1
|
Chen Z, Herzog RW, Kaufman RJ. Cellular stress and coagulation factor production: when more is not necessarily better. J Thromb Haemost 2023; 21:3329-3341. [PMID: 37839613 PMCID: PMC10760459 DOI: 10.1016/j.jtha.2023.10.005] [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: 10/10/2023] [Accepted: 10/11/2023] [Indexed: 10/17/2023]
Abstract
Remarkably, it has been 40 years since the isolation of the 2 genes involved in hemophilia A (HA) and hemophilia B (HB), encoding clotting factor (F) VIII (FVIII) and FIX, respectively. Over the years, these advances led to the development of purified recombinant protein factors that are free of contaminating viruses from human pooled plasma for hemophilia treatments, reducing the morbidity and mortality previously associated with human plasma-derived clotting factors. These discoveries also paved the way for modified factors that have increased plasma half-lives. Importantly, more recent advances have led to the development and Food and Drug Administration approval of a hepatocyte-targeted, adeno-associated viral vector-mediated gene transfer approach for HA and HB. However, major concerns regarding the durability and safety of HA gene therapy remain to be resolved. Compared with FIX, FVIII is a much larger protein that is prone to misfolding and aggregation in the endoplasmic reticulum and is poorly secreted by the mammalian cells. Due to the constraint of the packaging capacity of adeno-associated viral vector, B-domain deleted FVIII rather than the full-length protein is used for HA gene therapy. Like full-length FVIII, B-domain deleted FVIII misfolds and is inefficiently secreted. Its expression in hepatocytes activates the cellular unfolded protein response, which is deleterious for hepatocyte function and survival and has the potential to drive hepatocellular carcinoma. This review is focused on our current understanding of factors limiting FVIII secretion and the potential pathophysiological consequences upon expression in hepatocytes.
Collapse
Affiliation(s)
- Zhouji Chen
- Degenerative Diseases Program, Center for Genetic Diseases and Aging Research, SBP Medical Discovery Institute, California, USA
| | - Roland W Herzog
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, Indiana, USA
| | - Randal J Kaufman
- Degenerative Diseases Program, Center for Genetic Diseases and Aging Research, SBP Medical Discovery Institute, California, USA.
| |
Collapse
|
2
|
Wu T, Jiang Y, Shi W, Wang Y, Li T. Endoplasmic reticulum stress: a novel targeted approach to repair bone defects by regulating osteogenesis and angiogenesis. J Transl Med 2023; 21:480. [PMID: 37464413 PMCID: PMC10353205 DOI: 10.1186/s12967-023-04328-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 07/06/2023] [Indexed: 07/20/2023] Open
Abstract
Bone regeneration therapy is clinically important, and targeted regulation of endoplasmic reticulum (ER) stress is important in regenerative medicine. The processing of proteins in the ER controls cell fate. The accumulation of misfolded and unfolded proteins occurs in pathological states, triggering ER stress. ER stress restores homeostasis through three main mechanisms, including protein kinase-R-like ER kinase (PERK), inositol-requiring enzyme 1ɑ (IRE1ɑ) and activating transcription factor 6 (ATF6), collectively known as the unfolded protein response (UPR). However, the UPR has both adaptive and apoptotic effects. Modulation of ER stress has therapeutic potential for numerous diseases. Repair of bone defects involves both angiogenesis and bone regeneration. Here, we review the effects of ER stress on osteogenesis and angiogenesis, with emphasis on ER stress under high glucose (HG) and inflammatory conditions, and the use of ER stress inducers or inhibitors to regulate osteogenesis and angiogenesis. In addition, we highlight the ability for exosomes to regulate ER stress. Recent advances in the regulation of ER stress mediated osteogenesis and angiogenesis suggest novel therapeutic options for bone defects.
Collapse
Affiliation(s)
- Tingyu Wu
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, No. 59, Haier Road, Qingdao, 266003, China
| | - Yaping Jiang
- Department of Oral Implantology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Weipeng Shi
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, No. 59, Haier Road, Qingdao, 266003, China
| | - Yingzhen Wang
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, No. 59, Haier Road, Qingdao, 266003, China
| | - Tao Li
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, No. 59, Haier Road, Qingdao, 266003, China.
| |
Collapse
|
3
|
Enhancement of recombinant human IL-24 (rhIL-24) protein production from site-specific integrated engineered CHO cells by sodium butyrate treatment. Bioprocess Biosyst Eng 2022; 45:1979-1991. [DOI: 10.1007/s00449-022-02801-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 10/07/2022] [Indexed: 11/27/2022]
Abstract
AbstractInterleukin-24 (IL-24) has specific inhibitory effects on the proliferation of various tumor cells with almost no toxicity to normal cells. The antitumor activity of recombinant human IL-24 protein produced in mammalian cells is much higher than that of bacteria, but its expression level is extremely low. Sodium butyrate (NaBu) was utilized as a media additive to increase protein expression in Chinese hamster ovary cells. The site-specific integrated engineered cells FCHO/IL-24 were treated with NaBu under different culture conditions (10% and 0.5% serum adherent culture, 0.5% serum suspension culture). First, 3 days of 1 mmol/L NaBu treatment significantly increased rhIL-24 expression level in FCHO/IL-24 cells by 119.94 ± 1.5% (**p < 0.01), 57.49 ± 2.4% (**p < 0.01), and 20.17 ± 3.03% (*p < 0.05) under the above culture conditions. Second, NaBu has a time- and dose-dependent inhibitory effect on FCHO/IL-24 proliferation and induces G0/G1 phase arrest. Under 10% and 0.5% serum adherent culture, G0/G1 phase cells were increased by 11.3 ± 0.5% (**p < 0.01) and 15.0 ± 2.6% (**p < 0.01), respectively. No induction of apoptosis was observed under a high dosage of NaBu treatment. These results suggest that NaBu increases rhIL-24 secretion via inhibiting cell cycle progression, thereby trapping cells in the highly productive G0/G1 phase. Finally, with increasing NaBu dose, glucose concentration increased (**p < 0.01) while lactic acid and ammonia concentrations reduced significantly (**p < 0.01) in 10% and 0.5% serum adherent culture supernatant. RNA-seq showed that NaBu treatment affected multiple tumor and immune-related pathways. In conclusion, NaBu treatment dramatically promoted rhIL-24 production in engineered FCHO/IL-24 cells by altering downstream pathways and inducing G0/G1 cell arrest with little effect on apoptosis.
Collapse
|
4
|
Abstract
INTRODUCTION Hemophilia A (HA) or B (HB) is an X-linked recessive disorder caused by a defect in the factor VIII (FVIII) or factor IX (FIX) gene which leads to the dysfunction of blood coagulation. Protein replacement therapy (PRT) uses recombinant proteins and plasma-derived products, which incurs high cost and inconvenience requiring routine intravenous infusions and life-time treatment. Understanding of detailed molecular mechanisms on FVIII gene function could provide innovative solutions to amend this disorder. In recent decades, gene therapeutics have advanced rapidly and a one-time cure solution has been proposed. AREAS COVERED This review summarizes current understanding of molecular pathways involved in blood coagulation, with emphasis on FVIII's functional role. The existing knowledge and challenges on FVIII gene expression, from transcription, translation, post-translational modification including glycosylation to protein processing and secretion, and co-factor interactions are deciphered and potential molecular interventions discussed. EXPERT OPINION This article reviews the potential treatment targets for HA and HB, including antibodies, small molecules and gene therapeutics, based on molecular mechanisms of FVIII biosynthesis, and further, assessing the pros and cons of these various treatment strategies. Understanding detailed FVIII protein synthesis and secretory pathways could provide exciting opportunities in identifying novel therapeutics to ameliorate hemophilia state.
Collapse
Affiliation(s)
- Jie Gong
- School of Medicine, University of Electronic Science and Technology of China, Sichuan, China
| | - Hao-Lin Wang
- School of Medicine, University of Electronic Science and Technology of China, Sichuan, China
| | - Lung-Ji Chang
- School of Medicine, University of Electronic Science and Technology of China, Sichuan, China.,Geno-Immune Medical Institute, Shenzhen, China
| |
Collapse
|
5
|
Barzadd MM, Lundqvist M, Harris C, Malm M, Volk AL, Thalén N, Chotteau V, Grassi L, Smith A, Abadi ML, Lambiase G, Gibson S, Hatton D, Rockberg J. Autophagy and intracellular product degradation genes identified by systems biology analysis reduce aggregation of bispecific antibody in CHO cells. N Biotechnol 2022; 68:68-76. [PMID: 35123066 DOI: 10.1016/j.nbt.2022.01.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 01/31/2022] [Accepted: 01/31/2022] [Indexed: 12/18/2022]
Abstract
Aggregation of therapeutic bispecific antibodies negatively affects the yield, shelf-life, efficacy and safety of these products. Pairs of stable Chinese hamster ovary (CHO) cell lines produced two difficult-to-express bispecific antibodies with different levels of aggregated product (10-75% aggregate) in a miniaturized bioreactor system. Here, transcriptome analysis was used to interpret the biological causes for the aggregation and to identify strategies to improve product yield and quality. Differential expression- and gene set analysis revealed upregulated proteasomal degradation, unfolded protein response and autophagy processes to be correlated with reduced protein aggregation. Fourteen candidate genes with the potential to reduce aggregation were co-expressed in the stable clones for validation. Of these, HSP90B1, DDIT3, AK1S1, and ATG16L1, were found to significantly lower aggregation in the stable producers and two (HSP90B1 and DNAJC3) increased titres of the anti-HER2 monoclonal antibody trastuzumab by 50% during transient expression. It is suggested that this approach could be of general use for defining aggregation bottlenecks in CHO cells.
Collapse
Affiliation(s)
- Mona Moradi Barzadd
- KTH - Royal Institute of Technology, School of Engineering Sciences in Chemistry, Biotechnology, and Health, Dept. of Protein Science, SE-106 91 Stockholm, Sweden.
| | - Magnus Lundqvist
- KTH - Royal Institute of Technology, School of Engineering Sciences in Chemistry, Biotechnology, and Health, Dept. of Protein Science, SE-106 91 Stockholm, Sweden.
| | - Claire Harris
- Cell Culture & Fermentation Sciences, BioPharmaceutical Development, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Magdalena Malm
- KTH - Royal Institute of Technology, School of Engineering Sciences in Chemistry, Biotechnology, and Health, Dept. of Protein Science, SE-106 91 Stockholm, Sweden
| | - Anna-Luisa Volk
- KTH - Royal Institute of Technology, School of Engineering Sciences in Chemistry, Biotechnology, and Health, Dept. of Protein Science, SE-106 91 Stockholm, Sweden
| | - Niklas Thalén
- KTH - Royal Institute of Technology, School of Engineering Sciences in Chemistry, Biotechnology, and Health, Dept. of Protein Science, SE-106 91 Stockholm, Sweden
| | - Veronique Chotteau
- KTH - Royal Institute of Technology, School of Engineering Sciences in Chemistry, Biotechnology, and Health, Dept. of Industrial Biotechnology, SE-106 91 Stockholm, Sweden
| | - Luigi Grassi
- Cell Culture & Fermentation Sciences, BioPharmaceutical Development, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Andrew Smith
- Cell Culture & Fermentation Sciences, BioPharmaceutical Development, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Marina Leal Abadi
- Cell Culture & Fermentation Sciences, BioPharmaceutical Development, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Giulia Lambiase
- Analytical Sciences, BioPharmaceutical Development, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK and Advanced Biomanufacturing Centre, Department of Chemical and Biological Engineering, University of Sheffield, Mappin Street, Sheffield, UK
| | - Suzanne Gibson
- Cell Culture & Fermentation Sciences, BioPharmaceutical Development, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Diane Hatton
- Cell Culture & Fermentation Sciences, BioPharmaceutical Development, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Johan Rockberg
- KTH - Royal Institute of Technology, School of Engineering Sciences in Chemistry, Biotechnology, and Health, Dept. of Protein Science, SE-106 91 Stockholm, Sweden.
| |
Collapse
|
6
|
Liu ZB, Zhang T, Ye X, Liu ZQ, Sun X, Zhang LL, Wu CJ. Natural substances derived from herbs or plants are promising sources of anticancer agents against colorectal cancer via triggering apoptosis. J Pharm Pharmacol 2021; 74:162-178. [PMID: 34559879 DOI: 10.1093/jpp/rgab130] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 08/18/2021] [Indexed: 12/21/2022]
Abstract
OBJECTIVES Nowadays, one of the most common gastrointestinal cancers is colorectal cancer (CRC). Chemotherapy is still one of the main methods to treat cancer. However, the currently available synthetic chemotherapy drugs often cause serious adverse reactions. Apoptosis is generally considered as an ideal way for induction the death of tumour cells without the body's inflammatory response, and it is reported that lots of natural agents could trigger various cancer cells to apoptosis. The overarching aim of this project was to elucidate the specific mechanisms by which natural substances induce apoptosis in CRC cells and to be used as an alternative therapeutic option in the future. KEY FINDINGS The mechanisms for the pro-apoptotic effects of natural substances derived from herbs or plants include death receptor pathway, mitochondrial pathway, endoplasmic reticulum stress pathway, related signal transduction pathways (PI3K/Akt, MAPK, p53 signalling), and so on. SUMMARY This paper updated this information regarding the anti-tumour effects of natural agents via induction of apoptosis against CRC, which would be beneficial for future new drug research regarding natural products from herbs or plants.
