51
|
Breczko W, Lemancewicz D, Dzięcioł J, Kłoczko J, Bołkun Ł. High immunoproteasome concentration in the plasma of patients with newly diagnosed multiple myeloma treated with bortezomib is predictive of longer OS. Adv Med Sci 2021; 66:21-27. [PMID: 33246214 DOI: 10.1016/j.advms.2020.11.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 10/23/2020] [Accepted: 11/17/2020] [Indexed: 11/28/2022]
Abstract
PURPOSE Proteasome inhibitors (PI) bortezomib or carfilzomib among them, play a crucial role in the modern standard therapy for multiple myeloma (MM). In this study, we intended to evaluate whether immunoproteasome (IMP) concentration could act as an effective biomarker which determines the probability of response to treatment with bortezomib, in order to detect groups of patients who are more likely to respond to treatment with PI. MATERIALS AND METHODS In our study, we evaluated IMP concentration in the plasma of 40 patients with monoclonal gammopathy of undetermined significance (MGUS) and 116 patients with newly diagnosed MM during treatment with or without PI. RESULTS The values of all the studied parameters after the applied chemotherapy in the responders' group of patients declined considerably during the consecutive cycles of chemotherapy compared to their initial levels. On the contrary, in the group of non-responders, we observed no change in the measured IMP parameters during the consecutive cycles of therapy. We also showed that higher baseline IMP concentration might indicate longer overall survival (OS) in all patients. CONCLUSIONS Our results indicate that assessing plasma IMP concentration can be applied as a strong biomarker for predicting clinical response to treatment and OS in patients with newly diagnosed MM.
Collapse
Affiliation(s)
- Wioletta Breczko
- Department of Hematology, Medical University of Bialystok, Bialystok, Poland
| | - Dorota Lemancewicz
- Department of Hematology, Medical University of Bialystok, Bialystok, Poland; Department of Human Anatomy, Medical University of Bialystok, Bialystok, Poland
| | - Janusz Dzięcioł
- Department of Human Anatomy, Medical University of Bialystok, Bialystok, Poland
| | - Janusz Kłoczko
- Department of Hematology, Medical University of Bialystok, Bialystok, Poland
| | - Łukasz Bołkun
- Department of Hematology, Medical University of Bialystok, Bialystok, Poland.
| |
Collapse
|
52
|
Zhou D, Borsa M, Simon AK. Hallmarks and detection techniques of cellular senescence and cellular ageing in immune cells. Aging Cell 2021; 20:e13316. [PMID: 33524238 PMCID: PMC7884036 DOI: 10.1111/acel.13316] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 01/03/2021] [Accepted: 01/09/2021] [Indexed: 12/15/2022] Open
Abstract
The ageing of the global population brings about unprecedented challenges. Chronic age-related diseases in an increasing number of people represent an enormous burden for health and social care. The immune system deteriorates during ageing and contributes to many of these age-associated diseases due to its pivotal role in pathogen clearance, tissue homeostasis and maintenance. Moreover, in order to develop treatments for COVID-19, we urgently need to acquire more knowledge about the aged immune system, as older adults are disproportionally and more severely affected. Changes with age lead to impaired responses to infections, malignancies and vaccination, and are accompanied by chronic, low-degree inflammation, which together is termed immunosenescence. However, the molecular and cellular mechanisms that underlie immunosenescence, termed immune cell senescence, are mostly unknown. Cellular senescence, characterised by an irreversible cell cycle arrest, is thought to be the cause of tissue and organismal ageing. Thus, better understanding of cellular senescence in immune populations at single-cell level may provide us with insight into how immune cell senescence develops over the life time of an individual. In this review, we will briefly introduce the phenotypic characterisation of aged innate and adaptive immune cells, which also contributes to overall immunosenescence, including subsets and function. Next, we will focus on the different hallmarks of cellular senescence and cellular ageing, and the detection techniques most suitable for immune cells. Applying these techniques will deepen our understanding of immune cell senescence and to discover potential druggable pathways, which can be modulated to reverse immune ageing.
Collapse
Affiliation(s)
- Dingxi Zhou
- The Kennedy Institute of RheumatologyUniversity of OxfordOxfordUK
| | - Mariana Borsa
- The Kennedy Institute of RheumatologyUniversity of OxfordOxfordUK
| | | |
Collapse
|
53
|
Maresh ME, Salazar-Chaparro AF, Trader DJ. Methods for the discovery of small molecules to monitor and perturb the activity of the human proteasome. Future Med Chem 2021; 13:99-116. [PMID: 33275045 PMCID: PMC7857359 DOI: 10.4155/fmc-2020-0288] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 10/14/2020] [Indexed: 12/12/2022] Open
Abstract
Regulating protein production and degradation is critical to maintaining cellular homeostasis. The proteasome is a key player in keeping proteins at the proper levels. However, proteasome activity can be altered in certain disease states, such as blood cancers and neurodegenerative diseases. Cancers often exhibit enhanced proteasomal activity, as protein synthesis is increased in these cells compared with normal cells. Conversely, neurodegenerative diseases are characterized by protein accumulation, leading to reduced proteasome activity. As a result, the proteasome has emerged as a target for therapeutic intervention. The potential of the proteasome as a therapeutic target has come from studies involving chemical stimulators and inhibitors, and the development of a suite of assays and probes that can be used to monitor proteasome activity with purified enzyme and in live cells.
Collapse
Affiliation(s)
- Marianne E Maresh
- Department of Medicinal Chemistry & Molecular Pharmacology, Purdue University, 575 West Stadium Avenue, West Lafayette, IN 47907, USA
| | - Andres F Salazar-Chaparro
- Department of Medicinal Chemistry & Molecular Pharmacology, Purdue University, 575 West Stadium Avenue, West Lafayette, IN 47907, USA
| | - Darci J Trader
- Department of Medicinal Chemistry & Molecular Pharmacology, Purdue University, 575 West Stadium Avenue, West Lafayette, IN 47907, USA
| |
Collapse
|
54
|
Gomez-Perosanz M, Ras-Carmona A, Lafuente EM, Reche PA. Identification of CD8 + T cell epitopes through proteasome cleavage site predictions. BMC Bioinformatics 2020; 21:484. [PMID: 33308150 PMCID: PMC7733697 DOI: 10.1186/s12859-020-03782-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 09/28/2020] [Indexed: 01/08/2023] Open
Abstract
Background We previously introduced PCPS (Proteasome Cleavage Prediction Server), a web-based tool to predict proteasome cleavage sites using n-grams. Here, we evaluated the ability of PCPS immunoproteasome cleavage model to discriminate CD8+ T cell epitopes. Results We first assembled an epitope dataset consisting of 844 unique virus-specific CD8+ T cell epitopes and their source proteins. We then analyzed cleavage predictions by PCPS immunoproteasome cleavage model on this dataset and compared them with those provided by a related method implemented by NetChop web server. PCPS was clearly superior to NetChop in term of sensitivity (0.89 vs. 0.79) but somewhat inferior with regard to specificity (0.55 vs. 0.60). Judging by the Mathew’s Correlation Coefficient, PCPS predictions were overall superior to those provided by NetChop (0.46 vs. 0.39). We next analyzed the power of C-terminal cleavage predictions provided by the same PCPS model to discriminate CD8+ T cell epitopes, finding that they could be discriminated from random peptides with an accuracy of 0.74. Following these results, we tuned the PCPS web server to predict CD8+ T cell epitopes and predicted the entire SARS-CoV-2 epitope space. Conclusions We report an improved version of PCPS named iPCPS for predicting proteasome cleavage sites and peptides with CD8+ T cell epitope features. iPCPS is available for free public use at https://imed.med.ucm.es/Tools/pcps/.
Collapse
Affiliation(s)
- Marta Gomez-Perosanz
- Laboratory of Immunomedicine, Department of Immunology, Faculty of Medicine, Complutense University of Madrid, Pza Ramon y Cajal, s/n, 28040, Madrid, Spain
| | - Alvaro Ras-Carmona
- Laboratory of Immunomedicine, Department of Immunology, Faculty of Medicine, Complutense University of Madrid, Pza Ramon y Cajal, s/n, 28040, Madrid, Spain
| | - Esther M Lafuente
- Laboratory of Immunomedicine, Department of Immunology, Faculty of Medicine, Complutense University of Madrid, Pza Ramon y Cajal, s/n, 28040, Madrid, Spain
| | - Pedro A Reche
- Laboratory of Immunomedicine, Department of Immunology, Faculty of Medicine, Complutense University of Madrid, Pza Ramon y Cajal, s/n, 28040, Madrid, Spain.
| |
Collapse
|
55
|
Sun C, Jia G, Wang X, Wang Y, Liu Y. Immunoproteasome is up-regulated in rotenone-induced Parkinson's disease rat model. Neurosci Lett 2020; 738:135360. [PMID: 32905834 DOI: 10.1016/j.neulet.2020.135360] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 08/14/2020] [Accepted: 09/03/2020] [Indexed: 12/21/2022]
Abstract
The study was to investigate whether immunoproteasome (i-proteasome) and its downstream pathway are related to the pathogenesis of Parkinson's disease (PD). Rats were treated with rotenone showed significant weight loss and dyskinesia, which is consistent with the degeneration of TH-positive neurons and the activation of Iba-1-positive microglia/macrophages. Two major catalytic subunits of i-proteasome (PSMB9 and PSMB8) were seldom expressed in rat substantia nigra (SN) under normal condition, but they were significantly up-regulated with the release of TNF-α and IFN-γ after exposure to rotenone. In addition, compared with control group, the antigen presentation-related proteins antigen peptide transporter (TAP) 1, TAP2, major histocompatibility complex (MHC)-I and MHC-II levels were significantly up-regulated in rotenone group, which was in line with the accumulation of α-syn. These findings suggested that i-proteasome and antigen presentation pathways (related proteins) were upregulated by rotenone in a PD rat model.
