Chile. .,Buck Institute for Research on Aging, Novato, CA, USA.
| | - Kezhong Zhang
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA. .,Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, MI, USA.
| | - Randal J Kaufman
- Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.
| |
Collapse
|
36
|
Xu C, Su X, Chen Y, Xu Y, Wang Z, Mo X. Proteomics analysis of plasma protein changes in patent ductus arteriosus patients. Ital J Pediatr 2020; 46:64. [PMID: 32430045 PMCID: PMC7236322 DOI: 10.1186/s13052-020-00831-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 05/11/2020] [Indexed: 11/23/2022] Open
Abstract
Objective Patent ductus arteriosus (PDA) is a congenital heart defect with an unclear etiology that occurs commonly among newborns. Adequately understanding the molecular pathogenesis of PDA can contribute to improved treatment and prevention. Plasma proteins may provide evidence to explore the molecular mechanisms of abnormal cardiac development. Methods Isobaric tags for relative and absolute quantitation (iTRAQ) proteomics technology was used to measure different plasma proteins in PDA patients (n = 4) and controls (n = 4). The candidate protein was validated by ELISA and Western blot (WB) assays in a larger sample. Validation of the location and expression of this protein was performed in mouse heart sections. Results There were three downregulated proteins and eight upregulated proteins identified in the iTRAQ proteomics data. Among these, protein disulfide-isomerase A6 (PDIA6) was further analyzed for validation. The plasma PDIA6 concentrations (3.2 ± 0.7 ng/ml) in PDA patients were significantly lower than those in normal controls (5.8 ± 1.2 ng/ml). In addition, a WB assay also supported these results. PDIA6 was widely expressed in mouse heart outflow tract on embryonic day 14.5. Conclusion Plasma proteomics profiles suggested novel candidate molecular markers for PDA. The findings may allow development of a new strategy to investigate the mechanism and etiology of PDA.
Collapse
Affiliation(s)
- Cheng Xu
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China
| | - Xiaoqi Su
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China
| | - Yong Chen
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China
| | - Yang Xu
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China
| | - Zhiqi Wang
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China
| | - Xuming Mo
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China.
| |
Collapse
|
37
|
Paraoan L, Sharif U, Carlsson E, Supharattanasitthi W, Mahmud NM, Kamalden TA, Hiscott P, Jackson M, Grierson I. Secretory proteostasis of the retinal pigmented epithelium: Impairment links to age-related macular degeneration. Prog Retin Eye Res 2020; 79:100859. [PMID: 32278708 DOI: 10.1016/j.preteyeres.2020.100859] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 03/24/2020] [Accepted: 03/26/2020] [Indexed: 12/19/2022]
Abstract
Secretory proteostasis integrates protein synthesis, processing, folding and trafficking pathways that are essential for efficient cellular secretion. For the retinal pigment epithelium (RPE), secretory proteostasis is of vital importance for the maintenance of the structural and functional integrity of apical (photoreceptors) and basal (Bruch's membrane/choroidal blood supply) sides of the environment it resides in. This integrity is achieved through functions governed by RPE secreted proteins, which include extracellular matrix modelling/remodelling, angiogenesis and immune response modulation. Impaired RPE secretory proteostasis affects not only the extracellular environment, but leads to intracellular protein aggregation and ER-stress with subsequent cell death. Ample recent evidence implicates dysregulated proteostasis as a key factor in the development of age-related macular degeneration (AMD), the leading cause of blindness in the developed world, and research aiming to characterise the roles of various proteins implicated in AMD-associated dysregulated proteostasis unveiled unexpected facets of the mechanisms involved in degenerative pathogenesis. This review analyses cellular processes unveiled by the study of the top 200 transcripts most abundantly expressed by the RPE/choroid in the light of the specialised secretory nature of the RPE. Functional roles of these proteins and the mechanisms of their impaired secretion, due to age and genetic-related causes, are analysed in relation to AMD development. Understanding the importance of RPE secretory proteostasis in relation to maintaining retinal health and how it becomes impaired in disease is of paramount importance for the development and assessment of future therapeutic advancements involving gene and cell therapies.