Collapse
Affiliation(s)
- Zi-Bo Liu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, P.R. China
| | - Ting Zhang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, P.R. China
| | - Xun Ye
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, P.R. China
| | - Zi-Qi Liu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, P.R. China
| | - Xue Sun
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, P.R. China
| | - Li-Lin Zhang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, P.R. China
| | | |
Collapse
|
7
|
Mustapha S, Mohammed M, Azemi AK, Jatau AI, Shehu A, Mustapha L, Aliyu IM, Danraka RN, Amin A, Bala AA, Ahmad WANW, Rasool AHG, Mustafa MR, Mokhtar SS. Current Status of Endoplasmic Reticulum Stress in Type II Diabetes. Molecules 2021; 26:4362. [PMID: 34299638 PMCID: PMC8307902 DOI: 10.3390/molecules26144362] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/10/2021] [Accepted: 07/17/2021] [Indexed: 12/12/2022] Open
Abstract
The endoplasmic reticulum (ER) plays a multifunctional role in lipid biosynthesis, calcium storage, protein folding, and processing. Thus, maintaining ER homeostasis is essential for cellular functions. Several pathophysiological conditions and pharmacological agents are known to disrupt ER homeostasis, thereby, causing ER stress. The cells react to ER stress by initiating an adaptive signaling process called the unfolded protein response (UPR). However, the ER initiates death signaling pathways when ER stress persists. ER stress is linked to several diseases, such as cancer, obesity, and diabetes. Thus, its regulation can provide possible therapeutic targets for these. Current evidence suggests that chronic hyperglycemia and hyperlipidemia linked to type II diabetes disrupt ER homeostasis, thereby, resulting in irreversible UPR activation and cell death. Despite progress in understanding the pathophysiology of the UPR and ER stress, to date, the mechanisms of ER stress in relation to type II diabetes remain unclear. This review provides up-to-date information regarding the UPR, ER stress mechanisms, insulin dysfunction, oxidative stress, and the therapeutic potential of targeting specific ER stress pathways.
Collapse
Affiliation(s)
- Sagir Mustapha
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu 16150, Kelantan, Malaysia; (S.M.); (A.K.A.); (A.H.G.R.)
- Department of Pharmacology and Therapeutics, Ahmadu Bello University, Zaria 810107, Kaduna, Nigeria; (A.S.); (I.M.A.); (R.N.D.)
| | - Mustapha Mohammed
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Penang 11800, Pulau Pinang, Malaysia;
- Department of Clinical Pharmacy and Pharmacy Practice, Ahmadu Bello University, Zaria 810107, Kaduna, Nigeria
| | - Ahmad Khusairi Azemi
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu 16150, Kelantan, Malaysia; (S.M.); (A.K.A.); (A.H.G.R.)
| | - Abubakar Ibrahim Jatau
- School of Pharmacy and Pharmacology, University of Tasmania, Hobart, TAS 7005, Australia;
| | - Aishatu Shehu
- Department of Pharmacology and Therapeutics, Ahmadu Bello University, Zaria 810107, Kaduna, Nigeria; (A.S.); (I.M.A.); (R.N.D.)
| | - Lukman Mustapha
- Department of Pharmaceutical and Medicinal Chemistry, Kaduna State University, Kaduna 800241, Kaduna, Nigeria;
| | - Ibrahim Muazzamu Aliyu
- Department of Pharmacology and Therapeutics, Ahmadu Bello University, Zaria 810107, Kaduna, Nigeria; (A.S.); (I.M.A.); (R.N.D.)
| | - Rabi’u Nuhu Danraka
- Department of Pharmacology and Therapeutics, Ahmadu Bello University, Zaria 810107, Kaduna, Nigeria; (A.S.); (I.M.A.); (R.N.D.)
| | - Abdulbasit Amin
- Department of Physiology, Faculty of Basic Medical Sciences, University of Ilorin, Ilorin 240103, Kwara, Nigeria;
- Membrane Traffic Group, Instituto Gulbenkian de Ciencia, 2784-156 Lisbon, Portugal
| | - Auwal Adam Bala
- Department of Pharmacology, College of Medicine and Health Sciences, Federal University Dutse, Dutse 720281, Jigawa, Nigeria;
- Department of Pharmacology and Therapeutics, Faculty of Pharmaceutical Sciences, Bayero University Kano, Kano 700241, Kano, Nigeria
| | - Wan Amir Nizam Wan Ahmad
- Biomedicine Programme, School of Health Sciences, Universiti Sains Malaysia, Kota Bharu 16150, Kelantan, Malaysia;
| | - Aida Hanum Ghulam Rasool
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu 16150, Kelantan, Malaysia; (S.M.); (A.K.A.); (A.H.G.R.)
| | - Mohd Rais Mustafa
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia;
| | - Siti Safiah Mokhtar
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu 16150, Kelantan, Malaysia; (S.M.); (A.K.A.); (A.H.G.R.)
| |
Collapse
|
8
|
Alterations of the Platelet Proteome in Lung Cancer: Accelerated F13A1 and ER Processing as New Actors in Hypercoagulability. Cancers (Basel) 2021; 13:cancers13092260. [PMID: 34066760 PMCID: PMC8125802 DOI: 10.3390/cancers13092260] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 05/05/2021] [Indexed: 12/21/2022] Open
Abstract
Simple Summary The risk of venous thromboembolism in cancer is nine times higher than in the general population and the second leading cause of death in these patients. Tissue factor and downstream plasmatic coagulation cascade are largely responsible for the risk of thrombosis in cancer. In recent years, it has been increasingly recognised that platelets also play a central role in tumour growth and cancer-associated thrombosis. The underlying molecular mechanisms are largely unknown. In order to comprehensively investigate the biochemical changes in platelets from cancers with high risk of thrombosis, we examined the platelet proteome of brain and lung cancer patients in comparison to sex and age-matched healthy controls. However, we only found alterations in lung cancer, where some of these platelet proteins directly promote thrombosis. One example is the increased amount of the enzyme protein disulfide isomerase, which is clinically investigated as an antithrombotic drug target of the plant-based flavonol quercetin. Abstract In order to comprehensively expose cancer-related biochemical changes, we compared the platelet proteome of two types of cancer with a high risk of thrombosis (22 patients with brain cancer, 19 with lung cancer) to 41 matched healthy controls using unbiased two-dimensional differential in-gel electrophoresis. The examined platelet proteome was unchanged in patients with brain cancer, but considerably affected in lung cancer with 15 significantly altered proteins. Amongst these, the endoplasmic reticulum (ER) proteins calreticulin (CALR), endoplasmic reticulum chaperone BiP (HSPA5) and protein disulfide-isomerase (P4HB) were significantly elevated. Accelerated conversion of the fibrin stabilising factor XIII was detected in platelets of patients with lung cancer by elevated levels of a coagulation factor XIII (F13A1) 55 kDa fragment. A significant correlation of this F13A1 cleavage product with plasma levels of the plasmin–α-2-antiplasmin complex and D-dimer suggests its enhanced degradation by the fibrinolytic system. Protein association network analysis showed that lung cancer-related proteins were involved in platelet degranulation and upregulated ER protein processing. As a possible outcome, plasma FVIII, an immediate end product for ER-mediated glycosylation, correlated significantly with the ER-executing chaperones CALR and HSPA5. These new data on the differential behaviour of platelets in various cancers revealed F13A1 and ER chaperones as potential novel diagnostic and therapeutic targets in lung cancer patients.
Collapse
|
9
|
Alassaf M, Halloran MC. Pregnancy-associated plasma protein-aa regulates endoplasmic reticulum-mitochondria associations. eLife 2021; 10:59687. [PMID: 33759764 PMCID: PMC8024009 DOI: 10.7554/elife.59687] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 03/23/2021] [Indexed: 11/13/2022] Open
Abstract
Endoplasmic reticulum (ER) and mitochondria form close physical associations to facilitate calcium transfer, thereby regulating mitochondrial function. Neurons with high metabolic demands, such as sensory hair cells, are especially dependent on precisely regulated ER-mitochondria associations. We previously showed that the secreted metalloprotease pregnancy-associated plasma protein-aa (Pappaa) regulates mitochondrial function in zebrafish lateral line hair cells (Alassaf et al., 2019). Here, we show that pappaa mutant hair cells exhibit excessive and abnormally close ER-mitochondria associations, suggesting increased ER-mitochondria calcium transfer. pappaa mutant hair cells are more vulnerable to pharmacological induction of ER-calcium transfer. Additionally, pappaa mutant hair cells display ER stress and dysfunctional downstream processes of the ER-mitochondria axis including altered mitochondrial morphology and reduced autophagy. We further show that Pappaa influences ER-calcium transfer and autophagy via its ability to stimulate insulin-like growth factor-1 bioavailability. Together our results identify Pappaa as a novel regulator of the ER-mitochondria axis.
Collapse
Affiliation(s)
- Mroj Alassaf
- Department of Integrative Biology, University of Wisconsin, Madison, United States.,Department of Neuroscience, University of Wisconsin, Madison, United States.,Neuroscience Training Program, University of Wisconsin, Madison, United States
| | - Mary C Halloran
- Department of Integrative Biology, University of Wisconsin, Madison, United States.,Department of Neuroscience, University of Wisconsin, Madison, United States.,Neuroscience Training Program, University of Wisconsin, Madison, United States
| |
Collapse
|
10
|
Cao W, Dong B, Horling F, Firrman JA, Lengler J, Klugmann M, de la Rosa M, Wu W, Wang Q, Wei H, Moore AR, Roberts SA, Booth CJ, Hoellriegl W, Li D, Konkle B, Miao C, Reipert BM, Scheiflinger F, Rottensteiner H, Xiao W. Minimal Essential Human Factor VIII Alterations Enhance Secretion and Gene Therapy Efficiency. Mol Ther Methods Clin Dev 2020; 19:486-495. [PMID: 33313336 PMCID: PMC7708868 DOI: 10.1016/j.omtm.2020.10.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 10/19/2020] [Indexed: 12/18/2022]
Abstract
One important limitation for achieving therapeutic expression of human factor VIII (FVIII) in hemophilia A gene therapy is inefficient secretion of the FVIII protein. Substitution of five amino acids in the A1 domain of human FVIII with the corresponding porcine FVIII residues generated a secretion-enhanced human FVIII variant termed B-domain-deleted (BDD)-FVIII-X5 that resulted in 8-fold higher FVIII activity levels in the supernatant of an in vitro cell-based assay system than seen with unmodified human BDD-FVIII. Analysis of purified recombinant BDD-FVIII-X5 and BDD-FVIII revealed similar specific activities for both proteins, indicating that the effect of the X5 alteration is confined to increased FVIII secretion. Intravenous delivery in FVIII-deficient mice of liver-targeted adeno-associated virus (AAV) vectors designed to express BDD-FVIII-X5 or BDD-FVIII achieved substantially higher plasma FVIII activity levels for BDD-FVIII-X5, even when highly efficient codon-optimized F8 nucleotide sequences were employed. A comprehensive immunogenicity assessment using in vitro stimulation assays and various in vivo preclinical models of hemophilia A demonstrated that the BDD-FVIII-X5 variant does not exhibit an increased immunogenicity risk compared to BDD-FVIII. In conclusion, BDD-FVIII-X5 is an effective FVIII variant molecule that can be further developed for use in gene- and protein-based therapeutics for patients with hemophilia A.