Collapse
Affiliation(s)
- Congcong Sun
- Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, 250012, China
| | - Guoyong Jia
- Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, 250012, China
| | - Xingbang Wang
- Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, 250012, China
| | - Yun Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu Province, 221000, China
| | - Yiming Liu
- Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, 250012, China.
| |
Collapse
|
56
|
Liao J, An X, Yang X, Lin QY, Liu S, Xie Y, Bai J, Xia YL, Li HH. Deficiency of LMP10 Attenuates Diet-Induced Atherosclerosis by Inhibiting Macrophage Polarization and Inflammation in Apolipoprotein E Deficient Mice. Front Cell Dev Biol 2020; 8:592048. [PMID: 33195259 PMCID: PMC7644912 DOI: 10.3389/fcell.2020.592048] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 10/05/2020] [Indexed: 12/22/2022] Open
Abstract
Macrophage polarization and inflammation are key factors for the onset and progression of atherosclerosis. The immunoproteasome complex consists of three inducible catalytic subunits (LMP2, LMP10, and LMP7) that play a critical role in the regulation of these risk factors. We recently demonstrated that the LMP7 subunit promotes diet-induced atherosclerosis via inhibition of MERTK-mediated efferocytosis. Here, we explored the role of another subunit of LMP10 in the disease process, using ApoE knockout (ko) mice fed on an atherogenic diet (ATD) containing 0.5% cholesterol and 20% fat for 8 weeks as an in vivo atherosclerosis model. We observed that ATD significantly upregulated LMP10 expression in aortic lesions, which were primarily co-localized with plaque macrophages. Conversely, deletion of LMP10 markedly attenuated atherosclerotic lesion area, CD68+ macrophage accumulation, and necrotic core expansion in the plaques, but did not change plasma metabolic parameters, lesional SM22α+ smooth muscle cells, or collagen content. Myeloid-specific deletion of LMP10 by bone marrow transplantation resulted in similar phenotypes. Furthermore, deletion of LMP10 remarkably reduced aortic macrophage infiltration and increased M2/M1 ratio, accompanied by decreased expression of pro-inflammatory M1 cytokines (MCP-1, IL-1, and IL-6) and increased expression of anti-inflammatory M2 cytokines (IL-4 and IL-10). In addition, we confirmed in cultured macrophages that LMP10 deletion blunted macrophage polarization and inflammation during ox-LDL-induced foam cell formation in vitro, which was associated with decreased IκBα degradation and NF-κB activation. Our results show that the immunoproteasome subunit LMP10 promoted diet-induced atherosclerosis in ApoE ko mice possibly through regulation of NF-κB-mediated macrophage polarization and inflammation. Targeting LMP10 may represent a new therapeutic approach for atherosclerosis.
Collapse
Affiliation(s)
- Jiawei Liao
- Department of Cardiology, Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xiangbo An
- Department of Interventional Therapy, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xiaolei Yang
- Department of Cardiology, Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Qiu-Yue Lin
- Department of Cardiology, Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Shuang Liu
- Department of Occupational and Environmental Health, School of Public Health, Dalian Medical University, Dalian, China
| | - Yunpeng Xie
- Department of Cardiology, Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jie Bai
- Department of Occupational and Environmental Health, School of Public Health, Dalian Medical University, Dalian, China
| | - Yun-Long Xia
- Department of Cardiology, Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Hui-Hua Li
- Department of Cardiology, Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, China.,Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Department of Emergency Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
57
|
Reduced Proteasome Activity and Enhanced Autophagy in Blood Cells of Psoriatic Patients. Int J Mol Sci 2020; 21:ijms21207608. [PMID: 33066703 PMCID: PMC7589048 DOI: 10.3390/ijms21207608] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/09/2020] [Accepted: 10/12/2020] [Indexed: 02/06/2023] Open
Abstract
Psoriasis is a skin disease that is accompanied by oxidative stress resulting in modification of cell components, including proteins. Therefore, we investigated the relationship between the intensity of oxidative stress and the expression and activity of the proteasomal system as well as autophagy, responsible for the degradation of oxidatively modified proteins in the blood cells of patients with psoriasis. Our results showed that the caspase-like, trypsin-like, and chymotrypsin-like activity of the 20S proteasome in lymphocytes, erythrocytes, and granulocytes was lower, while the expression of constitutive proteasome and immunoproteasome subunits in lymphocytes was increased cells of psoriatic patients compared to healthy subjects. Conversely, the expression of constitutive subunits in erythrocytes, and both constitutive and immunoproteasomal subunits in granulocytes were reduced. However, a significant increase in the autophagy flux (assessed using LC3BII/LC3BI ratio) independent of the AKT pathway was observed. The levels of 4-HNE, 4-HNE-protein adducts, and proteins carbonyl groups were significantly higher in the blood cells of psoriatic patients. The decreased activity of the 20S proteasome together with the increased autophagy and the significantly increased level of proteins carbonyl groups and 4-HNE-protein adducts indicate a proteostatic imbalance in the blood cells of patients with psoriasis.
Collapse
|
58
|
Proteostasis Disturbances and Inflammation in Neurodegenerative Diseases. Cells 2020; 9:cells9102183. [PMID: 32998318 PMCID: PMC7601929 DOI: 10.3390/cells9102183] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/21/2020] [Accepted: 09/24/2020] [Indexed: 12/11/2022] Open
Abstract
Protein homeostasis (proteostasis) disturbances and inflammation are evident in normal aging and some age-related neurodegenerative diseases. While the proteostasis network maintains the integrity of intracellular and extracellular functional proteins, inflammation is a biological response to harmful stimuli. Cellular stress conditions can cause protein damage, thus exacerbating protein misfolding and leading to an eventual overload of the degradation system. The regulation of proteostasis network is particularly important in postmitotic neurons due to their limited regenerative capacity. Therefore, maintaining balanced protein synthesis, handling unfolding, refolding, and degrading misfolded proteins are essential to preserve all cellular functions in the central nervous sysytem. Failing proteostasis may trigger inflammatory responses in glial cells, and the consequent release of inflammatory mediators may lead to disturbances in proteostasis. Here, we review the mechanisms of proteostasis and inflammatory response, emphasizing their role in the pathological hallmarks of neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. Furthermore, we discuss the interplay between proteostatic stress and excessive immune response that activates inflammation and leads to dysfunctional proteostasis.
Collapse
|
59
|
Kiuchi T, Tomaru U, Ishizu A, Imagawa M, Iwasaki S, Suzuki A, Otsuka N, Ohhara Y, Kinoshita I, Matsuno Y, Dosaka-Akita H, Kasahara M. Expression of the immunoproteasome subunit β5i in non-small cell lung carcinomas. J Clin Pathol 2020; 74:300-306. [PMID: 32943490 DOI: 10.1136/jclinpath-2020-206618] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 06/24/2020] [Accepted: 06/27/2020] [Indexed: 12/25/2022]
Abstract
AIM The immunoproteasome is a specific proteasome isoform whose proteolytic activity enhances the generation of antigenic peptides to be presented by major histocompatibility complex class I molecules to CD8+ T cells. Physiologically, it is expressed abundantly in immune cells and is induced in somatic cells by cytokines, especially interferon-γ. Recently, variable expression of immunoproteasomes has been demonstrated in different types of cancers. However, the clinical significance of immunoproteasome expression in malignant tumours is poorly understood. In this study, we performed clinicopathological evaluation of immunoproteasome subunit β5i in non-small cell lung carcinomas (NSCLCs). METHODS Tumour tissues were collected from 155 patients with NSCLCs, and immunohistochemical analysis for β5i was performed in relation to the prognosis of patients. RESULTS High expression of β5i was found in about 20% of all NSCLCs and was found significantly more frequently (40%) in the adenocarcinoma subset. High expression of β5i was associated with a better 5-year relative survival rate in patients with pStage I to II adenocarcinoma and was also a significant and independent favourable prognostic factor in adenocarcinoma patients. In addition, when we performed in vitro analysis using NSCLC cell lines, combined treatment with the immunoproteasome-specific inhibitor ONX0914 and the proteasome inhibitor MG132 enhanced cell death in β5i-expressing NSCLC cell lines. CONCLUSION The expression of immunoproteasome can be explored as both a prognostic factor and a potential therapeutic target in NSCLCs. Since immunoproteasomes have crucial role in the antigen presentation, further studies may help to provide essential knowledge for therapeutic strategies in anticancer immunotherapy.
Collapse
Affiliation(s)
- Takayuki Kiuchi
- Department of Pathology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Utano Tomaru
- Department of Pathology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Akihiro Ishizu
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Makoto Imagawa
- Department of Diagnostic Pathology, KKR Sapporo Medical Center, Sapporo, Hokkaido, Japan
| | - Sari Iwasaki
- Department of Diagnostic Pathology, KKR Sapporo Medical Center, Sapporo, Hokkaido, Japan
| | - Akira Suzuki
- Department of Diagnostic Pathology, KKR Sapporo Medical Center, Sapporo, Hokkaido, Japan
| | - Noriyuki Otsuka
- Department of Pathology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yoshihito Ohhara
- Department of Medical Oncology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Ichiro Kinoshita
- Department of Medical Oncology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yoshihiro Matsuno
- Department of Surgical Pathology, Hokkaido University Hospital, Sapporo, Japan
| | - Hirotoshi Dosaka-Akita
- Department of Medical Oncology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Masanori Kasahara
- Department of Pathology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
60
|
Chang HH, Cheng YC, Tsai WC, Chen Y. PSMB8 inhibition decreases tumor angiogenesis in glioblastoma through vascular endothelial growth factor A reduction. Cancer Sci 2020; 111:4142-4153. [PMID: 32816328 PMCID: PMC7648028 DOI: 10.1111/cas.14625] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 08/10/2020] [Accepted: 08/13/2020] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma, also known as glioblastoma multiforme (GBM), is a fast‐growing tumor and the most aggressive brain malignancy. Proteasome subunit beta type‐8 (PSMB8) is one of the 17 essential subunits for the complete assembly of the 20S proteasome complex. The aim of the present study was to evaluate the role of PSMB8 expression in GBM progression and angiogenesis. PSMB8 expression in glioblastoma LN229 and U87MG was knocked down by siRNA or inducible shRNA both in vitro and in vivo. After PSMB8 reduction, cell survival, migration, invasion, angiogenesis, and the related signaling cascades were evaluated. An orthotopic mouse tumor model was also provided to examine the angiogenesis within tumors. A GEO profile analysis indicated that high expression of PSMB8 mRNA in GBM patients was correlated with a low survival rate. In immunohistochemistry analysis, PSMB8 expression was higher in high‐grade than in low‐grade brain tumors. The proliferation, migration, and angiogenesis of human GBM cells were decreased by PSMB8 knockdown in vitro. Furthermore, phosphorylated focal adhesion kinase (p‐FAK), p‐paxillin, MMP2, MMP9, and cathepsin B were significantly reduced in LN229 cells. Integrin β1 and β3 were reduced in HUVEC after incubation with LN229‐conditioned medium. In an orthotopic mouse tumor model, inducible knockdown of PSMB8 reduced the expression of vascular endothelial growth factor (VEGF), VEGF receptor, and CD31 as well as the progression of human glioblastoma. In this article, we demonstrated the role of PSMB8 in glioblastoma progression, especially neovascularization in vitro and in vivo. These results may provide a target for the anti–angiogenic effect of PSMB8 in glioblastoma therapy in the future.