Collapse
Affiliation(s)
- Luminita Paraoan
- Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom.
| | - Umar Sharif
- Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | - Emil Carlsson
- Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | - Wasu Supharattanasitthi
- Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom; Department of Physiology, Faculty of Pharmacy, Mahidol University, Bangkok, Thailand
| | - Nur Musfirah Mahmud
- Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | - Tengku Ain Kamalden
- Eye Research Centre, Department of Ophthalmology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Paul Hiscott
- Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | - Malcolm Jackson
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | - Ian Grierson
- Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
38
|
Lebeaupin C, Yong J, Kaufman RJ. The Impact of the ER Unfolded Protein Response on Cancer Initiation and Progression: Therapeutic Implications. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1243:113-131. [PMID: 32297215 PMCID: PMC7243802 DOI: 10.1007/978-3-030-40204-4_8] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Cellular stress induced by the accumulation of misfolded proteins in the endoplasmic reticulum (ER) activates an elaborate signalling network termed the unfolded protein response (UPR). This adaptive response is mediated by the transmembrane signal transducers IRE1, PERK, and ATF6 to decide cell fate of recovery or death. In malignant cells, UPR signalling may be required to maintain ER homeostasis and survival in the tumor microenvironment characterized by oxidative stress, hypoxia, lactic acidosis and compromised protein folding. Here we provide an overview of the ER response to cellular stress and how the sustained activation of this network enables malignant cells to develop tumorigenic, metastatic and drug-resistant capacities to thrive under adverse conditions. Understanding the complexity of ER stress responses and how to target the UPR in disease will have significant potential for novel future therapeutics.
Collapse
Affiliation(s)
- Cynthia Lebeaupin
- Degenerative Diseases Program, SBP Medical Discovery Institute, La Jolla, CA, USA
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Jing Yong
- Degenerative Diseases Program, SBP Medical Discovery Institute, La Jolla, CA, USA
| | - Randal J Kaufman
- Degenerative Diseases Program, SBP Medical Discovery Institute, La Jolla, CA, USA.
| |
Collapse
|
39
|
Wang WA, Agellon LB, Michalak M. Organellar Calcium Handling in the Cellular Reticular Network. Cold Spring Harb Perspect Biol 2019; 11:cshperspect.a038265. [PMID: 31358518 DOI: 10.1101/cshperspect.a038265] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Ca2+ is an important intracellular messenger affecting diverse cellular processes. In eukaryotic cells, Ca2+ is handled by a myriad of Ca2+-binding proteins found in organelles that are organized into the cellular reticular network (CRN). The network is comprised of the endoplasmic reticulum, Golgi apparatus, lysosomes, membranous components of the endocytic and exocytic pathways, peroxisomes, and the nuclear envelope. Membrane contact sites between the different components of the CRN enable the rapid movement of Ca2+, and communication of Ca2+ status, within the network. Ca2+-handling proteins that reside in the CRN facilitate Ca2+ sensing, buffering, and cellular signaling to coordinate the many processes that operate within the cell.