Collapse
Affiliation(s)
- Wenjing Cao
- Sol Sherry Thrombosis Research Center, Temple University Medical School, 3400 North Broad Street, Philadelphia, PA, 19140, USA
| | - Biao Dong
- Sol Sherry Thrombosis Research Center, Temple University Medical School, 3400 North Broad Street, Philadelphia, PA, 19140, USA
| | - Franziska Horling
- Drug Discovery Austria, Baxalta Innovations GmbH (now part of Takeda), Donau-City Str. 7, Vienna 1220, Austria
| | - Jenni A. Firrman
- Dairy and Functional Foods Research Unit, ARS, USDA, 600 East Mermaid Lane, Wyndmoor, PA 19038, USA
| | - Johannes Lengler
- Drug Discovery Austria, Baxalta Innovations GmbH (now part of Takeda), Donau-City Str. 7, Vienna 1220, Austria
| | - Matthias Klugmann
- Drug Discovery Austria, Baxalta Innovations GmbH (now part of Takeda), Donau-City Str. 7, Vienna 1220, Austria
| | - Maurus de la Rosa
- Drug Discovery Austria, Baxalta Innovations GmbH (now part of Takeda), Donau-City Str. 7, Vienna 1220, Austria
| | - Wenman Wu
- Sol Sherry Thrombosis Research Center, Temple University Medical School, 3400 North Broad Street, Philadelphia, PA, 19140, USA
| | - Qizhao Wang
- Sol Sherry Thrombosis Research Center, Temple University Medical School, 3400 North Broad Street, Philadelphia, PA, 19140, USA
| | - Hongying Wei
- Sol Sherry Thrombosis Research Center, Temple University Medical School, 3400 North Broad Street, Philadelphia, PA, 19140, USA
| | - Andrea R. Moore
- Sol Sherry Thrombosis Research Center, Temple University Medical School, 3400 North Broad Street, Philadelphia, PA, 19140, USA
| | - Sean A. Roberts
- Sol Sherry Thrombosis Research Center, Temple University Medical School, 3400 North Broad Street, Philadelphia, PA, 19140, USA
| | - Carmen J. Booth
- Department of Comparative Medicine, Yale University School of Medicine, 310 Cedar St., BML 330, New Haven, CT 06510, USA
| | - Werner Hoellriegl
- Drug Discovery Austria, Baxalta Innovations GmbH (now part of Takeda), Donau-City Str. 7, Vienna 1220, Austria
| | - Dong Li
- Sol Sherry Thrombosis Research Center, Temple University Medical School, 3400 North Broad Street, Philadelphia, PA, 19140, USA
| | - Barbara Konkle
- Seattle Children’s Research Institute, University of Washington, 1900 9 Ave, Seattle, WA 98195, USA
| | - Carol Miao
- Department of Medicine/Hematology, University of Washington, 1900 9 Ave, Seattle, WA 98195, USA
| | - Birgit M. Reipert
- Drug Discovery Austria, Baxalta Innovations GmbH (now part of Takeda), Donau-City Str. 7, Vienna 1220, Austria
| | - Friedrich Scheiflinger
- Drug Discovery Austria, Baxalta Innovations GmbH (now part of Takeda), Donau-City Str. 7, Vienna 1220, Austria
| | - Hanspeter Rottensteiner
- Drug Discovery Austria, Baxalta Innovations GmbH (now part of Takeda), Donau-City Str. 7, Vienna 1220, Austria
| | - Weidong Xiao
- Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, IN 46202, USA
| |
Collapse
|
11
|
Tao Y, Qiu T, Yao X, Jiang L, Wang N, Jiang J, Jia X, Wei S, Zhang J, Zhu Y, Tian W, Yang G, Liu X, Liu S, Ding Y, Sun X. IRE1α/NOX4 signaling pathway mediates ROS-dependent activation of hepatic stellate cells in NaAsO 2 -induced liver fibrosis. J Cell Physiol 2020; 236:1469-1480. [PMID: 32776539 DOI: 10.1002/jcp.29952] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 07/07/2020] [Accepted: 07/09/2020] [Indexed: 12/12/2022]
Abstract
Liver fibrosis is a severe health problem worldwide, and it is characterized by the activation of hepatic stellate cells (HSCs) and excessive deposition of collagen. Prolonged arsenic exposure can induce HSCs activation and liver fibrosis. In the present study, the results showed that chronic NaAsO2 ingestion could result in liver fibrosis and oxidative stress in Sprague-Dawley rats, along with representative collagen deposition and HSCs activation. In addition, the inositol-requiring enzyme 1α (IRE1α)-endoplasmic reticulum (ER)-stress pathway was activated, and the activity of nicotinamide adenine dinucleotide phosphate oxidase 4 (NOX4) was upregulated in rat livers. Simultaneously, the excessive production of reactive oxygen species (ROS) could induce HSCs activation, and NOX4 played an important role in generating ROS in vitro. Moreover, ER stress occurred with HSCs activation at the same time under NaAsO2 exposure, and during ER stress, the IRE1α pathway was responsible for NOX4 activation. Therefore, inhibition of IRE1α activation could attenuate the HSCs activation induced by NaAsO2 . In conclusion, the present study manifested that inorganic arsenic exposure could activate HSCs through IRE1α/NOX4-mediated ROS generation.
Collapse
Affiliation(s)
- Ye Tao
- Department of Occupational and Environmental Health, Dalian Medical University, Dalian, China
| | - Tianming Qiu
- Department of Occupational and Environmental Health, Dalian Medical University, Dalian, China
| | - Xiaofeng Yao
- Department of Occupational and Environmental Health, Dalian Medical University, Dalian, China
| | - Liping Jiang
- Experimental Teaching Center of Public Health, Dalian Medical University, Dalian, China
| | - Ningning Wang
- Department of Nutrition and Food Hygiene, Dalian Medical University, Dalian, China
| | - Jintong Jiang
- School of Foreign Languages, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Xue Jia
- Department of Occupational and Environmental Health, Dalian Medical University, Dalian, China
| | - Sen Wei
- Department of Occupational and Environmental Health, Dalian Medical University, Dalian, China
| | - Jingyuan Zhang
- Department of Occupational and Environmental Health, Dalian Medical University, Dalian, China
| | - Yuhan Zhu
- Department of Occupational and Environmental Health, Dalian Medical University, Dalian, China
| | - Wenyue Tian
- Department of Infectious Diseases, Shengjing Hospital of China Medical University, Shenyang, China
| | - Guang Yang
- Department of Nutrition and Food Hygiene, Dalian Medical University, Dalian, China
| | - Xiaofang Liu
- Department of Nutrition and Food Hygiene, Dalian Medical University, Dalian, China
| | - Shuang Liu
- Department of Occupational and Environmental Health, Dalian Medical University, Dalian, China
| | - Yang Ding
- Department of Infectious Diseases, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiance Sun
- Department of Occupational and Environmental Health, Dalian Medical University, Dalian, China.,Global Health Research Center, Dalian Medical University, Dalian, China
| |
Collapse
|
12
|
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: 1231] [Impact Index Per Article: 307.8] [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
|
13
|
Poothong J, Pottekat A, Siirin M, Campos AR, Paton AW, Paton JC, Lagunas-Acosta J, Chen Z, Swift M, Volkmann N, Hanein D, Yong J, Kaufman RJ. Factor VIII exhibits chaperone-dependent and glucose-regulated reversible amyloid formation in the endoplasmic reticulum. Blood 2020; 135:1899-1911. [PMID: 32128578 PMCID: PMC7243144 DOI: 10.1182/blood.2019002867] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 02/13/2020] [Indexed: 12/14/2022] Open
Abstract
Hemophilia A, an X-linked bleeding disorder caused by deficiency of factor VIII (FVIII), is treated by protein replacement. Unfortunately, this regimen is costly due to the expense of producing recombinant FVIII as a consequence of its low-level secretion from mammalian host cells. FVIII expression activates the endoplasmic reticulum (ER) stress response, causes oxidative stress, and induces apoptosis. Importantly, little is known about the factors that cause protein misfolding and aggregation in metazoans. Here, we identified intrinsic and extrinsic factors that cause FVIII to form aggregates. We show that FVIII forms amyloid-like fibrils within the ER lumen upon increased FVIII synthesis or inhibition of glucose metabolism. Significantly, FVIII amyloids can be dissolved upon restoration of glucose metabolism to produce functional secreted FVIII. Two ER chaperone families and their cochaperones, immunoglobulin binding protein (BiP) and calnexin/calreticulin, promote FVIII solubility in the ER, where the former is also required for disaggregation. A short aggregation motif in the FVIII A1 domain (termed Aggron) is necessary and sufficient to seed β-sheet polymerization, and BiP binding to this Aggron prevents amyloidogenesis. Our findings provide novel insight into mechanisms that limit FVIII secretion and ER protein aggregation in general and have implication for ongoing hemophilia A gene-therapy clinical trials.
Collapse
Affiliation(s)
| | | | | | - Alexandre Rosa Campos
- Proteomics Core Facility, Sanford Burnham Prebys (SBP) Medical Discovery Institute, La Jolla, CA
| | - Adrienne W Paton
- Research Centre for Infectious Diseases, Department of Molecular and Biomedical Science, University of Adelaide, Adelaide, SA, Australia; and
| | - James C Paton
- Research Centre for Infectious Diseases, Department of Molecular and Biomedical Science, University of Adelaide, Adelaide, SA, Australia; and
| | | | | | - Mark Swift
- Immunity and Pathogenesis Program, SBP Medical Discovery Institute, La Jolla, CA
| | - Niels Volkmann
- Immunity and Pathogenesis Program, SBP Medical Discovery Institute, La Jolla, CA
| | - Dorit Hanein
- Immunity and Pathogenesis Program, SBP Medical Discovery Institute, La Jolla, CA
| | | | | |
Collapse
|
14
|
Patel SR, Lundgren TS, Spencer HT, Doering CB. The Immune Response to the fVIII Gene Therapy in Preclinical Models. Front Immunol 2020; 11:494. [PMID: 32351497 PMCID: PMC7174743 DOI: 10.3389/fimmu.2020.00494] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 03/04/2020] [Indexed: 12/14/2022] Open
Abstract
Neutralizing antibodies to factor VIII (fVIII), referred to as "inhibitors," remain the most challenging complication post-fVIII replacement therapy. Preclinical development of novel fVIII products involves studies incorporating hemophilia A (HA) and wild-type animal models. Though immunogenicity is a critical aspect of preclinical pharmacology studies, gene therapy studies tend to focus on fVIII expression levels without major consideration for immunogenicity. Therefore, little clarity exists on whether preclinical testing can be predictive of clinical immunogenicity risk. Despite this, but perhaps due to the potential for transformative benefits, clinical gene therapy trials have progressed rapidly. In more than two decades, no inhibitors have been observed. However, all trials are conducted in previously treated patients without a history of inhibitors. The current review thus focuses on our understanding of preclinical immunogenicity for HA gene therapy candidates and the potential indication for inhibitor treatment, with a focus on product- and platform-specific determinants, including fVIII transgene sequence composition and tissue/vector biodistribution. Currently, the two leading clinical gene therapy vectors are adeno-associated viral (AAV) and lentiviral (LV) vectors. For HA applications, AAV vectors are liver-tropic and employ synthetic, high-expressing, liver-specific promoters. Factors including vector serotype and biodistribution, transcriptional regulatory elements, transgene sequence, dosing, liver immunoprivilege, and host immune status may contribute to tipping the scale between immunogenicity and tolerance. Many of these factors can also be important in delivery of LV-fVIII gene therapy, especially when delivered intravenously for liver-directed fVIII expression. However, ex vivo LV-fVIII targeting and transplantation of hematopoietic stem and progenitor cells (HSPC) has been demonstrated to achieve durable and curative fVIII production without inhibitor development in preclinical models. A critical variable appears to be pre-transplantation conditioning regimens that suppress and/or ablate T cells. Additionally, we and others have demonstrated the potential of LV-fVIII HSPC and liver-directed AAV-fVIII gene therapy to eradicate pre-existing inhibitors in murine and canine models of HA, respectively. Future preclinical studies will be essential to elucidate immune mechanism(s) at play in the context of gene therapy for HA, as well as strategies for preventing adverse immune responses and promoting immune tolerance even in the setting of pre-existing inhibitors.
Collapse
Affiliation(s)
- Seema R. Patel
- Hemostasis and Thrombosis Program, Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Emory University, Atlanta, GA, United States
| | - Taran S. Lundgren
- Cell and Gene Therapy Program, Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Emory University, Atlanta, GA, United States
- Graduate Program in Molecular and Systems Pharmacology, Laney Graduate School, Emory University, Atlanta, GA, United States
| | - H. Trent Spencer
- Cell and Gene Therapy Program, Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Emory University, Atlanta, GA, United States
| | - Christopher B. Doering
- Cell and Gene Therapy Program, Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Emory University, Atlanta, GA, United States
| |
Collapse
|
15
|
Chen CY, Tran DM, Cavedon A, Cai X, Rajendran R, Lyle MJ, Martini PGV, Miao CH. Treatment of Hemophilia A Using Factor VIII Messenger RNA Lipid Nanoparticles. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 20:534-544. [PMID: 32330871 PMCID: PMC7178004 DOI: 10.1016/j.omtn.2020.03.015] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 03/23/2020] [Accepted: 03/30/2020] [Indexed: 12/17/2022]
Abstract
Hemophilia A (HemA) patients are currently treated with costly and inconvenient replacement therapy of short-lived factor VIII (FVIII) protein. Development of lipid nanoparticle (LNP)-encapsulated mRNA encoding FVIII can change this paradigm. LNP technology constitutes a biocompatible and scalable system to efficiently package and deliver mRNA to the target site. Mice intravenously infused with the luciferase mRNA LNPs showed luminescence signals predominantly in the liver 4 h after injection. Repeated injections of LNPs did not induce elevation of liver transaminases. We next injected LNPs carrying mRNAs encoding different variants of human FVIII (F8 LNPs) into HemA mice. A single injection of B domain-deleted F8 LNPs using different dosing regimens achieved a wide range of therapeutic activities rapidly, which can be beneficial for various usages in hemophilia treatment. The expression slowly declined yet remained above therapeutic levels up to 5–7 days post-injection. Furthermore, routine repeated injections of F8 LNPs in immunodeficient mice produced consistent expression of FVIII over time. In conclusion, F8 LNP treatment produced rapid and prolonged duration of FVIII expression that could be applied to prophylactic treatment and potentially various other treatment options. Our study showed potential for a safe and effective platform of new mRNA therapies for HemA.