Collapse
Affiliation(s)
- Hsin-Han Chang
- Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan.,Graduate Institute of Life Science, National Defense Medical Center, Taipei, Taiwan
| | - Yu-Chen Cheng
- Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan.,Graduate Institute of Life Science, National Defense Medical Center, Taipei, Taiwan
| | - Wen-Chiuan Tsai
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Ying Chen
- Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan.,Graduate Institute of Life Science, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
61
|
Tundo GR, Sbardella D, Santoro AM, Coletta A, Oddone F, Grasso G, Milardi D, Lacal PM, Marini S, Purrello R, Graziani G, Coletta M. The proteasome as a druggable target with multiple therapeutic potentialities: Cutting and non-cutting edges. Pharmacol Ther 2020; 213:107579. [PMID: 32442437 PMCID: PMC7236745 DOI: 10.1016/j.pharmthera.2020.107579] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 05/05/2020] [Indexed: 01/10/2023]
Abstract
Ubiquitin Proteasome System (UPS) is an adaptable and finely tuned system that sustains proteostasis network under a large variety of physiopathological conditions. Its dysregulation is often associated with the onset and progression of human diseases; hence, UPS modulation has emerged as a promising new avenue for the development of treatments of several relevant pathologies, such as cancer and neurodegeneration. The clinical interest in proteasome inhibition has considerably increased after the FDA approval in 2003 of bortezomib for relapsed/refractory multiple myeloma, which is now used in the front-line setting. Thereafter, two other proteasome inhibitors (carfilzomib and ixazomib), designed to overcome resistance to bortezomib, have been approved for treatment-experienced patients, and a variety of novel inhibitors are currently under preclinical and clinical investigation not only for haematological malignancies but also for solid tumours. However, since UPS collapse leads to toxic misfolded proteins accumulation, proteasome is attracting even more interest as a target for the care of neurodegenerative diseases, which are sustained by UPS impairment. Thus, conceptually, proteasome activation represents an innovative and largely unexplored target for drug development. According to a multidisciplinary approach, spanning from chemistry, biochemistry, molecular biology to pharmacology, this review will summarize the most recent available literature regarding different aspects of proteasome biology, focusing on structure, function and regulation of proteasome in physiological and pathological processes, mostly cancer and neurodegenerative diseases, connecting biochemical features and clinical studies of proteasome targeting drugs.
Collapse
Affiliation(s)
- G R Tundo
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy.
| | | | - A M Santoro
- CNR, Institute of Crystallography, Catania, Italy
| | - A Coletta
- Department of Chemistry, University of Aarhus, Aarhus, Denmark
| | - F Oddone
- IRCCS-Fondazione Bietti, Rome, Italy
| | - G Grasso
- Department of Chemical Sciences, University of Catania, Catania, Italy
| | - D Milardi
- CNR, Institute of Crystallography, Catania, Italy
| | - P M Lacal
- Laboratory of Molecular Oncology, IDI-IRCCS, Rome, Italy
| | - S Marini
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy
| | - R Purrello
- Department of Chemical Sciences, University of Catania, Catania, Italy
| | - G Graziani
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.
| | - M Coletta
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy.
| |
Collapse
|
62
|
Kao SST, Bassiouni A, Ramezanpour M, Finnie J, Chegeni N, Colella AD, Chataway TK, Wormald PJ, Vreugde S, Psaltis AJ. Proteomic analysis of nasal mucus samples of healthy patients and patients with chronic rhinosinusitis. J Allergy Clin Immunol 2020; 147:168-178. [PMID: 32750382 DOI: 10.1016/j.jaci.2020.06.037] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 06/08/2020] [Accepted: 06/18/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Chronic rhinosinusitis (CRS) has a complex and multifactorial pathogenesis with a heterogeneous inflammatory profile. Proteomic analysis of nasal mucus may enable further understanding of protein abundances and biologic processes present in CRS and its endotypes compared with in healthy patients. OBJECTIVE Our aim was to determine differences in the nasal mucus proteome of healthy patients and patients with CRS. METHODS Nasal mucus was obtained from healthy patients, patients with CRS without nasal polyps (CRSsNP), and patients with CRS with nasal polyps (CRSwNP) before surgery. Gel electrophoresis was performed to fractionate the complex protein extracts before mass spectrometry analysis. Gene set enrichment analysis was performed on differentially expressed proteins. RESULTS A total of 33 patients were included in this study (12 healthy, 10 with CRSsNP, and 11 with CRSwNP). In all, 1142 proteins were identified in mucus samples from healthy patients, 761 in mucus samples from patients with CRSsNP, and 998 in mucus samples from patients with CRSwNP. Dysfunction in immunologic pathways, reduced cellular signaling, and increased cellular metabolism with associated tissue remodeling pathways were present in patients with CRS compared with in healthy patients. CONCLUSION Significant downregulation of mucosal immunity and antioxidant pathways with increased tissue modeling processes may account for the clinical manifestations of CRS. Ultimately, the differing proteome and biologic processes provide further insight into CRS pathogenesis and its endotypes.
Collapse
Affiliation(s)
- Stephen Shih-Teng Kao
- Department of Surgery-Otolaryngology Head and Neck Surgery, The University of Adelaide, Woodville South, Australia
| | - Ahmed Bassiouni
- Department of Surgery-Otolaryngology Head and Neck Surgery, The University of Adelaide, Woodville South, Australia
| | - Mahnaz Ramezanpour
- Department of Surgery-Otolaryngology Head and Neck Surgery, The University of Adelaide, Woodville South, Australia
| | - John Finnie
- Discipline of Anatomy and Pathology, Adelaide Medical School, The University of Adelaide and South Australia Pathology, Adelaide, Australia
| | - Nusha Chegeni
- Discipline of Anatomy and Pathology, Adelaide Medical School, The University of Adelaide and South Australia Pathology, Adelaide, Australia; Flinders Proteomic Facility, Department of Human Physiology, Flinders University, Bedford Park, Australia
| | - Alex D Colella
- Discipline of Anatomy and Pathology, Adelaide Medical School, The University of Adelaide and South Australia Pathology, Adelaide, Australia; Flinders Proteomic Facility, Department of Human Physiology, Flinders University, Bedford Park, Australia
| | - Timothy K Chataway
- Discipline of Anatomy and Pathology, Adelaide Medical School, The University of Adelaide and South Australia Pathology, Adelaide, Australia; Flinders Proteomic Facility, Department of Human Physiology, Flinders University, Bedford Park, Australia
| | - Peter-John Wormald
- Department of Surgery-Otolaryngology Head and Neck Surgery, The University of Adelaide, Woodville South, Australia
| | - Sarah Vreugde
- Department of Surgery-Otolaryngology Head and Neck Surgery, The University of Adelaide, Woodville South, Australia
| | - Alkis James Psaltis
- Department of Surgery-Otolaryngology Head and Neck Surgery, The University of Adelaide, Woodville South, Australia.
| |
Collapse
|
63
|
Nousbeck J, McAleer M, Hurault G, Kenny E, Harte K, Kezic S, Tanaka R, Irvine A. MicroRNA analysis of childhood atopic dermatitis reveals a role for miR‐451a*. Br J Dermatol 2020; 184:514-523. [DOI: 10.1111/bjd.19254] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/25/2020] [Indexed: 12/13/2022]
Affiliation(s)
- J. Nousbeck
- National Children’s Research Centre Crumlin, Dublin Ireland
- Clinical Medicine Trinity College Dublin Dublin Ireland
| | - M.A. McAleer
- National Children’s Research Centre Crumlin, Dublin Ireland
- Paediatric Dermatology Our Lady’s Children’s Hospital Crumlin, Dublin Ireland
| | - G. Hurault
- Department of Bioengineering Imperial College London London UK
| | - E. Kenny
- Department of Psychiatry Trinity Translational Medicine Institute St James’s Hospital Dublin Ireland
| | - K. Harte
- Department of Psychiatry Trinity Translational Medicine Institute St James’s Hospital Dublin Ireland
| | - S. Kezic
- Coronel Institute of Occupational Health Academic Medical Center Amsterdam the Netherlands
| | - R.J. Tanaka
- Department of Bioengineering Imperial College London London UK
| | - A.D. Irvine
- National Children’s Research Centre Crumlin, Dublin Ireland
- Clinical Medicine Trinity College Dublin Dublin Ireland
- Paediatric Dermatology Our Lady’s Children’s Hospital Crumlin, Dublin Ireland
| |
Collapse
|
64
|
Hypoxia-induced shift in the phenotype of proteasome from 26S toward immunoproteasome triggers loss of immunoprivilege of mesenchymal stem cells. Cell Death Dis 2020; 11:419. [PMID: 32499535 PMCID: PMC7272449 DOI: 10.1038/s41419-020-2634-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 05/01/2020] [Accepted: 05/05/2020] [Indexed: 02/06/2023]
Abstract
Allogeneic mesenchymal stem cells (MSCs) are immunoprivileged and are being investigated in phase I and phase II clinical trials to treat different degenerative and autoimmune diseases. In spite of encouraging outcome of initial trials, the long-term poor survival of transplanted cells in the host tissue has declined the overall enthusiasm. Recent analyses of allogeneic MSCs based studies confirm that after transplantation in the hypoxic or ischemic microenvironment of diseased tissues, MSCs become immunogenic and are rejected by recipient immune system. The immunoprivilege of MSCs is preserved by absence or negligible expression of cell surface antigen, human leukocyte antigen (HLA)-DRα. We found that in normoxic MSCs, 26S proteasome degrades HLA-DRα and maintains immunoprivilege of MSCs. The exposure to hypoxia leads to inactivation of 26S proteasome and formation of immunoproteasome in MSCs, which is associated with upregulation and activation of HLA-DRα, and as a result, MSCs become immunogenic. Furthermore, inhibition of immunoproteasome formation in hypoxic MSCs preserves the immunoprivilege. Therefore, hypoxia-induced shift in the phenotype of proteasome from 26S toward immunoproteasome triggers loss of immunoprivilege of allogeneic MSCs. The outcome of the current study may provide molecular targets to plan interventions to preserve immunoprivilege of allogeneic MSCs in the hypoxic or ischemic environment.