Collapse
Affiliation(s)
- Wen-An Wang
- Department of Biochemistry, University of Alberta, Edmonton, Alberta T6G 2S7, Canada
| | - Luis B Agellon
- School of Human Nutrition, McGill University, Ste. Anne de Bellevue, Quebec H9X 3V9, Canada
| | - Marek Michalak
- Department of Biochemistry, University of Alberta, Edmonton, Alberta T6G 2S7, Canada
| |
Collapse
|
40
|
Nam SM, Jeon YJ. Proteostasis In The Endoplasmic Reticulum: Road to Cure. Cancers (Basel) 2019; 11:E1793. [PMID: 31739582 PMCID: PMC6895847 DOI: 10.3390/cancers11111793] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 11/04/2019] [Accepted: 11/12/2019] [Indexed: 12/14/2022] Open
Abstract
The endoplasmic reticulum (ER) is an interconnected organelle that is responsible for the biosynthesis, folding, maturation, stabilization, and trafficking of transmembrane and secretory proteins. Therefore, cells evolve protein quality-control equipment of the ER to ensure protein homeostasis, also termed proteostasis. However, disruption in the folding capacity of the ER caused by a large variety of pathophysiological insults leads to the accumulation of unfolded or misfolded proteins in this organelle, known as ER stress. Upon ER stress, unfolded protein response (UPR) of the ER is activated, integrates ER stress signals, and transduces the integrated signals to relive ER stress, thereby leading to the re-establishment of proteostasis. Intriguingly, severe and persistent ER stress and the subsequently sustained unfolded protein response (UPR) are closely associated with tumor development, angiogenesis, aggressiveness, immunosuppression, and therapeutic response of cancer. Additionally, the UPR interconnects various processes in and around the tumor microenvironment. Therefore, it has begun to be delineated that pharmacologically and genetically manipulating strategies directed to target the UPR of the ER might exhibit positive clinical outcome in cancer. In the present review, we summarize recent advances in our understanding of the UPR of the ER and the UPR of the ER-mitochondria interconnection. We also highlight new insights into how the UPR of the ER in response to pathophysiological perturbations is implicated in the pathogenesis of cancer. We provide the concept to target the UPR of the ER, eventually discussing the potential of therapeutic interventions for targeting the UPR of the ER for cancer treatment.
Collapse
Affiliation(s)
- Su Min Nam
- Department of Biochemistry, Chungnam National University College of Medicine, Daejeon 35015, Korea;
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon 35015, Korea
| | - Young Joo Jeon
- Department of Biochemistry, Chungnam National University College of Medicine, Daejeon 35015, Korea;
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon 35015, Korea
| |
Collapse
|
41
|
Agellon LB, Michalak M. Avoiding raising the ire of IRE1α. Cell Calcium 2019; 83:102056. [DOI: 10.1016/j.ceca.2019.102056] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 07/17/2019] [Indexed: 01/06/2023]
|
42
|
Transcription Factor and miRNA Interplays Can Manifest the Survival of ccRCC Patients. Cancers (Basel) 2019; 11:cancers11111668. [PMID: 31661791 PMCID: PMC6895828 DOI: 10.3390/cancers11111668] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 10/21/2019] [Accepted: 10/24/2019] [Indexed: 12/15/2022] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) still remains a higher mortality rate in worldwide. Obtaining promising biomakers is very crucial for improving the diagnosis and prognosis of ccRCC patients. Herein, we firstly identified eight potentially prognostic miRNAs (hsa-miR-144-5p, hsa-miR-223-3p, hsa-miR-365b-3p, hsa-miR-3613-5p, hsa-miR-9-5p, hsa-miR-183-5p, hsa-miR-335-3p, hsa-miR-1269a). Secondly, we found that a signature containing these eight miRNAs showed obviously superior to a single miRNA in the prognostic effect and credibility for predicting the survival of ccRCC patients. Thirdly, we discovered that twenty-two transcription factors (TFs) interact with these eight miRNAs, and a signature combining nine TFs (TFAP2A, KLF5, IRF1, RUNX1, RARA, GATA3, IKZF1, POU2F2, and FOXM1) could promote the prognosis of ccRCC patients. Finally, we further identified eleven genes (hsa-miR-365b-3p, hsa-miR-223-3p, hsa-miR-1269a, hsa-miR-144-5p, hsa-miR-183-5p, hsa-miR-335-3p, TFAP2A, KLF5, IRF1, MYC, IKZF1) that could combine as a signature to improve the prognosis effect of ccRCC patients, which distinctly outperformed the eight-miRNA signature and the nine-TF signature. Overall, we identified several new prognosis factors for ccRCC, and revealed a potential mechanism that TFs and miRNAs interplay cooperatively or oppositely regulate a certain number of tumor suppressors, driver genes, and oncogenes to facilitate the survival of ccRCC patients.