Collapse
Affiliation(s)
- Chun-Yu Chen
- Seattle Children's Research Institute, Seattle, WA, USA
| | | | | | - Xiaohe Cai
- Seattle Children's Research Institute, Seattle, WA, USA
| | | | - Meghan J Lyle
- Seattle Children's Research Institute, Seattle, WA, USA
| | | | - Carol H Miao
- Seattle Children's Research Institute, Seattle, WA, USA; Department of Pediatrics, University of Washington, Seattle, WA, USA.
| |
Collapse
|
16
|
Ballar Kirmizibayrak P, Erbaykent-Tepedelen B, Gozen O, Erzurumlu Y. Divergent Modulation of Proteostasis in Prostate Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1233:117-151. [PMID: 32274755 DOI: 10.1007/978-3-030-38266-7_5] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Proteostasis regulates key cellular processes such as cell proliferation, differentiation, transcription, and apoptosis. The mechanisms by which proteostasis is regulated are crucial and the deterioration of cellular proteostasis has been significantly associated with tumorigenesis since it specifically targets key oncoproteins and tumor suppressors. Prostate cancer (PCa) is the second most common cause of cancer death in men worldwide. Androgens mediate one of the most central signaling pathways in all stages of PCa via the androgen receptor (AR). In addition to their regulation by hormones, PCa cells are also known to be highly secretory and are particularly prone to ER stress as proper ER function is essential. Alterations in various complex signaling pathways and cellular processes including cell cycle control, transcription, DNA repair, apoptosis, cell adhesion, epithelial-mesenchymal transition (EMT), and angiogenesis are critical factors influencing PCa development through key molecular changes mainly by posttranslational modifications in PCa-related proteins, including AR, NKX3.1, PTEN, p53, cyclin D1, and p27. Several ubiquitin ligases like MDM2, Siah2, RNF6, CHIP, and substrate-binding adaptor SPOP; deubiquitinases such as USP7, USP10, USP26, and USP12 are just some of the modifiers involved in the regulation of these key proteins via ubiquitin-proteasome system (UPS). Some ubiquitin-like modifiers, especially SUMOs, have been also closely associated with PCa. On the other hand, the proteotoxicity resulting from misfolded proteins and failure of ER adaptive capacity induce unfolded protein response (UPR) that is an indispensable signaling mechanism for PCa development. Lastly, ER-associated degradation (ERAD) also plays a crucial role in prostate tumorigenesis. In this section, the relationship between prostate cancer and proteostasis will be discussed in terms of UPS, UPR, SUMOylation, ERAD, and autophagy.
Collapse
Affiliation(s)
| | | | - Oguz Gozen
- Faculty of Medicine, Department of Physiology, Ege University, Izmir, Turkey
| | - Yalcin Erzurumlu
- Faculty of Pharmacy, Department of Biochemistry, Suleyman Demirel University, Isparta, Turkey
| |
Collapse
|
17
|
Yong J, Bischof H, Burgstaller S, Siirin M, Murphy A, Malli R, Kaufman RJ. Mitochondria supply ATP to the ER through a mechanism antagonized by cytosolic Ca 2. eLife 2019; 8:49682. [PMID: 31498082 PMCID: PMC6763289 DOI: 10.7554/elife.49682] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 09/09/2019] [Indexed: 12/15/2022] Open
Abstract
The endoplasmic reticulum (ER) imports ATP and uses energy from ATP hydrolysis for protein folding and trafficking. However, little is known about how this vital ATP transport occurs across the ER membrane. Here, using three commonly used cell lines (CHO, INS1 and HeLa), we report that ATP enters the ER lumen through a cytosolic Ca2+-antagonized mechanism, or CaATiER (Ca2+-Antagonized Transport into ER). Significantly, we show that mitochondria supply ATP to the ER and a SERCA-dependent Ca2+ gradient across the ER membrane is necessary for ATP transport into the ER, through SLC35B1/AXER. We propose that under physiological conditions, increases in cytosolic Ca2+ inhibit ATP import into the ER lumen to limit ER ATP consumption. Furthermore, the ATP level in the ER is readily depleted by oxidative phosphorylation (OxPhos) inhibitors and that ER protein misfolding increases ATP uptake from mitochondria into the ER. These findings suggest that ATP usage in the ER may increase mitochondrial OxPhos while decreasing glycolysis, i.e. an ‘anti-Warburg’ effect.
Collapse
Affiliation(s)
- Jing Yong
- Degenerative Diseases Program, SBP Medical Discovery Institute, La Jolla, United States
| | - Helmut Bischof
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Sandra Burgstaller
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Marina Siirin
- Degenerative Diseases Program, SBP Medical Discovery Institute, La Jolla, United States
| | - Anne Murphy
- Department of Pharmacology, University of California, San Diego, La Jolla, United States
| | - Roland Malli
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Randal J Kaufman
- Degenerative Diseases Program, SBP Medical Discovery Institute, La Jolla, United States.,Department of Pharmacology, University of California, San Diego, La Jolla, United States
| |
Collapse
|
18
|
Yasuno K, Igura S, Yamaguchi Y, Imaoka M, Kai K, Mori K. Pathological examination of spontaneous vacuolation of pancreatic acinar cells in mice. J Toxicol Pathol 2019; 32:105-109. [PMID: 31092977 PMCID: PMC6511541 DOI: 10.1293/tox.2018-0065] [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] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 01/15/2019] [Indexed: 12/18/2022] Open
Abstract
Pancreatic acinar cell vacuolation is spontaneously observed in mice; however, the lesion is rare and has not been well documented. Herein, we present a detailed pathological examination of this lesion. Vacuoles in pancreatic acinar cells were present in 2/15 X gene knockout mice with a C57BL/6J mouse background, 4/298 ICR(CD-1) mice, 1/110 B6C3F1 mice, and 3/399 CByB6F1-Tg(HRAS)2Jic mice. The vacuoles were usually observed in a unit of the acinus, and the lesions were spread throughout the pancreas. These vacuoles contained weakly basophilic material that was positive for the periodic acid-Schiff reaction. Immunohistochemically, the vacuoles were positive for calreticulin antibody. Electron microscopy revealed globular dilatation of the rough endoplasmic reticulum (rER). According to these findings, vacuolation of pancreatic acinar cells is caused by the accumulation of misfolded proteins and enlargement of the rER.
Collapse
Affiliation(s)
- Kyohei Yasuno
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., 1-16-13 Kita-Kasai, Edogawa-ku, Tokyo 134-8630, Japan
| | - Saori Igura
- Pathology Department, BoZo Research Center Inc, Gotemba-shi, Shizuoka 412-0039, Japan
| | - Yuko Yamaguchi
- Pathology Department, BoZo Research Center Inc, Gotemba-shi, Shizuoka 412-0039, Japan
| | - Masako Imaoka
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., 1-16-13 Kita-Kasai, Edogawa-ku, Tokyo 134-8630, Japan
| | - Kiyonori Kai
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., 1-16-13 Kita-Kasai, Edogawa-ku, Tokyo 134-8630, Japan
| | - Kazuhiko Mori
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., 1-16-13 Kita-Kasai, Edogawa-ku, Tokyo 134-8630, Japan
| |
Collapse
|
19
|
Adulcikas J, Sonda S, Norouzi S, Sohal SS, Myers S. Targeting the Zinc Transporter ZIP7 in the Treatment of Insulin Resistance and Type 2 Diabetes. Nutrients 2019; 11:nu11020408. [PMID: 30781350 PMCID: PMC6412268 DOI: 10.3390/nu11020408] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 01/13/2019] [Accepted: 02/12/2019] [Indexed: 02/07/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a disease associated with dysfunctional metabolic processes that lead to abnormally high levels of blood glucose. Preceding the development of T2DM is insulin resistance (IR), a disorder associated with suppressed or delayed responses to insulin. The effects of this response are predominately mediated through aberrant cell signalling processes and compromised glucose uptake into peripheral tissue including adipose, liver and skeletal muscle. Moreover, a major factor considered to be the cause of IR is endoplasmic reticulum (ER) stress. This subcellular organelle plays a pivotal role in protein folding and processes that increase ER stress, leads to maladaptive responses that result in cell death. Recently, zinc and the proteins that transport this metal ion have been implicated in the ER stress response. Specifically, the ER-specific zinc transporter ZIP7, coined the "gate-keeper" of zinc release from the ER into the cytosol, was shown to be essential for maintaining ER homeostasis in intestinal epithelium and myeloid leukaemia cells. Moreover, ZIP7 controls essential cell signalling pathways similar to insulin and activates glucose uptake in skeletal muscle. Accordingly, ZIP7 may be essential for the control of ER localized zinc and mechanisms that disrupt this process may lead to ER-stress and contribute to IR. Accordingly, understanding the mechanisms of ZIP7 action in the context of IR may provide opportunities to develop novel therapeutic options to target this transporter in the treatment of IR and subsequent T2DM.
Collapse
Affiliation(s)
- John Adulcikas
- College of Health and Medicine, School of Health Sciences, University of Tasmania, TAS 7005, Australia.
| | - Sabrina Sonda
- College of Health and Medicine, School of Health Sciences, University of Tasmania, TAS 7005, Australia.
| | - Shaghayegh Norouzi
- College of Health and Medicine, School of Health Sciences, University of Tasmania, TAS 7005, Australia.
| | - Sukhwinder Singh Sohal
- College of Health and Medicine, School of Health Sciences, University of Tasmania, TAS 7005, Australia.
| | - Stephen Myers
- College of Health and Medicine, School of Health Sciences, University of Tasmania, TAS 7005, Australia.
| |
Collapse
|
20
|
Koubek EJ, Weissenrieder JS, Neighbors JD, Hohl RJ. Schweinfurthins: Lipid Modulators with Promising Anticancer Activity. Lipids 2018; 53:767-784. [DOI: 10.1002/lipd.12088] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 08/30/2018] [Accepted: 08/31/2018] [Indexed: 12/12/2022]
Affiliation(s)
- Emily J. Koubek
- Departments of Medicine and Pharmacology, The Pennsylvania State Cancer Institute; The Pennsylvania State College of Medicine, 500 University Drive Hershey; Hershey PA 17033 USA
| | - Jillian S. Weissenrieder
- Departments of Medicine and Pharmacology; The Pennsylvania State College of Medicine, 500 University Drive Hershey; Hershey PA 17033 USA
| | - Jeffrey D. Neighbors
- Departments of Pharmacology and Medicine; The Pennsylvania State College of Medicine, 500 University Drive Hershey; Hershey PA 17033 USA
| | - Raymond J. Hohl
- Departments of Medicine and Pharmacology, The Pennsylvania State Cancer Institute; The Pennsylvania State College of Medicine, 500 University Drive Hershey; Hershey PA 17033 USA
| |
Collapse
|
21
|
Kaushik P, Henry M, Clynes M, Meleady P. The Expression Pattern of the Phosphoproteome Is Significantly Changed During the Growth Phases of Recombinant CHO Cell Culture. Biotechnol J 2018; 13:e1700221. [DOI: 10.1002/biot.201700221] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 07/13/2018] [Indexed: 12/31/2022]
Affiliation(s)
- Prashant Kaushik
- National Institute for Cellular Biotechnology; Dublin City University; Dublin 9 Ireland
| | - Michael Henry
- National Institute for Cellular Biotechnology; Dublin City University; Dublin 9 Ireland
| | - Martin Clynes
- National Institute for Cellular Biotechnology; Dublin City University; Dublin 9 Ireland
| | - Paula Meleady
- National Institute for Cellular Biotechnology; Dublin City University; Dublin 9 Ireland
| |
Collapse
|
22
|
Ciavattini A, Delli Carpini G, Serri M, Tozzi A, Leoni F, Di Loreto E, Saccucci F. Unfolded protein response, a link between endometrioid ovarian carcinoma and endometriosis: A pilot study. Oncol Lett 2018; 16:5449-5454. [PMID: 30250617 DOI: 10.3892/ol.2018.9256] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 06/28/2018] [Indexed: 12/25/2022] Open
Abstract
The aim of the present study was to analyze the expression profile of unfolded protein response (UPR) genes in endometrioid ovarian carcinoma and to evaluate its possible involvement in the neoplastic progression of endometriosis. An experimental retrospective pilot study was conducted on women with a diagnosis of endometrioid ovarian carcinoma at FIGO stage IA, ovarian endometriotic cysts or healthy subjects without a previous diagnosis of endometriosis. The expression profiles of UPR genes (ATF6, GRP78, CHOP and XBP1) were compared among ovaries with endometrioid ovarian cancer, endometriotic ovarian cysts, healthy contralateral ovaries and eutopic and healthy endometrial tissues. A significantly higher expression of ATF6 and GRP78 was detected in the affected ovaries in comparison with the healthy contralateral ovaries, while CHOP and XBP1 exhibited a significantly lower expression. XBP1 was overexpressed in endometrial tissues and its expression gradually decreased in endometriosis cysts and endometrioid ovarian carcinoma. These results support the hypothesis that alterations in the UPR genes CHOP and XBP1 are involved in the neoplastic progression of endometrioid ovarian cancer and are acquired following ovarian localization of ectopic endometrial cells.