Collapse
|
65
|
Gender-specific changes in energy metabolism and protein degradation as major pathways affected in livers of mice treated with ibuprofen. Sci Rep 2020; 10:3386. [PMID: 32099006 PMCID: PMC7042271 DOI: 10.1038/s41598-020-60053-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 02/03/2020] [Indexed: 12/12/2022] Open
Abstract
Ibuprofen, an inhibitor of prostanoid biosynthesis, is a common pharmacological agent used for the management of pain, inflammation and fever. However, the chronic use of ibuprofen at high doses is associated with increased risk for cardiovascular, renal, gastrointestinal and liver injuries. The underlying mechanisms of ibuprofen-mediated effects on liver remain unclear. To determine the mechanisms and signaling pathways affected by ibuprofen (100 mg/kg/day for seven days), we performed proteomic profiling of male mice liver with quantitative liquid chromatography tandem mass spectrometry (LC-MS/MS) using ten-plex tandem mass tag (TMT) labeling. More than 300 proteins were significantly altered between the control and ibuprofen-treated groups. The data suggests that several major pathways including (1) energy metabolism, (2) protein degradation, (3) fatty acid metabolism and (4) antioxidant system are altered in livers from ibuprofen treated mice. Independent validation of protein changes in energy metabolism and the antioxidant system was carried out by Western blotting and showed sex-related differences. Proteasome and immunoproteasome activity/expression assays showed ibuprofen induced gender-specific proteasome and immunoproteasome dysfunction in liver. The study observed multifactorial gender-specific ibuprofen-mediated effects on mice liver and suggests that males and females are affected differently by ibuprofen.
Collapse
|
66
|
Proteasome Inhibitors: Harnessing Proteostasis to Combat Disease. Molecules 2020; 25:molecules25030671. [PMID: 32033280 PMCID: PMC7037493 DOI: 10.3390/molecules25030671] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/25/2020] [Accepted: 01/28/2020] [Indexed: 02/07/2023] Open
Abstract
The proteasome is the central component of the main cellular protein degradation pathway. During the past four decades, the critical function of the proteasome in numerous physiological processes has been revealed, and proteasome activity has been linked to various human diseases. The proteasome prevents the accumulation of misfolded proteins, controls the cell cycle, and regulates the immune response, to name a few important roles for this macromolecular "machine." As a therapeutic target, proteasome inhibitors have been approved for the treatment of multiple myeloma and mantle cell lymphoma. However, inability to sufficiently inhibit proteasome activity at tolerated doses has hampered efforts to expand the scope of proteasome inhibitor-based therapies. With emerging new modalities in myeloma, it might seem challenging to develop additional proteasome-based therapies. However, the constant development of new applications for proteasome inhibitors and deeper insights into the intricacies of protein homeostasis suggest that proteasome inhibitors might have novel therapeutic applications. Herein, we summarize the latest advances in proteasome inhibitor development and discuss the future of proteasome inhibitors and other proteasome-based therapies in combating human diseases.
Collapse
|
67
|
Clarke EC, Bradfute SB. The use of mice lacking type I or both type I and type II interferon responses in research on hemorrhagic fever viruses. Part 1: Potential effects on adaptive immunity and response to vaccination. Antiviral Res 2020; 174:104703. [DOI: 10.1016/j.antiviral.2019.104703] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 12/10/2019] [Accepted: 12/20/2019] [Indexed: 12/25/2022]
|
68
|
Liao J, Xie Y, Lin Q, Yang X, An X, Xia Y, Du J, Wang F, Li HH. Immunoproteasome subunit β5i regulates diet-induced atherosclerosis through altering MERTK-mediated efferocytosis in Apoe knockout mice. J Pathol 2020; 250:275-287. [PMID: 31758542 DOI: 10.1002/path.5368] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 11/01/2019] [Accepted: 11/20/2019] [Indexed: 12/29/2022]
Abstract
The immunoproteasome contains three catalytic subunits (β1i, β2i and β5i) that are important modulators of immune cell homeostasis. A previous study showed a correlation between β5i and human atherosclerotic plaque instability; however, the causative role of β5i in atherosclerosis and the underlying mechanisms remain unknown. Here we explored this issue in apolipoprotein E (Apoe) knockout (eKO) mice with genetic deletion or pharmacological inhibition of β5i. We found that β5i expression was upregulated in lesional macrophages after an atherogenic diet (ATD). β5i/Apoe double KO (dKO) mice fed on the ATD had a significant decrease in both lesion area and necrotic core area, compared with eKO controls. Moreover, dKO mice had less caspase-3+ apoptotic cell accumulation but enhanced efferocytosis of apoptotic cells and increased expression of Mer receptor tyrosine kinase (MERTK). Consistently, similar phenotypes were observed in eKO mice transplanted with dKO bone marrow or treated with β5i-specific inhibitor PR-957. Mechanistic studies in vitro revealed that β5i deletion reduced IκBα degradation and inhibited NF-κB activation, promoting Mertk transcription and efferocytosis, thereby attenuating apoptotic cell accumulation. In conclusion, we demonstrate that β5i plays an important role in diet-induced atherosclerosis by altering MERTK-mediated efferocytosis. β5i might be a potential pharmaceutical target against atherosclerosis. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Jiawei Liao
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, PR China
| | - Yunpeng Xie
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, PR China
| | - Qiuyue Lin
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, PR China
| | - Xiaolei Yang
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, PR China
| | - Xiangbo An
- Department of Interventional Therapy, First Affiliated Hospital of Dalian Medical University, Dalian, PR China
| | - Yunlong Xia
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, PR China
| | - Jie Du
- Beijing AnZhen Hospital, The Key Laboratory of Remodeling-Related Cardiovascular Diseases, School of Basic Medical Sciences, Capital Medical University, Beijing, PR China
| | - Feng Wang
- Department of Interventional Therapy, First Affiliated Hospital of Dalian Medical University, Dalian, PR China
| | - Hui-Hua Li
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, PR China
| |
Collapse
|
69
|
Kwon D, Kim SM, Correia MA. Cytochrome P450 endoplasmic reticulum-associated degradation (ERAD): therapeutic and pathophysiological implications. Acta Pharm Sin B 2020; 10:42-60. [PMID: 31993306 PMCID: PMC6976991 DOI: 10.1016/j.apsb.2019.11.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 10/30/2019] [Accepted: 10/31/2019] [Indexed: 02/07/2023] Open
Abstract
The hepatic endoplasmic reticulum (ER)-anchored cytochromes P450 (P450s) are mixed-function oxidases engaged in the biotransformation of physiologically relevant endobiotics as well as of myriad xenobiotics of therapeutic and environmental relevance. P450 ER-content and hence function is regulated by their coordinated hemoprotein syntheses and proteolytic turnover. Such P450 proteolytic turnover occurs through a process known as ER-associated degradation (ERAD) that involves ubiquitin-dependent proteasomal degradation (UPD) and/or autophagic-lysosomal degradation (ALD). Herein, on the basis of available literature reports and our own recent findings of in vitro as well as in vivo experimental studies, we discuss the therapeutic and pathophysiological implications of altered P450 ERAD and its plausible clinical relevance. We specifically (i) describe the P450 ERAD-machinery and how it may be repurposed for the generation of antigenic P450 peptides involved in P450 autoantibody pathogenesis in drug-induced acute hypersensitivity reactions and liver injury, or viral hepatitis; (ii) discuss the relevance of accelerated or disrupted P450-ERAD to the pharmacological and/or toxicological effects of clinically relevant P450 drug substrates; and (iii) detail the pathophysiological consequences of disrupted P450 ERAD, contributing to non-alcoholic fatty liver disease (NAFLD)/non-alcoholic steatohepatitis (NASH) under certain synergistic cellular conditions.
Collapse
Key Words
- 3MA, 3-methyladenine
- AAA, ATPases associated with various cellular activities
- ACC1, acetyl-CoA carboxylase 1
- ACC2, acetyl-CoA carboxylase 2
- ACHE, acetylcholinesterase
- ACOX1, acyl-CoA oxidase 1
- ALD, autophagic-lysosomal degradation
- AMPK1
- AP-1, activator protein 1
- ASK1, apoptosis signal-regulating kinase
- ATF2, activating transcription factor 2
- AdipoR1, gene of adiponectin receptor 1
- Atg14, autophagy-related 14
- CBZ, carbamazepine
- CHIP E3 ubiquitin ligase
- CHIP, carboxy-terminus of Hsc70-interacting protein
- Cytochromes P450
- Endoplasmic reticulum-associated degradation
- FOXO, forkhead box O
- Fas, fatty acid synthase
- GAPDH, glyceraldehyde 3-phosphate dehydrogenase
- INH, isoniazid
- IRS1, insulin receptor substrate 1
- Il-1β, interleukin 1 β
- Il-6, interleukin 6
- Insig1, insulin-induced gene 1
- JNK1
- Lpl, lipoprotein lipase
- Mcp1, chemokine (C–C motif) ligand 1
- Non-alcoholic fatty liver disease
- Non-alcoholic steatohepatitis
- Pgc1, peroxisome proliferator-activated receptor coactivator 1
- SREBP1c, sterol regulatory element binding transcription factor 1c
- Scd1, stearoyl-coenzyme A desaturase
- Tnf, tumor necrosis factor
- UPD, ubiquitin (Ub)-dependent proteasomal degradation
- Ub, ubiquitin
- gp78/AMFR E3 ubiquitin ligase
- gp78/AMFR, autocrine motility factor receptor
- shRNAi, shRNA interference
Collapse
|
70
|
Gómez-Flores M, Villarreal-Villarreal CD, Welsh O, Cárdenas-de la Garza JA, Sánchez-Meza E, Áncer-Arellano J, Martinez-Moreno A, Ocampo-Candiani J, Herz-Ruelas ME. Lower than expected mortality with dexamethasone in toxic epidermal necrolysis: 30-year experience of a Northern Mexico reference hospital. Int J Dermatol 2020; 59:e4-e5. [PMID: 31592544 DOI: 10.1111/ijd.14666] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 07/30/2019] [Accepted: 09/03/2019] [Indexed: 01/24/2023]
Affiliation(s)
- Minerva Gómez-Flores
- Department of Dermatology, University Hospital "Dr. José Eleuterio González", Universidad Autónoma de Nuevo León, Monterrey, México
| | - Cesar D Villarreal-Villarreal
- Department of Dermatology, University Hospital "Dr. José Eleuterio González", Universidad Autónoma de Nuevo León, Monterrey, México
| | - Oliverio Welsh
- Department of Dermatology, University Hospital "Dr. José Eleuterio González", Universidad Autónoma de Nuevo León, Monterrey, México
| | - Jesus A Cárdenas-de la Garza
- Department of Dermatology, University Hospital "Dr. José Eleuterio González", Universidad Autónoma de Nuevo León, Monterrey, México
| | - Emmanuel Sánchez-Meza
- Department of Dermatology, University Hospital "Dr. José Eleuterio González", Universidad Autónoma de Nuevo León, Monterrey, México
| | - Jesus Áncer-Arellano
- Department of Dermatology, University Hospital "Dr. José Eleuterio González", Universidad Autónoma de Nuevo León, Monterrey, México
| | - Adrian Martinez-Moreno
- Department of Dermatology, University Hospital "Dr. José Eleuterio González", Universidad Autónoma de Nuevo León, Monterrey, México
| | - Jorge Ocampo-Candiani
- Department of Dermatology, University Hospital "Dr. José Eleuterio González", Universidad Autónoma de Nuevo León, Monterrey, México
| | - Maira E Herz-Ruelas
- Department of Dermatology, University Hospital "Dr. José Eleuterio González", Universidad Autónoma de Nuevo León, Monterrey, México
| |
Collapse
|
71
|
Platelet Proteasome Activity and Metabolism Is Upregulated during Bacterial Sepsis. Int J Mol Sci 2019; 20:ijms20235961. [PMID: 31783490 PMCID: PMC6928740 DOI: 10.3390/ijms20235961] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 11/21/2019] [Accepted: 11/25/2019] [Indexed: 11/30/2022] Open
Abstract
Dysregulation of platelet function can contribute to the disease progression in sepsis. The proteasome represents a critical and vital element of cellular protein metabolism in platelets and its proteolytic activity has been associated with platelet function. However, the role of the platelet proteasome as well as its response to infection under conditions of sepsis have not been studied so far. We measured platelet proteasome activity by fluorescent substrates, degradation of poly-ubiquitinated proteins and cleavage of the proteasome substrate Talin-1 in the presence of living E. coli strains and in platelets isolated from sepsis patients. Upregulation of the proteasome activator PA28 (PSME1) was assessed by quantitative real-time PCR in platelets from sepsis patients. We show that co-incubation of platelets with living E. coli (UTI89) results in increased degradation of poly-ubiquitinated proteins and cleavage of Talin-1 by the proteasome. Proteasome activity and cleavage of Talin-1 was significantly increased in α-hemolysin (HlyA)-positive E. coli strains. Supporting these findings, proteasome activity was also increased in platelets of patients with sepsis. Finally, the proteasome activator PA28 (PSME1) was upregulated in this group of patients. In this study we demonstrate for the first time that the proteasome in platelets is activated in the septic milieu.