Collapse
|
43
|
Wang Q, Groenendyk J, Paskevicius T, Qin W, Kor KC, Liu Y, Hiess F, Knollmann BC, Chen SRW, Tang J, Chen XZ, Agellon LB, Michalak M. Two pools of IRE1α in cardiac and skeletal muscle cells. FASEB J 2019; 33:8892-8904. [PMID: 31051095 PMCID: PMC6662970 DOI: 10.1096/fj.201802626r] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 04/08/2019] [Indexed: 12/23/2022]
Abstract
The endoplasmic reticulum (ER) plays a central role in cellular stress responses via mobilization of ER stress coping responses, such as the unfolded protein response (UPR). The inositol-requiring 1α (IRE1α) is an ER stress sensor and component of the UPR. Muscle cells also have a well-developed and highly subspecialized membrane network of smooth ER called the sarcoplasmic reticulum (SR) surrounding myofibrils and specialized for Ca2+ storage, release, and uptake to control muscle excitation-contraction coupling. Here, we describe 2 distinct pools of IRE1α in cardiac and skeletal muscle cells, one localized at the perinuclear ER and the other at the junctional SR. We discovered that, at the junctional SR, calsequestrin binds to the ER luminal domain of IRE1α, inhibiting its dimerization. This novel interaction of IRE1α with calsequestrin, one of the highly abundant Ca2+ handling proteins at the junctional SR, provides new insights into the regulation of stress coping responses in muscle cells.-Wang, Q., Groenendyk, J., Paskevicius, T., Qin, W., Kor, K. C., Liu, Y., Hiess, F., Knollmann, B. C., Chen, S. R. W., Tang, J., Chen, X.-Z., Agellon, L. B., Michalak, M. Two pools of IRE1α in cardiac and skeletal muscle cells.
Collapse
Affiliation(s)
- Qian Wang
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Jody Groenendyk
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | | | - Wenying Qin
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, China
| | - Kaylen C. Kor
- Division of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Yingjie Liu
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Florian Hiess
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Bjorn C. Knollmann
- Division of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - S. R. Wayne Chen
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Jingfeng Tang
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, China
| | - Xing-Zhen Chen
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, China
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada; and
| | - Luis B. Agellon
- School of Human Nutrition, McGill University, Ste. Anne de Bellevue, Quebec, Canada
| | - Marek Michalak
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, China
| |
Collapse
|
44
|
Urra H, Hetz C. Fine-tuning PERK signaling to control cell fate under stress. Nat Struct Mol Biol 2019; 24:789-790. [PMID: 28981072 DOI: 10.1038/nsmb.3478] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Hery Urra
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile, the Center for Geroscience, Brain Health and Metabolism, Santiago, Chile, and the Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Claudio Hetz
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile, the Center for Geroscience, Brain Health and Metabolism, Santiago, Chile, and the Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile.,Buck Institute for Research in Aging, Novato, California, USA and in the Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, Massachusetts, USA
| |
Collapse
|
45
|
Groenendyk J, Fan X, Peng Z, Kurgan L, Michalak M. Endoplasmic reticulum and the microRNA environment in the cardiovascular system 1. Can J Physiol Pharmacol 2019; 97:515-527. [PMID: 31063413 DOI: 10.1139/cjpp-2018-0720] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Stress responses are important to human physiology and pathology, and the inability to adapt to cellular stress leads to cell death. To mitigate cellular stress and re-establish homeostasis, cells, including those in the cardiovascular system, activate stress coping response mechanisms. The endoplasmic reticulum, a component of the cellular reticular network in cardiac cells, mobilizes so-called endoplasmic reticulum stress coping responses, such as the unfolded protein response. MicroRNAs play an important part in the maintenance of cellular and tissue homeostasis, perform a central role in the biology of the cardiac myocyte, and are involved in pathological cardiac function and remodeling. In this paper, we review a link between endoplasmic reticulum homeostasis and microRNA with an emphasis on the impact on stress responses in the cardiovascular system.