Collapse
Affiliation(s)
- Andrea Ciavattini
- Woman's Health Sciences Department, Gynecologic Section, Polytechnic University of Marche, Ancona I-60123, Italy
| | - Giovanni Delli Carpini
- Woman's Health Sciences Department, Gynecologic Section, Polytechnic University of Marche, Ancona I-60123, Italy
| | - Matteo Serri
- Woman's Health Sciences Department, Gynecologic Section, Polytechnic University of Marche, Ancona I-60123, Italy
| | - Alessandra Tozzi
- Woman's Health Sciences Department, Gynecologic Section, Polytechnic University of Marche, Ancona I-60123, Italy
| | - Francesca Leoni
- Department of Biochemistry, Biology and Genetics, Polytechnic University of Marche, Ancona I-60121, Italy
| | - Eugenia Di Loreto
- Woman's Health Sciences Department, Gynecologic Section, Polytechnic University of Marche, Ancona I-60123, Italy
| | - Franca Saccucci
- Department of Biochemistry, Biology and Genetics, Polytechnic University of Marche, Ancona I-60121, Italy
| |
Collapse
|
23
|
Sharma SH, Rajamanickam V, Nagarajan S. Antiproliferative effect of p-Coumaric acid targets UPR activation by downregulating Grp78 in colon cancer. Chem Biol Interact 2018; 291:16-28. [PMID: 29879413 DOI: 10.1016/j.cbi.2018.06.001] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 05/08/2018] [Accepted: 06/03/2018] [Indexed: 02/07/2023]
Abstract
p-CA is a naturally occurring phenolic acid present in most plants and in all commonly consumed vegetables and fruits. Here we demonstrated the anti-cancer effect of the food borne phytochemical p-CA both in vitro and in vivo models of colon cancer using growth rate and tumor incidence as endpoints. Glucose regulated protein (GRP78) induction and UPR activation plays a key role in oncogenic progression, therefore increased dependence of cancer cells on these UPR signaling pathways for survival can be exploited for anti-cancer research. Hence we investigated the effect of p-CA on Grp78 a molecular chaperone often upregulated in colon cancer and its impact on unfolded protein response (UPR). Administration of the procarcinogen 1,2- dimethylhydrazine (DMH) causes Grp78 upregulation and tumor adaptation via UPR activation. The adaptive activity of UPR activates antiapoptotic NF-κB that results in upregulation of the markers of inflammation and angiogenesis. Supplementation of p-CA downregulated Grp78 and activated UPR mediated apoptosis both in in vitro and in vivo models of colon cancer. Further we observed that p-CA significantly reduced inflammation by decreasing the expression of cytokines COX-2, IL-6, TNF-α and PGE2 as analyzed by q-PCR and also reduced the expression of p-p65 and p-IκBα as analyzed by western blot. Further mechanistic insights revealed that p-CA inhibits Grp78 upregulation in cancer cells through activation of PERK-eIF2α-ATF-4-CHOP pathway that culminates in apoptosis inducing effect of p-CA.
Collapse
Affiliation(s)
- Sharada H Sharma
- School of Chemical and Biotechnology, SASTRA Deemed University, Thirumalaisamudram, Thanjavur, 613401, Tamil Nadu, India
| | - Vinothkumar Rajamanickam
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, PR China
| | - Sangeetha Nagarajan
- School of Chemical and Biotechnology, SASTRA Deemed University, Thirumalaisamudram, Thanjavur, 613401, Tamil Nadu, India.
| |
Collapse
|
24
|
Chen Y, Chen L, Zhang H, Huang S, Xiong Y, Xia C. Interaction of Sulfonylureas with Liver Uptake Transporters OATP1B1 and OATP1B3. Basic Clin Pharmacol Toxicol 2018; 123:147-154. [PMID: 29498478 DOI: 10.1111/bcpt.12992] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 02/13/2018] [Indexed: 12/20/2022]
Abstract
Sulfonylureas (SUs) such as glibenclamide, gliclazide, glimepiride, glipizide and gliquidone are one of the first oral medicines available for the treatment of type 2 diabetes and are widely used for the treatment of hyperglycaemia. The hepatic transporters, organic anion transporting polypeptide 1B1 (OATP1B1) and organic anion transporting polypeptide 1B3 (OATP1B3), play an important role in the disposition of a variety of drugs by mediating their uptake from blood into hepatocytes. Drug-drug interactions mediated by OATP1B1/1B3 may result in the hepatic transporting change for drug substrates. The inhibitory effects of glibenclamide and glimepiride on sulfobromophthalein (BSP) uptake have been previously studied, and glibenclamide has been reported as the substrate of OATP1B3, but it remains unclear whether other SUs such as gliclazide, glipizide and gliquidone are substrates of OATP1B1 and OATP1B3. Here, we investigated the relationship between the five most commonly applied SUs (glibenclamide, gliclazide, glimepiride, glipizide, gliquidone) and OATP1B1 and OATP1B3. We performed uptake and inhibition assays in HEK293T cells stably expressing OATP1B1 or OATP1B3, respectively, and established a liquid chromatography-mass spectrometry (LC-MS) method for the simultaneous measurement of five SUs. We demonstrated that gliclazide and glimepiride are substrates of OATP1B1 and glibenclamide and glipizide are substrates of OATP1B3. We also confirmed the interaction between these SUs and rosuvastatin. No transporting was observed for gliquidone, suggesting that it is not a substrate of either transporter.
Collapse
Affiliation(s)
- Yu Chen
- Clinical Pharmacology Institute, Nanchang University, Nanchang, China.,Jiangxi Provincial Children's Hospital, Nanchang, China
| | - Lin Chen
- Clinical Pharmacology Institute, Nanchang University, Nanchang, China
| | - Hong Zhang
- Clinical Pharmacology Institute, Nanchang University, Nanchang, China
| | - Shibo Huang
- Clinical Pharmacology Institute, Nanchang University, Nanchang, China
| | - Yuqing Xiong
- Clinical Pharmacology Institute, Nanchang University, Nanchang, China
| | - Chunhua Xia
- Clinical Pharmacology Institute, Nanchang University, Nanchang, China
| |
Collapse
|
25
|
Yin B, Wang Q, Chung CY, Ren X, Bhattacharya R, Yarema KJ, Betenbaugh MJ. Butyrated ManNAc analog improves protein expression in Chinese hamster ovary cells. Biotechnol Bioeng 2018; 115:1531-1541. [DOI: 10.1002/bit.26560] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 01/29/2018] [Accepted: 02/04/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Bojiao Yin
- Department of Chemical and Biomolecular Engineering; Johns Hopkins University; Baltimore Maryland
| | - Qiong Wang
- Department of Chemical and Biomolecular Engineering; Johns Hopkins University; Baltimore Maryland
| | - Cheng-Yu Chung
- Department of Chemical and Biomolecular Engineering; Johns Hopkins University; Baltimore Maryland
| | - Xiaozhi Ren
- Department of Chemical and Biomolecular Engineering; Johns Hopkins University; Baltimore Maryland
| | - Rahul Bhattacharya
- Department of Biomedical Engineering; Johns Hopkins University; Baltimore Maryland
| | - Kevin J. Yarema
- Department of Biomedical Engineering; Johns Hopkins University; Baltimore Maryland
| | - Michael J. Betenbaugh
- Department of Chemical and Biomolecular Engineering; Johns Hopkins University; Baltimore Maryland
| |
Collapse
|
26
|
Hussain H, Fisher DI, Abbott WM, Roth RG, Dickson AJ. Use of a protein engineering strategy to overcome limitations in the production of “Difficult to Express” recombinant proteins. Biotechnol Bioeng 2017. [DOI: 10.1002/bit.26358] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Hirra Hussain
- Manchester Institute of Biotechnology; Faculty of Science and Engineering; University of Manchester; M1 7DN Manchester United Kingdom
| | - David I. Fisher
- AstraZeneca, Cambridge Science Park; Milton Cambridge United Kingdom
| | - W. Mark Abbott
- AstraZeneca, Cambridge Science Park; Milton Cambridge United Kingdom
| | | | - Alan J. Dickson
- Manchester Institute of Biotechnology; Faculty of Science and Engineering; University of Manchester; M1 7DN Manchester United Kingdom
| |
Collapse
|
27
|
Zitzmann J, Weidner T, Czermak P. Optimized expression of the antimicrobial protein Gloverin from Galleria mellonella using stably transformed Drosophila melanogaster S2 cells. Cytotechnology 2017; 69:371-389. [PMID: 28132128 PMCID: PMC5366974 DOI: 10.1007/s10616-017-0068-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 01/10/2017] [Indexed: 12/11/2022] Open
Abstract
Antimicrobial proteins and peptides (AMPs) are valuable as leads in the pharmaceutical industry for the development of novel anti-infective drugs. Here we describe the efficient heterologous expression and basic characterization of a Gloverin-family AMP derived from the greater wax moth Galleria mellonella. Highly productive single-cell clones prepared by limiting dilution achieved a 100% increase in productivity compared to the original polyclonal Drosophila melanogaster S2 cell line. Comprehensive screening for suitable expression conditions using statistical experimental designs revealed that optimal induction was achieved using 600 µM CuSO4 at the mid-exponential growth phase. Under these conditions, 25 mg/L of the AMP was expressed at the 1-L bioreactor scale, with optimal induction and harvest times ensured by dielectric spectroscopy and the online measurement of optical density. Gloverin was purified from the supernatant by immobilized metal ion affinity chromatography followed by dialysis. In growth assays, the purified protein showed specific antimicrobial activity against two different strains of Escherichia coli.
Collapse
Affiliation(s)
- Jan Zitzmann
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Giessen, Germany
| | - Tobias Weidner
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Giessen, Germany
| | - Peter Czermak
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Giessen, Germany.
- Department of Chemical Engineering, Kansas State University, Manhattan, KS, USA.
- Faculty of Biology and Chemistry, Justus-Liebig University of Giessen, Giessen, Germany.
- Project Group Bioresources, Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), Giessen, Germany.
| |
Collapse
|
28
|
Lund AM, Kaas CS, Brandl J, Pedersen LE, Kildegaard HF, Kristensen C, Andersen MR. Network reconstruction of the mouse secretory pathway applied on CHO cell transcriptome data. BMC SYSTEMS BIOLOGY 2017; 11:37. [PMID: 28298216 PMCID: PMC5353859 DOI: 10.1186/s12918-017-0414-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 02/27/2017] [Indexed: 11/21/2022]
Abstract
Background Protein secretion is one of the most important processes in eukaryotes. It is based on a highly complex machinery involving numerous proteins in several cellular compartments. The elucidation of the cell biology of the secretory machinery is of great importance, as it drives protein expression for biopharmaceutical industry, a 140 billion USD global market. However, the complexity of secretory process is difficult to describe using a simple reductionist approach, and therefore a promising avenue is to employ the tools of systems biology. Results On the basis of manual curation of the literature on the yeast, human, and mouse secretory pathway, we have compiled a comprehensive catalogue of characterized proteins with functional annotation and their interconnectivity. Thus we have established the most elaborate reconstruction (RECON) of the functional secretion pathway network to date, counting 801 different components in mouse. By employing our mouse RECON to the CHO-K1 genome in a comparative genomic approach, we could reconstruct the protein secretory pathway of CHO cells counting 764 CHO components. This RECON furthermore facilitated the development of three alternative methods to study protein secretion through graphical visualizations of omics data. We have demonstrated the use of these methods to identify potential new and known targets for engineering improved growth and IgG production, as well as the general observation that CHO cells seem to have less strict transcriptional regulation of protein secretion than healthy mouse cells. Conclusions The RECON of the secretory pathway represents a strong tool for interpretation of data related to protein secretion as illustrated with transcriptomic data of Chinese Hamster Ovary (CHO) cells, the main platform for mammalian protein production. Electronic supplementary material The online version of this article (doi:10.1186/s12918-017-0414-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anne Mathilde Lund
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads 223, DK-2800, Kgs. Lyngby, Denmark
| | - Christian Schrøder Kaas
- Recombinant Expression Technologies, Global Research Unit, Novo Nordisk A/S, Novo Nordisk Park, DK-2760, Måløv, Denmark
| | - Julian Brandl
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads 223, DK-2800, Kgs. Lyngby, Denmark
| | - Lasse Ebdrup Pedersen
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kemitorvet 220, DK-2800, Kgs. Lyngby, Denmark
| | - Helene Faustrup Kildegaard
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kemitorvet 220, DK-2800, Kgs. Lyngby, Denmark
| | - Claus Kristensen
- Faculty of Health and Medical Sciences, Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3B, DK-2200, København N, Denmark
| | - Mikael Rørdam Andersen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads 223, DK-2800, Kgs. Lyngby, Denmark.
| |
Collapse
|
29
|
Valeric acid supplementation combined to mild hypothermia increases productivity in CHO cell cultivations. Biochem Eng J 2016. [DOI: 10.1016/j.bej.2016.06.031] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
30
|
Fantacini DMC, Fontes AM, de Abreu Neto MS, Covas DT, Picanço-Castro V. The F309S mutation increases factor VIII secretion in human cell line. Rev Bras Hematol Hemoter 2016; 38:135-40. [PMID: 27208572 PMCID: PMC4877930 DOI: 10.1016/j.bjhh.2016.04.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 04/01/2016] [Indexed: 11/25/2022] Open
Abstract
OBJECTIVES The capacity of a human cell line to secrete recombinant factor VIII with a F309S point mutation was investigated, as was the effect of the addition of chemical chaperones (betaine and sodium-4-phenylbutyrate) on the secretion of factor VIII. METHODS This work used a vector with a F309S mutation in the A1 domain to investigate FVIII production in the HEK 293 human cell line. Factor VIII activity was measured by chromogenic assay. Furthermore, the effects of chemical drugs on the culture were evaluated. RESULTS The addition of the F309S mutation to a previously described FVIII variant increased FVIII secretion by 4.5 fold. Moreover, the addition of betaine or sodium-4-phenylbutyrate increased the secretion rate of FVIIIΔB proteins in HEK 293 cells, but the same effect was not seen for FVIIIΔB-F309S indicating that all the recombinant protein produced had been efficiently secreted. CONCLUSION Bioengineering factor VIII expressed in human cells may lead to an efficient production of recombinant factor VIII and contribute toward low-cost coagulation factor replacement therapy for hemophilia A. FVIII-F309S produced in human cells can be effective in vivo.