Collapse
|
72
|
Zerfas BL, Maresh ME, Trader DJ. The Immunoproteasome: An Emerging Target in Cancer and Autoimmune and Neurological Disorders. J Med Chem 2019; 63:1841-1858. [PMID: 31670954 DOI: 10.1021/acs.jmedchem.9b01226] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The immunoproteasome (iCP) is an isoform of the 20S proteasome that is expressed when cells are stressed or receive an inflammatory signal. The primary role of the iCP is to hydrolyze proteins into peptides that are compatible with being loaded into a MHC-I complex. When the activity of the iCP is dysregulated or highly expressed, it can lead to unwanted cell death. Some cancer types express the iCP rather than the standard proteasome, and selective inhibitors have been developed to exploit this difference. Here, we describe diseases known to be influenced by iCP activity and the current status for targeting the iCP to elicit a therapeutic response. We also describe a variety of chemical tools that have been developed to monitor the activity of the iCP in cells. Finally, we present the future outlook for targeting the iCP in a variety of disease types and with mechanisms besides inhibition.
Collapse
Affiliation(s)
- Breanna L Zerfas
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, 575 West Stadium Avenue, West Lafayette, Indiana 47907, United States
| | - Marianne E Maresh
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, 575 West Stadium Avenue, West Lafayette, Indiana 47907, United States
| | - Darci J Trader
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, 575 West Stadium Avenue, West Lafayette, Indiana 47907, United States
| |
Collapse
|
73
|
Jarvela J, Moyer M, Leahy P, Bonfield T, Fletcher D, Mkono WN, Aung H, Canaday DH, Dazard JE, Silver RF. Mycobacterium tuberculosis-Induced Bronchoalveolar Lavage Gene Expression Signature in Latent Tuberculosis Infection Is Dominated by Pleiotropic Effects of CD4 + T Cell-Dependent IFN-γ Production despite the Presence of Polyfunctional T Cells within the Airways. THE JOURNAL OF IMMUNOLOGY 2019; 203:2194-2209. [PMID: 31541022 DOI: 10.4049/jimmunol.1900230] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 08/08/2019] [Indexed: 11/19/2022]
Abstract
Tuberculosis (TB) remains a worldwide public health threat. Development of a more effective vaccination strategy to prevent pulmonary TB, the most common and contagious form of the disease, is a research priority for international TB control. A key to reaching this goal is improved understanding of the mechanisms of local immunity to Mycobacterium tuberculosis, the causative organism of TB. In this study, we evaluated global M. tuberculosis-induced gene expression in airway immune cells obtained by bronchoalveolar lavage (BAL) of individuals with latent TB infection (LTBI) and M. tuberculosis-naive controls. In prior studies, we demonstrated that BAL cells from LTBI individuals display substantial enrichment for M. tuberculosis-responsive CD4+ T cells compared with matched peripheral blood samples. We therefore specifically assessed the impact of the depletion of CD4+ and CD8+ T cells on M. tuberculosis-induced BAL cell gene expression in LTBI. Our studies identified 12 canonical pathways and a 47-gene signature that was both sensitive and specific for the contribution of CD4+ T cells to local recall responses to M. tuberculosis In contrast, depletion of CD8+ cells did not identify any genes that fit our strict criteria for inclusion in this signature. Although BAL CD4+ T cells in LTBI displayed polyfunctionality, the observed gene signature predominantly reflected the impact of IFN-γ production on a wide range of host immune responses. These findings provide a standard for comparison of the efficacy of standard bacillus Calmette-Guérin vaccination as well as novel TB vaccines now in development at impacting the initial response to re-exposure to M. tuberculosis in the human lung.
Collapse
Affiliation(s)
- Jessica Jarvela
- Division of Pulmonary, Critical Care and Sleep Medicine, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH 44106.,Division of Pulmonary, Critical Care and Sleep Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106
| | - Michelle Moyer
- Division of Pulmonary, Critical Care and Sleep Medicine, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH 44106.,Division of Pulmonary, Critical Care and Sleep Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106
| | - Patrick Leahy
- Case Western Reserve University Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106
| | - Tracey Bonfield
- Division of Pediatric Pulmonology, Allergy, and Immunology, Case Western Reserve University School of Medicine, Cleveland, OH 44106
| | - David Fletcher
- Division of Pediatric Pulmonology, Allergy, and Immunology, Case Western Reserve University School of Medicine, Cleveland, OH 44106
| | - Wambura N Mkono
- Division of Pulmonary, Critical Care and Sleep Medicine, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH 44106.,Division of Pulmonary, Critical Care and Sleep Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106.,Division of Pulmonary, Critical Care and Sleep Medicine, University Hospitals Cleveland Medical Center, Cleveland, OH 44106
| | - Htin Aung
- Division of Infectious Diseases and HIV Medicine, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH 44106.,Division of Infectious Diseases and HIV Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106; and
| | - David H Canaday
- Division of Infectious Diseases and HIV Medicine, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH 44106.,Division of Infectious Diseases and HIV Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106; and
| | - Jean-Eudes Dazard
- Center for Proteomics and Bioinformatics, Case Western Reserve University School of Medicine, Cleveland, OH 44106
| | - Richard F Silver
- Division of Pulmonary, Critical Care and Sleep Medicine, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH 44106; .,Division of Pulmonary, Critical Care and Sleep Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106.,Division of Pulmonary, Critical Care and Sleep Medicine, University Hospitals Cleveland Medical Center, Cleveland, OH 44106
| |
Collapse
|
74
|
Liu Q, Wang HY, He XJ. Induction of immunoproteasomes in porcine kidney (PK)-15 cells by interferon-γ and tumor necrosis factor-α. J Vet Med Sci 2019; 81:1776-1782. [PMID: 31548474 PMCID: PMC6943335 DOI: 10.1292/jvms.19-0157] [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] [Indexed: 11/30/2022] Open
Abstract
Immunoproteasome (i-proteasome) has both immune and non-immune functions and plays
important roles in controlling infections and combating illnesses. Our previous studies
suggest that interferon (IFN)-γ induces the expression of three immune-specific catalytic
subunits of the 20S proteasome that can replace their constitutive homologues to form the
i-proteasome in immune cells, such as porcine alveolar macrophages (AMs) in
vitro. However, i-proteasome levels and their modulation in non-immune cells
such as the epithelial cells in pigs remain unknown. Here, we investigated the expression
of i-proteasomes in non-immune cells (porcine kidney (PK)-15 cells) to determine
i-proteasome modulation upon stimulation of PK-15 cells with IFN-γ and tumor necrosis
factor (TNF)-α in vitro. The expression of i-proteasome subunits in PK-15
cells were regulated by IFN-γ and TNF-α. Remarkably, we found that the combination
treatment of IFN-γ and TNF-α increased the expression of i-proteasome subunits LMP2, LMP7,
and MECL-1 in PK-15 cells at transcriptional levels, but may decrease their expression at
translational level, compared to their expression levels induced by individual cytokine
treatments. These results provide critical insight into i-proteasome modulation in porcine
non-immune cells, contribute further to our understanding of i-proteasome function in
tissue pathogenesis and the development of antiviral adaptive immune responses against
intracellular infections.
Collapse
Affiliation(s)
- Qiang Liu
- Nanchong Key Laboratory of Disease Prevention, Control and Detection in Livestock and Poultry, Nanchong Vocational and Technical College, Nanchong 637131, China
| | - Huai Yu Wang
- Nanchong Key Laboratory of Disease Prevention, Control and Detection in Livestock and Poultry, Nanchong Vocational and Technical College, Nanchong 637131, China
| | - Xi-Jun He
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin 150069, China
| |
Collapse
|
75
|
Abstract
The proteasome degrades most cellular proteins in a controlled and tightly regulated manner and thereby controls many processes, including cell cycle, transcription, signalling, trafficking and protein quality control. Proteasomal degradation is vital in all cells and organisms, and dysfunction or failure of proteasomal degradation is associated with diverse human diseases, including cancer and neurodegeneration. Target selection is an important and well-established way to control protein degradation. In addition, mounting evidence indicates that cells adjust proteasome-mediated degradation to their needs by regulating proteasome abundance through the coordinated expression of proteasome subunits and assembly chaperones. Central to the regulation of proteasome assembly is TOR complex 1 (TORC1), which is the master regulator of cell growth and stress. This Review discusses how proteasome assembly and the regulation of proteasomal degradation are integrated with cellular physiology, including the interplay between the proteasome and autophagy pathways. Understanding these mechanisms has potential implications for disease therapy, as the misregulation of proteasome function contributes to human diseases such as cancer and neurodegeneration.