Collapse
Affiliation(s)
- Jody Groenendyk
- a Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2S7, Canada
| | - Xiao Fan
- b Irving Medical Center, Columbia University, New York, NY 10032, USA
| | - Zhenling Peng
- c Center for Applied Mathematics, Tianjin University, Tianjin 300072, China
| | - Lukasz Kurgan
- d Department of Computer Science, Virginia Commonwealth University, Richmond, VA 23284, USA.,e Department of Electrical and Computer Engineering, University of Alberta, Edmonton, AB T6G 2V4, Canada
| | - Marek Michalak
- a Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2S7, Canada
| |
Collapse
|
46
|
Emerging Roles of the Endoplasmic Reticulum Associated Unfolded Protein Response in Cancer Cell Migration and Invasion. Cancers (Basel) 2019; 11:cancers11050631. [PMID: 31064137 PMCID: PMC6562633 DOI: 10.3390/cancers11050631] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 04/29/2019] [Accepted: 05/01/2019] [Indexed: 12/21/2022] Open
Abstract
Endoplasmic reticulum (ER) proteostasis is often altered in tumor cells due to intrinsic (oncogene expression, aneuploidy) and extrinsic (environmental) challenges. ER stress triggers the activation of an adaptive response named the Unfolded Protein Response (UPR), leading to protein translation repression, and to the improvement of ER protein folding and clearance capacity. The UPR is emerging as a key player in malignant transformation and tumor growth, impacting on most hallmarks of cancer. As such, the UPR can influence cancer cells’ migration and invasion properties. In this review, we overview the involvement of the UPR in cancer progression. We discuss its cross-talks with the cell migration and invasion machinery. Specific aspects will be covered including extracellular matrix (ECM) remodeling, modification of cell adhesion, chemo-attraction, epithelial-mesenchymal transition (EMT), modulation of signaling pathways associated with cell mobility, and cytoskeleton remodeling. The therapeutic potential of targeting the UPR to treat cancer will also be considered with specific emphasis in the impact on metastasis and tissue invasion.
Collapse
|
47
|
Matsusaki M, Kanemura S, Kinoshita M, Lee YH, Inaba K, Okumura M. The Protein Disulfide Isomerase Family: from proteostasis to pathogenesis. Biochim Biophys Acta Gen Subj 2019; 1864:129338. [PMID: 30986509 DOI: 10.1016/j.bbagen.2019.04.003] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 03/08/2019] [Accepted: 04/02/2019] [Indexed: 12/13/2022]
Abstract
In mammalian cells, nearly one-third of proteins are inserted into the endoplasmic reticulum (ER), where they undergo oxidative folding and chaperoning assisted by approximately 20 members of the protein disulfide isomerase family (PDIs). PDIs consist of multiple thioredoxin-like domains and recognize a wide variety of proteins via highly conserved interdomain flexibility. Although PDIs have been studied intensely for almost 50 years, exactly how they maintain protein homeostasis in the ER remains unknown, and is important not only for fundamental biological understanding but also for protein misfolding- and aggregation-related pathophysiology. Herein, we review recent advances in structural biology and biophysical approaches that explore the underlying mechanism by which PDIs fulfil their distinct functions to promote productive protein folding and scavenge misfolded proteins in the ER, the primary factory for efficient production of the secretome.
Collapse
Affiliation(s)
- Motonori Matsusaki
- Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Aramaki aza Aoba 6-3, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Shingo Kanemura
- Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Aramaki aza Aoba 6-3, Aoba-ku, Sendai, Miyagi 980-8578, Japan; School of Science and Technology, Kwansei Gakuin University, Gakuen 2-1, Sanda, Hyogo 669-1337, Japan
| | - Misaki Kinoshita
- Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Aramaki aza Aoba 6-3, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Young-Ho Lee
- Protein Structure Group, Korea Basic Science Institute, Ochang, Chungbuk 28199, South Korea; Bio-Analytical Science, University of Science and Technology, Daejeon 34113, South Korea
| | - Kenji Inaba
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Katahira 2-1-1, Aoba-ku, Sendai, Miyagi 980-8577, Japan.