Collapse
Affiliation(s)
| | | | | | - Dimas Tadeu Covas
- Universidade de São Paulo (USP), Ribeirão Preto, SP, Brazil; Hemocentro de Ribeirão Preto, Ribeirão Preto, SP, Brazil
| | - Virgínia Picanço-Castro
- Universidade de São Paulo (USP), Ribeirão Preto, SP, Brazil; Hemocentro de Ribeirão Preto, Ribeirão Preto, SP, Brazil
| |
Collapse
|
31
|
Aroso M, Ferreira R, Freitas A, Vitorino R, Gomez-Lazaro M. New insights on the mitochondrial proteome plasticity in Parkinson's disease. Proteomics Clin Appl 2016; 10:416-29. [PMID: 26749507 DOI: 10.1002/prca.201500092] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 12/09/2015] [Accepted: 01/04/2016] [Indexed: 12/21/2022]
Abstract
Parkinson's disease (PD) is one of the most common neurodegenerative diseases whose relentless progression results in severe disability. Although PD aetiology is unknown, growing evidences point to the mitochondrial involvement in the pathobiology of this disorder. So, it seems imperative to understand the means by which the molecular pathways harboured in this organelle are regulated. With the advances in MS-based proteomics, there is a substantial expectation in the increased knowledge of mitochondrial protein dynamics. Still, few studies have been performed on mitochondrial protein profiling in the context of PD. In order to integrate data from these studies, network analyses were performed taking into consideration variables such as model of PD, cell line, or tissue origin. Overall, data retrieved from these analyses highlighted the modulation of the biological processes related with "generation of energy," "cellular metabolism," and "mitochondrial transport" in PD. However, it was noted that the impact of sample type and/or PD model on the biological processes was modulated by the disease. Moreover, technical considerations related to protein characterization using gel-based or gel-free MS approaches should be considered in data comparison among different studies. Data from the present review will help to envisage future studies targeting these mechanisms.
Collapse
Affiliation(s)
- Miguel Aroso
- Department of Medical Sciences, iBiMED, University of Aveiro, Aveiro, Portugal
| | - Rita Ferreira
- Department of Chemistry, QOPNA, Mass Spectrometry Center, University of Aveiro, Aveiro, Portugal
| | - Ana Freitas
- Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal.,INEB - Instituto de Engenharia Biomédica, University of Porto, Porto, Portugal.,Faculty of Medicine, University of Porto, Porto, Portugal
| | - Rui Vitorino
- Department of Medical Sciences, iBiMED, University of Aveiro, Aveiro, Portugal.,Department of Chemistry, QOPNA, Mass Spectrometry Center, University of Aveiro, Aveiro, Portugal.,Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal.,Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Maria Gomez-Lazaro
- Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal.,INEB - Instituto de Engenharia Biomédica, University of Porto, Porto, Portugal
| |
Collapse
|
32
|
Testosterone production by a Leydig tumor cell line is suppressed by hyperthermia-induced endoplasmic reticulum stress in mice. Life Sci 2016; 146:184-91. [DOI: 10.1016/j.lfs.2015.12.042] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 12/21/2015] [Accepted: 12/22/2015] [Indexed: 11/21/2022]
|
33
|
Versatile microscale screening platform for improving recombinant protein productivity in Chinese hamster ovary cells. Sci Rep 2015; 5:18016. [PMID: 26657798 PMCID: PMC4676018 DOI: 10.1038/srep18016] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 11/10/2015] [Indexed: 11/09/2022] Open
Abstract
Chinese hamster ovary (CHO) cells are widely used as cell factories for the production of biopharmaceuticals. In contrast to the highly optimized production processes for monoclonal antibody (mAb)-based biopharmaceuticals, improving productivity of non-mAb therapeutic glycoproteins is more likely to reduce production costs significantly. The aim of this study was to establish a versatile target gene screening platform for improving productivity for primarily non-mAb glycoproteins with complete interchangeability of model proteins and target genes using transient expression. The platform consists of four techniques compatible with 96-well microplates: lipid-based transient transfection, cell cultivation in microplates, cell counting and antibody-independent product titer determination based on split-GFP complementation. We were able to demonstrate growth profiles and volumetric productivity of CHO cells in 96-half-deepwell microplates comparable with those obtained in shake flasks. In addition, we demonstrate that split-GFP complementation can be used to accurately measure relative titers of therapeutic glycoproteins. Using this platform, we were able to detect target gene-specific increase in titer and specific productivity of two non-mAb glycoproteins. In conclusion, the platform provides a novel miniaturized and parallelisable solution for screening target genes and holds the potential to unravel genes that can enhance the secretory capacity of CHO cells.
Collapse
|
34
|
Hassler JR, Scheuner DL, Wang S, Han J, Kodali VK, Li P, Nguyen J, George JS, Davis C, Wu SP, Bai Y, Sartor M, Cavalcoli J, Malhi H, Baudouin G, Zhang Y, Yates III JR, Itkin-Ansari P, Volkmann N, Kaufman RJ. The IRE1α/XBP1s Pathway Is Essential for the Glucose Response and Protection of β Cells. PLoS Biol 2015; 13:e1002277. [PMID: 26469762 PMCID: PMC4607427 DOI: 10.1371/journal.pbio.1002277] [Citation(s) in RCA: 122] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 09/08/2015] [Indexed: 12/11/2022] Open
Abstract
Although glucose uniquely stimulates proinsulin biosynthesis in β cells, surprisingly little is known of the underlying mechanism(s). Here, we demonstrate that glucose activates the unfolded protein response transducer inositol-requiring enzyme 1 alpha (IRE1α) to initiate X-box-binding protein 1 (Xbp1) mRNA splicing in adult primary β cells. Using mRNA sequencing (mRNA-Seq), we show that unconventional Xbp1 mRNA splicing is required to increase and decrease the expression of several hundred mRNAs encoding functions that expand the protein secretory capacity for increased insulin production and protect from oxidative damage, respectively. At 2 wk after tamoxifen-mediated Ire1α deletion, mice develop hyperglycemia and hypoinsulinemia, due to defective β cell function that was exacerbated upon feeding and glucose stimulation. Although previous reports suggest IRE1α degrades insulin mRNAs, Ire1α deletion did not alter insulin mRNA expression either in the presence or absence of glucose stimulation. Instead, β cell failure upon Ire1α deletion was primarily due to reduced proinsulin mRNA translation primarily because of defective glucose-stimulated induction of a dozen genes required for the signal recognition particle (SRP), SRP receptors, the translocon, the signal peptidase complex, and over 100 other genes with many other intracellular functions. In contrast, Ire1α deletion in β cells increased the expression of over 300 mRNAs encoding functions that cause inflammation and oxidative stress, yet only a few of these accumulated during high glucose. Antioxidant treatment significantly reduced glucose intolerance and markers of inflammation and oxidative stress in mice with β cell-specific Ire1α deletion. The results demonstrate that glucose activates IRE1α-mediated Xbp1 splicing to expand the secretory capacity of the β cell for increased proinsulin synthesis and to limit oxidative stress that leads to β cell failure.
Collapse
Affiliation(s)
- Justin R. Hassler
- Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, United States of America
- Department of Biological Chemistry, University of Michigan Medical Center, Ann Arbor, Michigan, United States of America
| | - Donalyn L. Scheuner
- Department of Biological Chemistry, University of Michigan Medical Center, Ann Arbor, Michigan, United States of America
- Lilly Research Laboratories, Eli Lilly & Company, Lilly Corporate Center, Indianapolis, Indiana, United States of America
| | - Shiyu Wang
- Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, United States of America
| | - Jaeseok Han
- Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, United States of America
| | - Vamsi K. Kodali
- Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, United States of America
| | - Philip Li
- Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, United States of America
| | - Julie Nguyen
- Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, United States of America
| | - Jenny S. George
- Department of Biological Chemistry, University of Michigan Medical Center, Ann Arbor, Michigan, United States of America
| | - Cory Davis
- Department of Biological Chemistry, University of Michigan Medical Center, Ann Arbor, Michigan, United States of America
| | - Shengyang P. Wu
- Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, Michigan, United States of America
| | - Yongsheng Bai
- NCIBI Department of Bioinformatics, University of Michigan Medical Center, Ann Arbor, Michigan, United States of America
- Department of Biology, Indiana State University, Terre Haute, Indiana, United States of America
| | - Maureen Sartor
- NCIBI Department of Bioinformatics, University of Michigan Medical Center, Ann Arbor, Michigan, United States of America
| | - James Cavalcoli
- NCIBI Department of Bioinformatics, University of Michigan Medical Center, Ann Arbor, Michigan, United States of America
| | - Harmeet Malhi
- Department of Biological Chemistry, University of Michigan Medical Center, Ann Arbor, Michigan, United States of America
| | - Gregory Baudouin
- Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, United States of America
| | - Yaoyang Zhang
- Department of Chemical Physiology and Cell Biology, The Scripps Research Institute, La Jolla, California, United States of America
| | - John R. Yates III
- Department of Chemical Physiology and Cell Biology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Pamela Itkin-Ansari
- Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, United States of America
| | - Niels Volkmann
- Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, United States of America
| | - Randal J. Kaufman
- Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, United States of America
- Department of Biological Chemistry, University of Michigan Medical Center, Ann Arbor, Michigan, United States of America
- Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, Michigan, United States of America
- * E-mail:
| |
Collapse
|
35
|
Abstract
Glioblastomas are devastating central nervous system tumors with abysmal prognoses. These tumors are often difficult to resect surgically, are highly invasive and proliferative, and are resistant to virtually all therapeutic attempts, making them universally lethal diseases. One key enabling feature of their tumor biology is the engagement of the unfolded protein response (UPR), a stress response originating in the endoplasmic reticulum (ER) designed to handle the pathologies of aggregating malfolded proteins in that organelle. Glioblastomas and other tumors have co-opted this stress response to allow their continued uncontrolled growth by enhanced protein production (maintained by chaperone-assisted protein folding) and lipid biosynthesis driven downstream of the UPR. These features can account for the extensive extracellular remodeling/invasiveness/angiogenesis and proliferative capacity, and ultimately result in tumor phenotypes of chemo- and radio-resistance. The UPR in general, and its chaperoning capacity in particular, are thus putative high-value targets for treatment intervention. Such therapeutic strategies, and potential problems with them, will be discussed and analyzed.
Collapse
|
36
|
Espinosa-Diez C, Miguel V, Mennerich D, Kietzmann T, Sánchez-Pérez P, Cadenas S, Lamas S. Antioxidant responses and cellular adjustments to oxidative stress. Redox Biol 2015; 6:183-197. [PMID: 26233704 PMCID: PMC4534574 DOI: 10.1016/j.redox.2015.07.008] [Citation(s) in RCA: 718] [Impact Index Per Article: 79.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 07/06/2015] [Indexed: 02/08/2023] Open
Abstract
Redox biological reactions are now accepted to bear the Janus faceted feature of promoting both physiological signaling responses and pathophysiological cues. Endogenous antioxidant molecules participate in both scenarios. This review focuses on the role of crucial cellular nucleophiles, such as glutathione, and their capacity to interact with oxidants and to establish networks with other critical enzymes such as peroxiredoxins. We discuss the importance of the Nrf2-Keap1 pathway as an example of a transcriptional antioxidant response and we summarize transcriptional routes related to redox activation. As examples of pathophysiological cellular and tissular settings where antioxidant responses are major players we highlight endoplasmic reticulum stress and ischemia reperfusion. Topologically confined redox-mediated post-translational modifications of thiols are considered important molecular mechanisms mediating many antioxidant responses, whereas redox-sensitive microRNAs have emerged as key players in the posttranscriptional regulation of redox-mediated gene expression. Understanding such mechanisms may provide the basis for antioxidant-based therapeutic interventions in redox-related diseases. Antioxidant responses are crucial for both redox signaling and redox damage. Glutathione-mediated reactions and Nrf2-Keap1 pathway are key antioxidant responses. Redox-related post-translational modifications activate specific signaling pathways. Redox-sensitive microRNAs contribute to redox-mediated gene expression regulation. ER stress and ischemia-reperfusion are antioxidant-related pathophysiological events.