Collapse
|
76
|
Galli G, Poissonnier A, Guégan JP, Charrier M, Sisirak V, Lazaro E, Truchetet ME, Richez C, Legembre P, Blanco P. HIV protease inhibitors and autoimmunity: An odd, but promising idea. Clin Exp Rheumatol 2019; 18:102370. [PMID: 31404704 DOI: 10.1016/j.autrev.2019.102370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Accepted: 05/08/2019] [Indexed: 10/26/2022]
Affiliation(s)
- Gaël Galli
- Service de Médecine Interne, FHU ACRONIM, Hôpital Haut Lévêque, Centre Hospitalier Universitaire, Avenue Magellan, 33600 Pessac, France; Université de Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux, France; CNRS-UMR 5164, ImmunoConcEpt, Université de Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux, France
| | - Amanda Poissonnier
- CLCC Eugène Marquis, INSERM-UMR 1242, Université de Rennes, rue Bataille Flandres Dunkerque, 35042 Rennes, France; Equipe Ligue Contre Le Cancer, rue Bataille Flandres Dunkerque, 35042 Rennes, France
| | - Jean Philippe Guégan
- CLCC Eugène Marquis, INSERM-UMR 1242, Université de Rennes, rue Bataille Flandres Dunkerque, 35042 Rennes, France; Equipe Ligue Contre Le Cancer, rue Bataille Flandres Dunkerque, 35042 Rennes, France
| | - Manon Charrier
- Université de Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux, France; CNRS-UMR 5164, ImmunoConcEpt, Université de Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux, France; Service de Néphrologie, FHU ACRONIM, Hôpital Pellegrin, Centre Hospitalier Universitaire, place Amélie Raba Léon, 33076 Bordeaux, France
| | - Vanja Sisirak
- Université de Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux, France; CNRS-UMR 5164, ImmunoConcEpt, Université de Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux, France
| | - Estibaliz Lazaro
- Service de Médecine Interne, FHU ACRONIM, Hôpital Haut Lévêque, Centre Hospitalier Universitaire, Avenue Magellan, 33600 Pessac, France; Université de Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux, France; CNRS-UMR 5164, ImmunoConcEpt, Université de Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux, France
| | - Marie-Elise Truchetet
- Université de Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux, France; CNRS-UMR 5164, ImmunoConcEpt, Université de Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux, France; Service de Rhumatologie, FHU ACRONIM, Hôpital Pellegrin, Centre Hospitalier Universitaire, place Amélie Raba Léon, 33076 Bordeaux, France
| | - Christophe Richez
- Université de Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux, France; CNRS-UMR 5164, ImmunoConcEpt, Université de Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux, France; Service de Rhumatologie, FHU ACRONIM, Hôpital Pellegrin, Centre Hospitalier Universitaire, place Amélie Raba Léon, 33076 Bordeaux, France
| | - Patrick Legembre
- CLCC Eugène Marquis, INSERM-UMR 1242, Université de Rennes, rue Bataille Flandres Dunkerque, 35042 Rennes, France; Equipe Ligue Contre Le Cancer, rue Bataille Flandres Dunkerque, 35042 Rennes, France
| | - Patrick Blanco
- Laboratoire d'Immunologie et Immunogénétique, FHU ACRONIM, Hôpital Pellegrin, Centre Hospitalier Universitaire, place Amélie Raba Léon, 33076 Bordeaux, France; Université de Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux, France; CNRS-UMR 5164, ImmunoConcEpt, Université de Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux, France.
| |
Collapse
|
77
|
Kors S, Geijtenbeek K, Reits E, Schipper-Krom S. Regulation of Proteasome Activity by (Post-)transcriptional Mechanisms. Front Mol Biosci 2019; 6:48. [PMID: 31380390 PMCID: PMC6646590 DOI: 10.3389/fmolb.2019.00048] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 06/11/2019] [Indexed: 12/23/2022] Open
Abstract
Intracellular protein synthesis, folding, and degradation are tightly controlled processes to ensure proper protein homeostasis. The proteasome is responsible for the degradation of the majority of intracellular proteins, which are often targeted for degradation via polyubiquitination. However, the degradation rate of proteins is also affected by the capacity of proteasomes to recognize and degrade these substrate proteins. This capacity is regulated by a variety of proteasome modulations including (1) changes in complex composition, (2) post-translational modifications, and (3) altered transcription of proteasomal subunits and activators. Various diseases are linked to proteasome modulation and altered proteasome function. A better understanding of these modulations may offer new perspectives for therapeutic intervention. Here we present an overview of these three proteasome modulating mechanisms to give better insight into the diversity of proteasomes.
Collapse
Affiliation(s)
- Suzan Kors
- Department of Medical Biology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Karlijne Geijtenbeek
- Department of Medical Biology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Eric Reits
- Department of Medical Biology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Sabine Schipper-Krom
- Department of Medical Biology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
78
|
Jiao Y, Yang S, Cao Y, Zheng Z, Deng Y, Wang Q, Huang R, Du X. Genome and transcriptome analyses providing insight into the immune response of pearl oysters after allograft and xenograft transplantations. FISH & SHELLFISH IMMUNOLOGY 2019; 90:109-117. [PMID: 31051240 DOI: 10.1016/j.fsi.2019.04.061] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 04/19/2019] [Accepted: 04/24/2019] [Indexed: 06/09/2023]
Abstract
The immune response after allograft or xenograft transplantation in the pearl oyster is a major factor that cause its nucleus rejection and death. To determine the mechanism underlying the immune response after allograft and xenograft transplantations in the pearl oyster Pinctada fucata martensii, we constructed two sets of transcriptomes of hemocytes at different times (6 and 12 h; 1, 3, 6, 12, and 30 d) after allograft and xenograft transplantations, in which the xenografted mantle tissue was from Pinctada maxima. The transcriptomic analysis reveals many genes are involved in the immune response to transplantation, such as transient receptor potential cation channel (TRP), calmodulin (CaM), DNA replication-related genes, and sugar and lipid metabolism-related genes. The expression of these identified genes was higher in the host pearl oyster transplanted with xenograft than that by allograft. The histological analysis of the pearl sac also confirmed that many hemocytes were still gathered around the transplanted nucleus, and no pearl sac was formed in the host pearl oysters at 30 d after xenograft transplantation. The genomic analysis indicated that pearl oysters evolved many copies of genes, such as TRP, CaM, and GST, to sense and cope with the immune response after transplantation. "Ribosome" and "Cytosolic DNA-sensing pathway" were specifically induced in the xenograft group, whereas "Notch signaling pathway" specifically responded to the allograft transplantation. These results can improve our understanding of the mechanism underlying the immune response of pearl oysters after allograft and xenograft transplantations.
Collapse
Affiliation(s)
- Yu Jiao
- Fishery College, Guangdong Ocean University, Zhanjiang, 524025, China
| | - Shuai Yang
- Fishery College, Guangdong Ocean University, Zhanjiang, 524025, China
| | - Yanfei Cao
- Fishery College, Guangdong Ocean University, Zhanjiang, 524025, China
| | - Zhe Zheng
- Fishery College, Guangdong Ocean University, Zhanjiang, 524025, China
| | - Yuewen Deng
- Fishery College, Guangdong Ocean University, Zhanjiang, 524025, China
| | - Qingheng Wang
- Fishery College, Guangdong Ocean University, Zhanjiang, 524025, China
| | - Ronglian Huang
- Fishery College, Guangdong Ocean University, Zhanjiang, 524025, China
| | - Xiaodong Du
- Fishery College, Guangdong Ocean University, Zhanjiang, 524025, China.
| |
Collapse
|
79
|
Wang N, Chen M, Wang T. Liposomes used as a vaccine adjuvant-delivery system: From basics to clinical immunization. J Control Release 2019; 303:130-150. [PMID: 31022431 PMCID: PMC7111479 DOI: 10.1016/j.jconrel.2019.04.025] [Citation(s) in RCA: 176] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 04/17/2019] [Accepted: 04/17/2019] [Indexed: 12/14/2022]
Abstract
Liposomes are widely utilized as a carrier to improve therapeutic efficacy of agents thanks to their merits of high loading capacity, targeting delivery, reliable protection of agents, good biocompatibility, versatile structure modification and adjustable characteristics, such as size, surface charge, membrane flexibility and the agent loading mode. In particular, in recent years, through modification with immunopotentiators and targeting molecules, and in combination with innovative immunization devices, liposomes are rapidly developed as a multifunctional vaccine adjuvant-delivery system (VADS) that has a high capability in inducing desired immunoresponses, as they can target immune cells and even cellular organelles, engender lysosome escape, and promote Ag cross-presentation, thus enormously enhancing vaccination efficacy. Moreover, after decades of development, several products developed on liposome VADS have already been authorized for clinical immunization and are showing great advantages over conventional vaccines. This article describes in depth some critical issues relevant to the development of liposomes as a VADS, including principles underlying immunization, physicochemical properties of liposomes as the immunity-influencing factors, functional material modification to enhance immunostimulatory functions, the state-of-the-art liposome VADSs, as well as the marketed vaccines based on a liposome VADS. Therefore, this article provides a comprehensive reference to the development of novel liposome vaccines.