| | - Masaki Okumura
- Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Aramaki aza Aoba 6-3, Aoba-ku, Sendai, Miyagi 980-8578, Japan.
| |
Collapse
|
48
|
Yang H, Xue Y, Kuang S, Zhang M, Chen J, Liu L, Shan Z, Lin Q, Li X, Yang M, Zhou H, Rao F, Deng C. Involvement of Orai1 in tunicamycin-induced endothelial dysfunction. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2019; 23:95-102. [PMID: 30820153 PMCID: PMC6384200 DOI: 10.4196/kjpp.2019.23.2.95] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Revised: 09/04/2018] [Accepted: 10/10/2018] [Indexed: 12/28/2022]
Abstract
Endoplasmic reticulum (ER) stress is mediated by disturbance of Ca2+ homeostasis. The store-operated calcium (SOC) channel is the primary Ca2+ channel in non-excitable cells, but its participation in agent-induced ER stress is not clear. In this study, the effects of tunicamycin on Ca2+ influx in human umbilical vein endothelial cells (HUVECs) were observed with the fluorescent probe Fluo-4 AM. The effect of tunicamycin on the expression of the unfolded protein response (UPR)-related proteins BiP and CHOP was assayed by western blotting with or without inhibition of Orai1. Tunicamycin induced endothelial dysfunction by activating ER stress. Orai1 expression and the influx of extracellular Ca2+ in HUVECs were both upregulated during ER stress. The SOC channel inhibitor SKF96365 reversed tunicamycin-induced endothelial cell dysfunction by inhibiting ER stress. Regulation of tunicamycin-induced ER stress by Orai1 indicates that modification of Orai1 activity may have therapeutic value for conditions with ER stress-induced endothelial dysfunction.
Collapse
Affiliation(s)
- Hui Yang
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangzhou, Guangdong 510080, China.,Research Center of Medical Sciences, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, China
| | - Yumei Xue
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangzhou, Guangdong 510080, China.,Research Center of Medical Sciences, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, China
| | - Sujuan Kuang
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangzhou, Guangdong 510080, China.,Research Center of Medical Sciences, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, China
| | - Mengzhen Zhang
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangzhou, Guangdong 510080, China.,Research Center of Medical Sciences, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, China
| | - Jinghui Chen
- Department of Anesthesiology, Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong 510623, China
| | - Lin Liu
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangzhou, Guangdong 510080, China.,Research Center of Medical Sciences, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, China
| | - Zhixin Shan
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangzhou, Guangdong 510080, China.,Research Center of Medical Sciences, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, China
| | - Qiuxiong Lin
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangzhou, Guangdong 510080, China.,Research Center of Medical Sciences, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, China
| | - Xiaohong Li
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangzhou, Guangdong 510080, China.,Research Center of Medical Sciences, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, China
| | - Min Yang
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangzhou, Guangdong 510080, China.,Research Center of Medical Sciences, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, China
| | - Hui Zhou
- Department of Pharmacy, Guangzhou Panyu Shiqiao Hospital, Guangzhou, Guangdong 511400, China
| | - Fang Rao
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangzhou, Guangdong 510080, China.,Research Center of Medical Sciences, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, China
| | - Chunyu Deng
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangzhou, Guangdong 510080, China.,Research Center of Medical Sciences, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, China
| |
Collapse
|
49
|
Mennerich D, Kellokumpu S, Kietzmann T. Hypoxia and Reactive Oxygen Species as Modulators of Endoplasmic Reticulum and Golgi Homeostasis. Antioxid Redox Signal 2019; 30:113-137. [PMID: 29717631 DOI: 10.1089/ars.2018.7523] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
SIGNIFICANCE Eukaryotic cells execute various functions in subcellular compartments or organelles for which cellular redox homeostasis is of importance. Apart from mitochondria, hypoxia and stress-mediated formation of reactive oxygen species (ROS) were shown to modulate endoplasmic reticulum (ER) and Golgi apparatus (GA) functions. Recent Advances: Research during the last decade has improved our understanding of disulfide bond formation, protein glycosylation and secretion, as well as pH and redox homeostasis in the ER and GA. Thus, oxygen (O2) itself, NADPH oxidase (NOX) formed ROS, and pH changes appear to be of importance and indicate the intricate balance of intercompartmental communication. CRITICAL ISSUES Although the interplay between hypoxia, ER stress, and Golgi function is evident, the existence of more than 20 protein disulfide isomerase family members and the relative mild phenotypes of, for example, endoplasmic reticulum oxidoreductin 1 (ERO1)- and NOX4-knockout mice clearly suggest the existence of redundant and alternative pathways, which remain largely elusive. FUTURE DIRECTIONS The identification of these pathways and the key players involved in intercompartmental communication needs suitable animal models, genome-wide association, as well as proteomic studies in humans. The results of those studies will be beneficial for the understanding of the etiology of diseases such as type 2 diabetes, Alzheimer's disease, and cancer, which are associated with ROS, protein aggregation, and glycosylation defects.