Collapse
Affiliation(s)
- Cristina Espinosa-Diez
- Department of Cell Biology and Immunology, Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Nicolás Cabrera 1, 28049 Madrid, Spain
| | - Verónica Miguel
- Department of Cell Biology and Immunology, Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Nicolás Cabrera 1, 28049 Madrid, Spain
| | - Daniela Mennerich
- Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, Aapistie 7, University of Oulu, FI-90230 Oulu, Finland
| | - Thomas Kietzmann
- Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, Aapistie 7, University of Oulu, FI-90230 Oulu, Finland
| | - Patricia Sánchez-Pérez
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM) and Departamento de Biología Molecular, Universidad Autónoma de Madrid, Nicolás Cabrera 1, 28049 Madrid, Spain; Instituto de Investigación Sanitaria Princesa (IIS-IP), 28006 Madrid, Spain
| | - Susana Cadenas
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM) and Departamento de Biología Molecular, Universidad Autónoma de Madrid, Nicolás Cabrera 1, 28049 Madrid, Spain; Instituto de Investigación Sanitaria Princesa (IIS-IP), 28006 Madrid, Spain
| | - Santiago Lamas
- Department of Cell Biology and Immunology, Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Nicolás Cabrera 1, 28049 Madrid, Spain.
| |
Collapse
|
37
|
Sheehy RM, Kuder CH, Bachman Z, Hohl RJ. Calcium and P-glycoprotein independent synergism between schweinfurthins and verapamil. Cancer Biol Ther 2015; 16:1259-68. [PMID: 26046259 DOI: 10.1080/15384047.2015.1056420] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Schweinfurthins are intriguing natural products with anti-cancer activities and as yet incompletely understood mechanisms of action. We investigated whether inhibitors of P-glycoprotein (Pgp), in a manner analogous to other natural products, might enhance schweinfurthins' growth inhibitory actions by increasing intracellular schweinfurthin levels. Both the schweinfurthin-sensitive glioblastoma multiforme cell line SF-295 and relatively insensitive lung carcinoma cell line A549 were treated with 2 schweinfurthin analogs: 3-deoxyschweinfurthin B-p-nitro bis-stilbene (3dSB-PNBS) and 5'-methylschweinfurthin G (methyl-G). There was a synergistic enhancement of growth inhibition with the combination of the Pgp inhibitor verapamil and both analogs in SF-295 cells. Methyl-G, verapamil, and the combination did not result in alterations to intracellular calcium concentration. Verapamil increased the intracellular concentration of 3dSB-PNBS in both SF-295 and A549 cells in a Pgp-independent manner. Methyl-G, verapamil, and the combination do not result in increased ER stress. Methyl-G increased the intracellular concentration of a known Pgp substrate, Rhodamine 123 in SF-295 cells. Reduction of cellular cholesterol leads to the accumulation of Pgp substrates, as Pgp requires cholesterol for proper function. Since 3dSB enhances lovastatin-induced upregulation of the cholesterol efflux pump ABCA1, it is intriguing that co-treatment with cholesterol rescued the methyl-G-induced increase in Rhodamine 123 intracellular concentration. These studies support the hypothesis that verapamil potentiates the schweinfurthin growth inhibitory effect by increasing its intracellular concentration.
Collapse
Key Words
- 3dSB, 3-deoxyschweinfurthin B
- 3dSB-PNBS, 3-deoxyschweinfurthin B p-nitro bis-stilbene
- BAPTA-AM, 1,2-bis(o-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid
- CI, combination index
- DMP-PNBS, 3,4-dimethoxypheny-p-nitro bis-stilbene
- ER, endoplasmic reticulum
- GBM, Glioblastoma Multiforme
- MTT, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide
- Methyl-G, 5'-methoxyschweinfurthin G
- NCI, National Cancer Institute
- PARP, poly-ADP-ribose polymerase
- Pgp, P-glycoprotein drug efflux pump
- cholesterol metabolism
- drug efflux pump
- glioblastoma multiforme
- oxysterol binding protein
- p-glycoprotein
- schweinfurthin
- verapamil
Collapse
Affiliation(s)
- Ryan M Sheehy
- a Department of Pharmacology ; University of Iowa ; Iowa City , IA USA
| | | | | | | |
Collapse
|
38
|
Wang M, Kaufman RJ. The impact of the endoplasmic reticulum protein-folding environment on cancer development. Nat Rev Cancer 2014; 14:581-97. [PMID: 25145482 DOI: 10.1038/nrc3800] [Citation(s) in RCA: 776] [Impact Index Per Article: 77.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The endoplasmic reticulum (ER) is an essential organelle in eukaryotic cells for the storage and regulated release of calcium and as the entrance to the secretory pathway. Protein misfolding in the ER causes accumulation of misfolded proteins (ER stress) and activation of the unfolded protein response (UPR), which has evolved to maintain a productive ER protein-folding environment. Both ER stress and UPR activation are documented in many different human cancers. In this Review, we summarize the impact of ER stress and UPR activation on every aspect of cancer and discuss outstanding questions for which answers will pave the way for therapeutics.
Collapse
Affiliation(s)
- Miao Wang
- Degenerative Diseases Program, Center for Cancer Research, Sanford-Burnham Medical Research Institute, 10901 N. Torrey Pines Rd, La Jolla, California 92037, USA
| | - Randal J Kaufman
- Degenerative Diseases Program, Center for Cancer Research, Sanford-Burnham Medical Research Institute, 10901 N. Torrey Pines Rd, La Jolla, California 92037, USA
| |
Collapse
|
39
|
O'Brien PD, Hinder LM, Sakowski SA, Feldman EL. ER stress in diabetic peripheral neuropathy: A new therapeutic target. Antioxid Redox Signal 2014; 21:621-33. [PMID: 24382087 DOI: 10.1089/ars.2013.5807] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
SIGNIFICANCE Diabetes and other diseases that comprise the metabolic syndrome have reached epidemic proportions. Diabetic peripheral neuropathy (DPN) is the most prevalent complication of diabetes, affecting ~50% of diabetic patients. Characterized by chronic pain or loss of sensation, recurrent foot ulcerations, and risk for amputation, DPN is associated with significant morbidity and mortality. Mechanisms underlying DPN pathogenesis are complex and not well understood, and no effective treatments are available. Thus, an improved understanding of DPN pathogenesis is critical for the development of successful therapeutic options. RECENT ADVANCES Recent research implicates endoplasmic reticulum (ER) stress as a novel mechanism in the onset and progression of DPN. ER stress activates the unfolded protein response (UPR), a well-orchestrated signaling cascade responsible for relieving stress and restoring normal ER function. CRITICAL ISSUES During times of extreme or chronic stress, such as that associated with diabetes, the UPR may be insufficient to alleviate ER stress, resulting in apoptosis. Here, we discuss the potential role of ER stress in DPN, as well as evidence demonstrating how ER stress intersects with pathways involved in DPN development and progression. An improved understanding of how ER stress contributes to peripheral nerve dysfunction in diabetes will provide important insight into DPN pathogenesis. FUTURE DIRECTIONS Future studies aimed at gaining the necessary insight into ER stress in DPN pathogenesis will ultimately facilitate the development of novel therapies.
Collapse
|
40
|
Hemopexin-dependent heme uptake via endocytosis regulates the Bach1 transcription repressor and heme oxygenase gene activation. Biochim Biophys Acta Gen Subj 2014; 1840:2351-60. [DOI: 10.1016/j.bbagen.2014.02.029] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Revised: 02/24/2014] [Accepted: 02/27/2014] [Indexed: 12/30/2022]
|
41
|
Beukes N, Levendal RA, Frost CL. Selected terpenoids from medicinal plants modulate endoplasmic reticulum stress in metabolic disorders. J Pharm Pharmacol 2014; 66:1505-25. [DOI: 10.1111/jphp.12267] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Accepted: 03/16/2014] [Indexed: 12/20/2022]
Abstract
Abstract
Objectives
The majority of research performed on cellular stress and apoptosis focuses on mitochondrial dysfunction; however, the importance of the endoplasmic reticulum dysfunction and the link to metabolic diseases has gained a substantial interest. This review focuses on the potential of terpenoids to influence endoplasmic reticulum stress and the possible role terpenoids play as the treatment of metabolic diseases.
Key findings
Metabolic diseases develop as a result of a cascade of cellular pathways. In most cases, cells are able to compensate for the disruption of the cellular homeostasis although the initiation of response pathways; however, chronic stress initiates apoptotic pathways. This reviewed (1) showed the importance of phytoterpenoids to influence endoplasmic reticulum (ER) stress and homeostasis, (2) showed how regulating ER stress affect the cell survival and death, and (3) highlighted some examples of how the progression of metabolic diseases can be influenced by ER.
Summary
Due to the substantial number of terpenoids that have been identified in literature, this review gave examples of 21 terpenoids that have been documented to have an effect on the different proteins associated with ER stress, how these plant terpenoids influence ER dysfunction and metabolic diseases such as diabetes, cancer, liver, and neurological diseases and parasitic infections.
Collapse
Affiliation(s)
- Natasha Beukes
- Department of Biochemistry and Microbiology, Nelson Mandela Metropolitan University, Port Elizabeth, South Africa
| | - Ruby-Ann Levendal
- Department of Biochemistry and Microbiology, Nelson Mandela Metropolitan University, Port Elizabeth, South Africa
| | - Carminita L Frost
- Department of Biochemistry and Microbiology, Nelson Mandela Metropolitan University, Port Elizabeth, South Africa
| |
Collapse
|
42
|
Hussain H, Maldonado-Agurto R, Dickson AJ. The endoplasmic reticulum and unfolded protein response in the control of mammalian recombinant protein production. Biotechnol Lett 2014; 36:1581-93. [PMID: 24752815 DOI: 10.1007/s10529-014-1537-y] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Accepted: 04/10/2014] [Indexed: 12/31/2022]
Abstract
The endoplasmic reticulum (ER) of eukaryotic cells is involved in the synthesis and processing of proteins and lipids in the secretory pathway. These processing events that proteins undergo in the ER may present major limiting steps for recombinant protein production. Increased protein synthesis, accumulation of improperly processed or mis-folded protein can induce ER stress. To cope with ER stress, the ER has quality control mechanisms, such as the unfolded protein response (UPR) and ER-associated degradation to restore homeostasis. ER stress and UPR activation trigger multiple physiological cellular changes. Here we review cellular mechanisms that cope with ER stress and illustrate how this knowledge can be applied to increase the efficiency of recombinant protein expression.
Collapse
Affiliation(s)
- Hirra Hussain
- Faculty of Life Sciences, The Michael Smith Building, University of Manchester, Oxford Road, Manchester, M13 9PT, UK
| | | | | |
Collapse
|
43
|
Wang Y, Li JR, Sun MX, Ni B, Huan C, Huang L, Li C, Fan HJ, Ren XF, Mao X. Triggering unfolded protein response by 2-Deoxy-D-glucose inhibits porcine epidemic diarrhea virus propagation. Antiviral Res 2014; 106:33-41. [PMID: 24681123 PMCID: PMC7113873 DOI: 10.1016/j.antiviral.2014.03.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Revised: 03/05/2014] [Accepted: 03/16/2014] [Indexed: 12/21/2022]
Abstract
The unfolded protein response (UPR) is cyto-protective machinery elicited towards an influx of large amount of protein synthesis in the endoplasmic reticulum (ER). Extensive studies suggest that the UPR can also be activated during virus infection. In the present studies, we first evaluated if porcine epidemic diarrhea virus (PEDV) infection activated the UPR pathways. Electron microscopy analysis demonstrated the morphology changes of ER post-PEDV infection. Western blot and real-time PCR identified the differences of UPR genes in response to PEDV infection. The results suggested that PEDV infection induced UPR in Vero cells. Meanwhile, we silenced the expression of PKR-like ER kinase (PERK) by shRNA, we found that the knockdown of PERK increased virus loads in the cells, which was consistent with the result on 4-phenylbutyrate (4-PBA) treatment. We next determined whether 2-Deoxy-d-glucose (2-DG), an ER stress inducer, possessed antiviral activity against PEDV infection. Plaque formation assay, RT-PCR and Western blot analysis suggested that 2-DG might inhibit virus infection by affecting viral protein translation during the early stage of virus infection. Interestingly, we also found that 2-DG treatment could affect virus assembly, which is similar to previous studies on influenza virus. All these results support the therapeutic potential of using 2-DG or glucose/mannose analogs to induce the UPR to block virus replication.