Collapse
Affiliation(s)
- Ning Wang
- School of Food and Bioengineering, Hefei University of Technology, 193 Tun Brook Road, Hefei, Anhui Province 230009, China
| | - Minnan Chen
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province 230032, China
| | - Ting Wang
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province 230032, China.
| |
Collapse
|
80
|
Buneeva OA, Medvedev AE. [Ubiquitin-independent protein degradation in proteasomes]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2019; 64:134-148. [PMID: 29723144 DOI: 10.18097/pbmc20186402134] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Proteasomes are large supramolecular protein complexes present in all prokaryotic and eukaryotic cells, where they perform targeted degradation of intracellular proteins. Until recently, it was generally accepted that prior proteolytic degradation in proteasomes the proteins had to be targeted by ubiquitination: the ATP-dependent addition of (typically four sequential) residues of the low-molecular ubiquitin protein, involving the ubiquitin-activating enzyme, ubiquitin-conjugating enzyme and ubiquitin ligase. The cytoplasm and nucleoplasm proteins labeled in this way are then digested in 26S proteasomes. However, in recent years it has become increasingly clear that using this route the cell eliminates only a part of unwanted proteins. Many proteins can be cleaved by the 20S proteasome in an ATP-independent manner and without previous ubiquitination. Ubiquitin-independent protein degradation in proteasomes is a relatively new area of studies of the role of the ubiquitin-proteasome system. However, recent data obtained in this direction already correct existing concepts about proteasomal degradation of proteins and its regulation. Ubiquitin-independent proteasome degradation needs the main structural precondition in proteins: the presence of unstructured regions in the amino acid sequences that provide interaction with the proteasome. Taking into consideration that in humans almost half of all genes encode proteins that contain a certain proportion of intrinsically disordered regions, it appears that the list of proteins undergoing ubiquitin-independent degradation will demonstrate further increase. Since 26S of proteasomes account for only 30% of the total proteasome content in mammalian cells, most of the proteasomes exist in the form of 20S complexes. The latter suggests that ubiquitin-independent proteolysis performed by the 20S proteasome is a natural process of removing damaged proteins from the cell and maintaining a constant level of intrinsically disordered proteins. In this case, the functional overload of proteasomes in aging and/or other types of pathological processes, if it is not accompanied by triggering more radical mechanisms for the elimination of damaged proteins, organelles and whole cells, has the most serious consequences for the whole organism.
Collapse
Affiliation(s)
- O A Buneeva
- Institute of Biomedical Chemistry, Moscow, Russia
| | - A E Medvedev
- Institute of Biomedical Chemistry, Moscow, Russia
| |
Collapse
|
81
|
Santambrogio L, Berendam SJ, Engelhard VH. The Antigen Processing and Presentation Machinery in Lymphatic Endothelial Cells. Front Immunol 2019; 10:1033. [PMID: 31134089 PMCID: PMC6513971 DOI: 10.3389/fimmu.2019.01033] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 04/23/2019] [Indexed: 12/24/2022] Open
Abstract
Until a few years ago, lymphatic vessels and lymphatic endothelial cells (LEC) were viewed as part of a passive conduit for lymph and immune cells to reach lymph nodes (LN). However, recent work has shown that LEC are active immunological players whose interaction with dendritic cells and T cells is of important immunomodulatory relevance. While the immunological interaction between LEC and other immune cells has taken a center stage, molecular analysis of LEC antigen processing and presentation machinery is still lagging. Herein we review the current knowledge of LEC MHC I and MHC II antigen processing and presentation pathways, Including the role of LEC in antigen phagocytosis, classical, and non-classical MHC II presentation, proteasome processing and MHC I presentation, and cross-presentation. The ultimate goal is to provide an overview of the LEC antigen processing and presentation machinery that constitutes the molecular basis for their role in MHC I and MHC II-restricted immune responses.
Collapse
Affiliation(s)
- Laura Santambrogio
- Department of Pathology, Microbiology and Immunology, Albert Einstein College of Medicine, New York, NY, United States
| | - Stella J Berendam
- Department of Microbiology, Immunology and Cancer Biology, Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Victor H Engelhard
- Department of Microbiology, Immunology and Cancer Biology, Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| |
Collapse
|
82
|
Limanaqi F, Biagioni F, Gaglione A, Busceti CL, Fornai F. A Sentinel in the Crosstalk Between the Nervous and Immune System: The (Immuno)-Proteasome. Front Immunol 2019; 10:628. [PMID: 30984192 PMCID: PMC6450179 DOI: 10.3389/fimmu.2019.00628] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 03/08/2019] [Indexed: 12/20/2022] Open
Abstract
The wealth of recent evidence about a bi-directional communication between nerve- and immune- cells revolutionized the traditional concept about the brain as an “immune-privileged” organ while opening novel avenues in the pathophysiology of CNS disorders. In fact, altered communication between the immune and nervous system is emerging as a common hallmark in neuro-developmental, neurodegenerative, and neuro-immunological diseases. At molecular level, the ubiquitin proteasome machinery operates as a sentinel at the crossroad between the immune system and brain. In fact, the standard proteasome and its alternative/inducible counterpart, the immunoproteasome, operate dynamically and coordinately in both nerve- and immune- cells to modulate neurotransmission, oxidative/inflammatory stress response, and immunity. When dysregulations of the proteasome system occur, altered amounts of standard- vs. immune-proteasome subtypes translate into altered communication between neurons, glia, and immune cells. This contributes to neuro-inflammatory pathology in a variety of neurological disorders encompassing Parkinson's, Alzheimer's, and Huntingtin's diseases, brain trauma, epilepsy, and Multiple Sclerosis. In the present review, we analyze those proteasome-dependent molecular interactions which sustain communication between neurons, glia, and brain circulating T-lymphocytes both in baseline and pathological conditions. The evidence here discussed converges in that upregulation of immunoproteasome to the detriment of the standard proteasome, is commonly implicated in the inflammatory- and immune- biology of neurodegeneration. These concepts may foster additional studies investigating the role of immunoproteasome as a potential target in neurodegenerative and neuro-immunological disorders.
Collapse
Affiliation(s)
- Fiona Limanaqi
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | | | | | | | - Francesco Fornai
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy.,I.R.C.C.S Neuromed, Pozzilli, Italy
| |
Collapse
|
83
|
Sun C, Mo M, Wang Y, Yu W, Song C, Wang X, Chen S, Liu Y. Activation of the immunoproteasome protects SH-SY5Y cells from the toxicity of rotenone. Neurotoxicology 2019; 73:112-119. [PMID: 30904435 DOI: 10.1016/j.neuro.2019.03.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 03/13/2019] [Accepted: 03/18/2019] [Indexed: 02/06/2023]
Abstract
This study investigated the expression and role of immunoproteasome (i-proteasome) in a cell model of Parkinson's disease (PD). The cytotoxicity of rotenone was measured by CCK-8 assay. The i-proteasome β1i subunit PSMB9 was suppressed by a specific shRNA or transfected with an overexpression plasmid in the SH-SY5Y cells. Under the exposure to rotenone or not, the expression of constitutive proteasome β subunits, i-proteasome βi subunits, antigen presentation related proteins, α-syn and TH were detected by Western blot in PSMB9-silenced or -overexpressed cells, and the proteasomal activities were detected by fluorogenic peptide substrates. The location of i-proteasome βi subunits and α-syn were detected by immunofluorescence staining. The levels of ROS, GSH and MDA were measured by commercial kits. Cell apoptosis was detected by flow cytometry. Besides impairing the constitutive proteasomes, rotenone induced the expression of βi subunits of i-proteasome and antigen presentation related proteins such as TAP1, TAP2 and MHC-I. Silencing or overexpressing PSMB9 had no obvious effect on the levels of other subunits, but could regulate the chymotrypsin-like activity of 20S proteasome and the expression of TAP1, TAP2 and MHC-I. Three βi subunits (PSMB9, PSMB10, PSMB8) of i-proteasome were all co-localized with α-syn. PSMB9 knockdown aggravated accumulation of α-syn, degradation of TH, release of ROS, increased level of MDA, decreased level of GSH and eventually promoted apoptosis in SH-SY5Y cells after rotenone treatment, while over-expression of PSMB9 could attenuate these toxic effects of rotenone. I-proteasome is activated in SH-SY5Y cells treated with rotenone and may play a neuroprotective role.
Collapse
Affiliation(s)
- Congcong Sun
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Mingshu Mo
- Department of Neurology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Yun Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221000, China
| | - Wenfei Yu
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Chengyuan Song
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Xingbang Wang
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Si Chen
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Yiming Liu
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, 250012, China; Brain Science Research Institute, Shandong University, Jinan, 250012, China.
| |
Collapse
|
84
|
Hungria VTDM, Crusoé EDQ, Bittencourt RI, Maiolino A, Magalhães RJP, Sobrinho JDN, Pinto JV, Fortes RC, Moreira EDS, Tanaka PY. New proteasome inhibitors in the treatment of multiple myeloma. Hematol Transfus Cell Ther 2019; 41:76-83. [PMID: 30793108 PMCID: PMC6371737 DOI: 10.1016/j.htct.2018.07.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 07/05/2018] [Indexed: 02/08/2023] Open
Abstract
The treatment of patients with relapsed and/or refractory multiple myeloma has improved considerably in the last 15 years, after the introduction of proteasome inhibitors and immunomodulatory drugs. The first clinical trials with new proteasome inhibitors have produced exciting results, particularly those comparing triplet regimens with standard doublet regimens, with a gain in progression-free survival accompanied by an acceptable safety profile and either similar or better health-related quality of life. New proteasome inhibitors hold the potential to fill unmet needs in multiple myeloma management regarding improvement of clinical outcomes, including delayed progression of disease in high-risk patients. This review summarizes the main pharmacological properties and clinical outcomes of these agents, and discusses their potential to change the whole multiple myeloma therapeutic landscape.
Collapse
Affiliation(s)
| | - Edvan de Queiroz Crusoé
- Universidade Federal da Bahia, Hospital Universitário Professor Edgar Santos, Serviço de Hematologia, Salvador, BA, Brazil
| | | | - Angelo Maiolino
- Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | | | | | | | | | | |
Collapse
|
85
|
Momose H, Sasaki E, Kuramitsu M, Hamaguchi I, Mizukami T. Gene expression profiling toward the next generation safety control of influenza vaccines and adjuvants in Japan. Vaccine 2018; 36:6449-6455. [PMID: 30243500 DOI: 10.1016/j.vaccine.2018.09.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 08/16/2018] [Accepted: 08/17/2018] [Indexed: 10/28/2022]
Abstract
Influenza becomes epidemic worldwide every year, and many individuals receive vaccination annually. Quality control relating to safety and potency of influenza vaccines is important to maintain public confidence. The safety of influenza vaccines has been assessed by clinical trials, and animal safety tests are performed to monitor the consistent quality between vaccines used for clinical trials and marketing; the biological responses in vaccinated animals are evaluated, including changes in body weight and white blood cell count. Animal safety tests have been contributing to the quality relating to the safety of influenza vaccines for decades, but improvements are needed. Although precise mechanisms involving biological changes in animal safety tests have not been fully elucidated, the application of cDNA microarray technology make it possible to reliably identify genes related to biological responses in vaccinated animals. From analysis of the expression profile of >10,000 genes of lung in animals treated with an inactivated whole virion influenza vaccine, we identified 17 marker genes whose expression patterns correlated well to changes in body weight and leukocyte count in vaccinated animals. In influenza HA vaccine-treated animals exhibiting subtle changes in biological responses, a robust expression pattern of marker genes was found. Furthermore, these marker genes could also be used in the evaluation of adjuvanted influenza vaccines. The expression profile of marker genes is expected to be an alternative indicator for safety control of various influenza vaccines conferring high sensitivity and short turnaround time. Thus, gene expression profiling may be a powerful tool for safety control of vaccines in the future.