Collapse
Affiliation(s)
- Daniela Mennerich
- Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu , Oulu, Finland
| | - Sakari Kellokumpu
- Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu , Oulu, Finland
| | - Thomas Kietzmann
- Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu , Oulu, Finland
| |
Collapse
|
50
|
Wohlrab P, Soto-Gonzales L, Benesch T, Winter MP, Lang IM, Markstaller K, Tretter V, Klein KU. Intermittent Hypoxia Activates Duration-Dependent Protective and Injurious Mechanisms in Mouse Lung Endothelial Cells. Front Physiol 2018; 9:1754. [PMID: 30574096 PMCID: PMC6291480 DOI: 10.3389/fphys.2018.01754] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 11/20/2018] [Indexed: 12/31/2022] Open
Abstract
Intermittent hypoxia is a major factor in clinical conditions like the obstructive sleep apnea syndrome or the cyclic recruitment and derecruitment of atelectasis in acute respiratory distress syndrome and positive pressure mechanical ventilation. In vivo investigations of the direct impact of intermittent hypoxia are frequently hampered by multiple co-morbidities of patients. Therefore, cell culture experiments are important model systems to elucidate molecular mechanisms that are involved in the cellular response to alternating oxygen conditions and could represent future targets for tailored therapies. In this study, we focused on mouse lung endothelial cells as a first frontier to encounter altered oxygen due to disturbances in airway or lung function, that play an important role in the development of secondary diseases like vascular disease and pulmonary hypertension. We analyzed key markers for endothelial function including cell adhesion molecules, molecules involved in regulation of fibrinolysis, hemostasis, redox balance, and regulators of gene expression like miRNAs. Results show that short-time exposure to intermittent hypoxia has little impact on vitality and health of cells. At early timepoints and up to 24 h, many endothelial markers are unchanged in their expression and some indicators of injury are even downregulated. However, in the long-term, multiple signaling pathways are activated, that ultimately result in cellular inflammation, oxidative stress, and apoptosis.
Collapse
Affiliation(s)
- Peter Wohlrab
- Department of Anaesthesia, General Intensive Care and Pain Management, Medical University of Vienna, Vienna, Austria
| | - Lourdes Soto-Gonzales
- Department of Anaesthesia, General Intensive Care and Pain Management, Medical University of Vienna, Vienna, Austria
| | - Thomas Benesch
- Institute for International Development, University of Vienna, Vienna, Austria
| | - Max Paul Winter
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Irene Marthe Lang
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Klaus Markstaller
- Department of Anaesthesia, General Intensive Care and Pain Management, Medical University of Vienna, Vienna, Austria
| | - Verena Tretter
- Department of Anaesthesia, General Intensive Care and Pain Management, Medical University of Vienna, Vienna, Austria
| | - Klaus Ulrich Klein
- Department of Anaesthesia, General Intensive Care and Pain Management, Medical University of Vienna, Vienna, Austria
| |
Collapse
|