Collapse
Affiliation(s)
- Yue Wang
- College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing, Jiangsu Province, 210095, China
| | - Jia-rong Li
- College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing, Jiangsu Province, 210095, China
| | - Ming-xia Sun
- College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing, Jiangsu Province, 210095, China
| | - Bo Ni
- College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing, Jiangsu Province, 210095, China
| | - Changchao Huan
- College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing, Jiangsu Province, 210095, China
| | - Li Huang
- College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing, Jiangsu Province, 210095, China
| | - Chen Li
- College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing, Jiangsu Province, 210095, China
| | - Hong-jie Fan
- College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing, Jiangsu Province, 210095, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China
| | - Xiao-feng Ren
- College of Veterinary Medicine, Northeast Agricultural University, 59 Mucai Street, Harbin, Xiangfang District, 150030, China.
| | - Xiang Mao
- College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing, Jiangsu Province, 210095, China.
| |
Collapse
|
44
|
Sakai M, Kankanamge PJ, Shoji J, Kawata S, Tochikura TS, Kawai A. Studies on the Conditions Required for Structural and Functional Maturation of Rabies Virus Glycoprotein (G) in G cDNA-Transfected Cells. Microbiol Immunol 2013; 48:853-64. [PMID: 15557743 DOI: 10.1111/j.1348-0421.2004.tb03617.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
When the rabies virus G cDNA was expressed with the help of T7 RNA polymerase provided by a recombinant vaccinia virus (RVV-T7), functional G proteins were produced in terms of their ability to induce low pH-dependent syncytium formation and the formation of conformational epitopes, including the acid-sensitive epitope recognized by mAb #1-30-44. Such an ability and the 1-30-44 epitope formation, however, were not associated with the G gene products when G cDNA was expressed without the help of RVV-T7 using a tetracycline-regulated expression vector (pTet-G), although they were normally transported to the surface of established G protein-producing BHK-21 (G-BHK) cells. But, when the G-BHK cells were treated with 2.5 m M sodium butyrate (NaB) after the removal of tetracycline, we could observe not only a much increased frequency of G protein-producing cells, but also the greatly enhanced maturation of the protein. Another short acylate, sodium propionate (NaP), similarly induced increased G protein synthesis at a concentration of 2.5 m M as NaB; however, such proteins were mostly not endowed with the fusion activity nor the 1-30-44 epitope, while NaP at a higher concentration as 5.0 m M did induce similarly the increased production and enhanced maturation of G protein, including the 1-30-44 epitope formation. From these results, we conclude that functional maturation of G protein to acquire fusogenic activity is correlated with 1-30-44 epitope formation, and 2.5 m M NaB not only stimulates G protein production, but also provides such cellular conditions as are required for the structural and functional maturation of the protein.
Collapse
Affiliation(s)
- Mai Sakai
- Department of Molecular Microbiology, Graduate School of Pharmaceutical Sciences, Kyoto University, Japan
| | | | | | | | | | | |
Collapse
|
45
|
Ferris SP, Jaber NS, Molinari M, Arvan P, Kaufman RJ. UDP-glucose:glycoprotein glucosyltransferase (UGGT1) promotes substrate solubility in the endoplasmic reticulum. Mol Biol Cell 2013; 24:2597-608. [PMID: 23864712 PMCID: PMC3756913 DOI: 10.1091/mbc.e13-02-0101] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Revised: 06/05/2013] [Accepted: 06/28/2013] [Indexed: 01/31/2023] Open
Abstract
Protein folding in the endoplasmic reticulum (ER) is error prone, and ER quality control (ERQC) processes ensure that only correctly folded proteins are exported from the ER. Glycoproteins can be retained in the ER by ERQC, and this retention contributes to multiple human diseases, termed ER storage diseases. UDP-glucose:glycoprotein glucosyltransferase (UGGT1) acts as a central component of glycoprotein ERQC, monoglucosylating deglucosylated N-glycans of incompletely folded glycoproteins and promoting subsequent reassociation with the lectin-like chaperones calreticulin and calnexin. The extent to which UGGT1 influences glycoprotein folding, however, has only been investigated for a few selected substrates. Using mouse embryonic fibroblasts lacking UGGT1 or those with UGGT1 complementation, we investigated the effect of monoglucosylation on the soluble/insoluble distribution of two misfolded α1-antitrypsin (AAT) variants responsible for AAT deficiency disease: null Hong Kong (NHK) and Z allele. Whereas substrate solubility increases directly with the number of N-linked glycosylation sites, our results indicate that additional solubility is conferred by UGGT1 enzymatic activity. Monoglucosylation-dependent solubility decreases both BiP association with NHK and unfolded protein response activation, and the solubility increase is blocked in cells deficient for calreticulin. These results suggest that UGGT1-dependent monoglucosylation of N-linked glycoproteins promotes substrate solubility in the ER.
Collapse
Affiliation(s)
- Sean P. Ferris
- Department of Biological Chemistry and Medical Scientist Training Program, University of Michigan Medical School, Ann Arbor, MI 48109-1621
| | - Nikita S. Jaber
- Michigan Comprehensive Diabetes Center, University of Michigan Medical School, Ann Arbor, MI 48105-1910
| | - Maurizio Molinari
- Institute for Research in Biomedicine, CH-6500 Bellinzona, Switzerland
- Ecole Polytechnique Federale de Lausanne, School of Life Sciences, CH-1015 Lausanne, Switzerland
| | - Peter Arvan
- Michigan Comprehensive Diabetes Center, University of Michigan Medical School, Ann Arbor, MI 48105-1910
| | - Randal J. Kaufman
- Degenerative Disease Research, Center for Neuroscience, Aging, and Stem Cell Research, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037
| |
Collapse
|
46
|
Dadehbeigi N, Dickson AJ. Application of a nonradioactive method of measuring protein synthesis in industrially relevant Chinese hamster ovary cells. Biotechnol Prog 2013; 29:1043-9. [PMID: 23749410 DOI: 10.1002/btpr.1750] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2013] [Revised: 04/11/2013] [Indexed: 01/08/2023]
Abstract
Due to the high medical and commercial value of recombinant proteins for clinical and diagnostic purposes, the protein synthesis machinery of mammalian host cells is the subject of extensive research by the biopharmaceutical industry. RNA translation and protein synthesis are steps that may determine the extent of growth and productivity of host cells. To address the problems of utilization of current radioisotope methods with proprietary media, we have focused on the application of an alternative method of measuring protein synthesis in recombinant Chinese hamster ovary (CHO) cells. This method employs puromycin as a nonradioactive label which incorporates into nascent polypeptide chains and is detectable by western blotting. This method, which is referred to as SUnSET, successfully demonstrated the expected changes in protein synthesis in conditions that inhibit and restore translation activity and was reproducibly quantifiable. The study of the effects of feed and sodium butyrate addition on protein synthesis by SUnSET revealed an increase following 1 h feed supplementation while a high concentration of sodium butyrate was able to decrease translation during the same treatment period. Finally, SUnSET was used to compare protein synthesis activity during batch culture of the CHO cell line in relation to growth. The results indicate that as the cells approached the end of batch culture, the global rate of protein synthesis declined in parallel with the decreasing growth rate. In conclusion, this method can be used as a "snapshot" to directly monitor the effects of different culture conditions and treatments on translation in recombinant host cells.
Collapse
Affiliation(s)
- Nazanin Dadehbeigi
- Faculty of Life Sciences, University of Manchester, Michael Smith Building, Manchester, UK, M13 9PT.
| | | |
Collapse
|
47
|
Abstract
The exocrine pancreas is the organ with the highest level of protein synthesis in the adult--each day the pancreas produces litres of fluid filled with enzymes that are capable of breaking down nearly all organic substances. For optimal health, the pancreas must produce sufficient enzymes of the right character to match the dietary intake. Disruption of normal pancreatic function occurs primarily as a result of dysfunction of the acinar cells that produce these digestive enzymes, and can lead to acute or chronic diseases. For many years, the prevailing dogma has been that inappropriate intracellular activation of the digestive enzymes produced by acinar cells was the key to pancreatic inflammatory diseases, as digestive enzymes themselves are potentially harmful to the cells that secrete them. However, we now know that many stressors can affect pancreatic acinar cells, and that these stressors can independently trigger pancreatic pathology through various mechanisms. This Review focuses on protein synthesis and active digestive enzymes--two key stressors faced by the acinar cell that are likely to be the major drivers of pathology encountered in the pancreas.
Collapse
|
48
|
Cao SS, Zimmermann EM, Chuang BM, Song B, Nwokoye A, Wilkinson JE, Eaton KA, Kaufman RJ. The unfolded protein response and chemical chaperones reduce protein misfolding and colitis in mice. Gastroenterology 2013; 144:989-1000.e6. [PMID: 23336977 PMCID: PMC3751190 DOI: 10.1053/j.gastro.2013.01.023] [Citation(s) in RCA: 168] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Revised: 01/10/2013] [Accepted: 01/13/2013] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Endoplasmic reticulum (ER) stress has been associated with development of inflammatory bowel disease. We examined the effects of ER stress-induced chaperone response and the orally active chemical chaperones tauroursodeoxycholate (TUDCA) and 4-phenylbutyrate (PBA), which facilitate protein folding and reduce ER stress, in mice with colitis. METHODS We used dextran sulfate sodium (DSS) to induce colitis in mice that do not express the transcription factor ATF6α or the protein chaperone P58(IPK). We examined the effects of TUDCA and PBA in cultured intestinal epithelial cells (IECs); in wild-type, P58(IPK-/-), and Atf6α(-/-) mice with colitis; and in Il10(-/-) mice. RESULTS P58(IPK-/-) and Atf6α(-/-) mice developed more severe colitis following administration of DSS than wild-type mice. IECs from P58(IPK-/-) mice had excessive ER stress, and apoptotic signaling was activated in IECs from Atf6α(-/-) mice. Inflammatory stimuli induced ER stress signals in cultured IECs, which were reduced by incubation with TUDCA or PBA. Oral administration of either PBA or TUDCA reduced features of DSS-induced acute and chronic colitis in wild-type mice, the colitis that develops in Il10(-/-) mice, and DSS-induced colitis in P58(IPK-/-) and Atf6α(-/-) mice. Reduced signs of colonic inflammation in these mice were associated with significantly decreased ER stress in colonic epithelial cells. CONCLUSIONS The unfolded protein response induces expression of genes that encode chaperones involved in ER protein folding; these factors prevent induction of colitis in mice. Chemical chaperones such as TUDCA and PBA alleviate different forms of colitis in mice and might be developed for treatment of inflammatory bowel diseases.
Collapse
Affiliation(s)
- Stewart Siyan Cao
- Del E. Webb Neuroscience, Aging and Stem Cell Research Center, Sanford Burnham Medical Research Institute, La Jolla, CA 92037, USA
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Suh DH, Kim MK, Kim HS, Chung HH, Song YS. Unfolded protein response to autophagy as a promising druggable target for anticancer therapy. Ann N Y Acad Sci 2013; 1271:20-32. [PMID: 23050960 PMCID: PMC3499662 DOI: 10.1111/j.1749-6632.2012.06739.x] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The endoplasmic reticulum (ER) is responsible for protein processing. In rapidly proliferating tumor cells, the ER tends to be overloaded with unfolded and misfolded proteins due to high metabolic demand. With the limited protein-folding capacity of the ER, tumor cells often suffer from more ER stress than do normal cells. Thus, cellular stress responses to cope with ER stress, such as the unfolded protein response (UPR) and autophagy, might be more activated in cancer cells than in normal cells. The complex signaling pathways from the UPR to autophagy provide promising druggable targets; a number of UPR/autophagy-targeted anticancer agents are currently in development in preclinical and clinical studies. In this short review we will discuss the potential anticancer efficacy of modulators of cellular stress responses, especially UPR and autophagy, on the basis of their signaling pathways. In addition, the current developmental status of the UPR/autophagy-targeted agents will be discussed.
Collapse
Affiliation(s)
- Dong Hoon Suh
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | | | | | | | | |
Collapse
|
50
|
4-Phenylbutyric acid reduces endoplasmic reticulum stress, trypsin activation, and acinar cell apoptosis while increasing secretion in rat pancreatic acini. Pancreas 2013; 42:92-101. [PMID: 22889983 DOI: 10.1097/mpa.0b013e318259f6ca] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVES Endoplasmic reticulum (ER) stress leads to misfolded proteins inside the ER and initiates unfolded protein response (UPR). Unfolded protein response components are involved in pancreatic function and activated during pancreatitis. However, the exact role of ER stress in the exocrine pancreas is unclear. The present study examined the effects of 4-phenylbutyric acid (4-PBA), an ER chaperone, on acini and UPR components. METHODS Rat acini were stimulated with cholecystokinin (10 pmol/L to 10 nmol/L) with or without preincubation of 4-PBA. The UPR components were analyzed, including chaperone-binding protein, protein kinaselike ER kinase, X-box-binding protein 1, c-Jun NH(2)-terminal kinase, CCAAT/enhancer-binding protein homologous protein, caspase 3, and apoptosis. Effects of 4-PBA were measured on secretion, calcium, and trypsin activation. RESULTS 4-Phenylbutyric acid led to an increase of secretion, whereas trypsin activation with supraphysiological cholecystokinin was significantly reduced. 4-Phenylbutyric acid prevented chaperone-binding protein up-regulation, diminished protein kinaselike ER kinase, and c-Jun NH2-terminal kinase phosphorylation, prohibited X-box-binding protein 1 splicing and CCAAT/enhancer-binding protein homologous protein expression, caspase 3 activation, and apoptosis caused by supraphysiological cholecystokinin. CONCLUSION By incubation with 4-PBA, beneficial in urea cycle deficiency, it was possible to enhance enzyme secretion to suppress trypsin activation, UPR activation, and proapoptotic pathways. The data hint new perspectives for the use of chemical chaperones in pancreatic diseases.
Collapse
|