Collapse
Affiliation(s)
- Haruka Momose
- Department of Safety Research on Blood and Biological Products, National Institute of Infectious Diseases, 4-7-1 Gakuen, Musashimurayama, Tokyo 208-0011, Japan
| | - Eita Sasaki
- Department of Safety Research on Blood and Biological Products, National Institute of Infectious Diseases, 4-7-1 Gakuen, Musashimurayama, Tokyo 208-0011, Japan
| | - Madoka Kuramitsu
- Department of Safety Research on Blood and Biological Products, National Institute of Infectious Diseases, 4-7-1 Gakuen, Musashimurayama, Tokyo 208-0011, Japan
| | - Isao Hamaguchi
- Department of Safety Research on Blood and Biological Products, National Institute of Infectious Diseases, 4-7-1 Gakuen, Musashimurayama, Tokyo 208-0011, Japan
| | - Takuo Mizukami
- Department of Safety Research on Blood and Biological Products, National Institute of Infectious Diseases, 4-7-1 Gakuen, Musashimurayama, Tokyo 208-0011, Japan.
| |
Collapse
|
86
|
Bastola P, Oien DB, Cooley M, Chien J. Emerging Cancer Therapeutic Targets in Protein Homeostasis. AAPS JOURNAL 2018; 20:94. [PMID: 30151644 DOI: 10.1208/s12248-018-0254-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 08/10/2018] [Indexed: 12/11/2022]
Abstract
Genomic aberrations inside malignant cells through copy number alterations, aneuploidy, and mutations can exacerbate misfolded and unfolded protein burden resulting in increased proteotoxic stress. Increased proteotoxic stress can be deleterious to malignant cells; therefore, these cells rely heavily on the protein quality control mechanisms for survival and proliferation. Components of the protein quality control, such as the unfolded protein response, heat shock proteins, autophagy, and the ubiquitin proteasome system, orchestrate a cascade of downstream events that allow the mitigation of the proteotoxic stress. This dependency makes components of the protein quality control mechanisms attractive targets in cancer therapeutics. In this review, we explore the components of the protein homeostasis especially focusing on the emerging cancer therapeutic agents/targets that are being actively pursued actively.
Collapse
Affiliation(s)
- Prabhakar Bastola
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud NE, Albuquerque, NM, 87131, USA.,Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, 66130, USA
| | - Derek B Oien
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud NE, Albuquerque, NM, 87131, USA
| | - Megan Cooley
- Methods Development, Small Molecules, PRA Health Sciences, Lenexa, KS, 66215, USA
| | - Jeremy Chien
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud NE, Albuquerque, NM, 87131, USA.
| |
Collapse
|
87
|
Buneeva OA, Medvedev AE. Ubiquitin-Independent Degradation of Proteins in Proteasomes. BIOCHEMISTRY (MOSCOW), SUPPLEMENT SERIES B: BIOMEDICAL CHEMISTRY 2018. [DOI: 10.1134/s1990750818030022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
88
|
Ogorevc E, Schiffrer ES, Sosič I, Gobec S. A patent review of immunoproteasome inhibitors. Expert Opin Ther Pat 2018; 28:517-540. [PMID: 29865878 DOI: 10.1080/13543776.2018.1484904] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION The ubiquitin-proteasome system is responsible for maintaining protein homeostasis and regulating a variety of cellular processes. The constitutive proteasome is expressed in all cells while the immunoproteasome (IP) is predominantly found in cells of hematopoietic origin. In other cells, the expression of IP can be induced under the influence of cytokines released by T cells during acute immune and stress responses. Inhibitors of IP are of significant interest, because it is expected that selective inhibition of the IP would cause fewer adverse effects. AREAS COVERED There is a considerable interest on patenting IP-specific inhibitors. Relevant patents and patent applications disclosing IP inhibitors are summarized and divided into two parts according to the chemical characteristics of compounds. We also briefly report on the biochemical methods used in the patents to profile the characteristics of IP inhibitors. EXPERT OPINION Several selective inhibitors of IP with a promising ability to address autoimmune and inflammatory diseases are being developed. Peptidic compounds are prevalent and the most advanced IP-selective compounds to date, ONX-0914 and KZR-616, are tripeptide epoxyketone-based molecules. However, some patents disclose that IP-selective inhibition is possible with compounds possessing non-peptidic scaffolds indicating countless possibilities to address inhibition of IP in the future.
Collapse
Affiliation(s)
- Eva Ogorevc
- a Faculty of Pharmacy , University of Ljubljana , Ljubljana , Slovenia
| | | | - Izidor Sosič
- a Faculty of Pharmacy , University of Ljubljana , Ljubljana , Slovenia
| | - Stanislav Gobec
- a Faculty of Pharmacy , University of Ljubljana , Ljubljana , Slovenia
| |
Collapse
|
89
|
Honda-Ozaki F, Terashima M, Niwa A, Saiki N, Kawasaki Y, Ito H, Hotta A, Nagahashi A, Igura K, Asaka I, Li HL, Yanagimachi M, Furukawa F, Kanazawa N, Nakahata T, Saito MK. Pluripotent Stem Cell Model of Nakajo-Nishimura Syndrome Untangles Proinflammatory Pathways Mediated by Oxidative Stress. Stem Cell Reports 2018; 10:1835-1850. [PMID: 29731430 PMCID: PMC5989695 DOI: 10.1016/j.stemcr.2018.04.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 04/04/2018] [Accepted: 04/05/2018] [Indexed: 01/10/2023] Open
Abstract
Nakajo-Nishimura syndrome (NNS) is an immunoproteasome-associated autoinflammatory disorder caused by a mutation of the PSMB8 gene. Although dysfunction of the immunoproteasome causes various cellular stresses attributed to the overproduction of inflammatory cytokines and chemokines in NNS, the underlying mechanisms of the autoinflammation are still largely unknown. To investigate and understand the mechanisms and signal pathways in NNS, we established a panel of isogenic pluripotent stem cell (PSC) lines with PSMB8 mutation. Activity of the immunoproteasome in PSMB8-mutant PSC-derived myeloid cell lines (MT-MLs) was reduced even without stimulation compared with non-mutant-MLs. In addition, MT-MLs showed an overproduction of inflammatory cytokines and chemokines, with elevated reactive oxygen species (ROS) and phosphorylated p38 MAPK levels. Treatment with p38 MAPK inhibitor and antioxidants decreased the abnormal production of cytokines and chemokines. The current PSC model revealed a specific ROS-mediated inflammatory pathway, providing a platform for the discovery of alternative therapeutic options for NNS and related immunoproteasome disorders. An isogenic PSC panel for Nakajo-Nishimura syndrome was prepared Mutant myeloid cell lines showed increased proinflammatory response p38 MAPK inhibitor and antioxidant treatment restored the proinflammatory phenotype
Collapse
Affiliation(s)
- Fumiko Honda-Ozaki
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Madoka Terashima
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Akira Niwa
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Norikazu Saiki
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Yuri Kawasaki
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Haruna Ito
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Akitsu Hotta
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Ayako Nagahashi
- Department of Fundamental Cell Technology, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Koichi Igura
- Department of Fundamental Cell Technology, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Isao Asaka
- Department of Fundamental Cell Technology, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Hongmei Lisa Li
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan; Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Masakatsu Yanagimachi
- Department of Pediatrics, Tokyo Medical and Dental University, Tokyo 113-8519, Japan
| | - Fukumi Furukawa
- Department of Dermatology, Wakayama Medical University, Wakayama 641-0012, Japan
| | - Nobuo Kanazawa
- Department of Dermatology, Wakayama Medical University, Wakayama 641-0012, Japan.
| | - Tatsutoshi Nakahata
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Megumu K Saito
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan.
| |
Collapse
|
90
|
Mishra R, Upadhyay A, Prajapati VK, Mishra A. Proteasome-mediated proteostasis: Novel medicinal and pharmacological strategies for diseases. Med Res Rev 2018; 38:1916-1973. [DOI: 10.1002/med.21502] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Revised: 03/13/2018] [Accepted: 04/04/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Ribhav Mishra
- Cellular and Molecular Neurobiology Unit; Indian Institute of Technology Jodhpur; Rajasthan India
| | - Arun Upadhyay
- Cellular and Molecular Neurobiology Unit; Indian Institute of Technology Jodhpur; Rajasthan India
| | - Vijay Kumar Prajapati
- Department of Biochemistry; School of Life Sciences; Central University of Rajasthan; Rajasthan India
| | - Amit Mishra
- Cellular and Molecular Neurobiology Unit; Indian Institute of Technology Jodhpur; Rajasthan India
| |
Collapse
|
91
|
Correale P, Botta C, Staropoli N, Nardone V, Pastina P, Ulivieri C, Gandolfo C, Baldari TC, Lazzi S, Ciliberto D, Giannicola R, Fioravanti A, Giordano A, Zappavigna S, Caraglia M, Tassone P, Pirtoli L, Cusi MG, Tagliaferri P. Systemic inflammatory status predict the outcome of k-RAS WT metastatic colorectal cancer patients receiving the thymidylate synthase poly-epitope-peptide anticancer vaccine. Oncotarget 2018; 9:20539-20554. [PMID: 29755670 PMCID: PMC5945541 DOI: 10.18632/oncotarget.24993] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 02/21/2018] [Indexed: 12/19/2022] Open
Abstract
TSPP is an anticancer poly-epitope peptide vaccine to thymidylate synthase, recently investigated in the multi-arm phase Ib TSPP/VAC1 trial. TSPP vaccination induced immune-biological effects and showed antitumor activity in metastatic colorectal cancer (mCRC) patients and other malignancies. Progression-free and overall survival of 41 mCRC patients enrolled in the study correlated with baseline levels of CEA, immune-inflammatory markers (neutrophil/lymphocyte ratio, CRP, ESR, LDH, ENA), IL-4 and with post-treatment change in p-ANCA and CD56dimCD16brightNKs (p < 0.04). A subset of 19 patients with activating k-ras mutations showed a different immune-inflammatory response to TSPP as compared to patients with k-ras/wt and a worse outcome in term of PFS (p = 0.048). In patients with k-ras/mut, inflammatory markers lost their predictive value and their survival directly correlated with the baseline levels of IL17/A over the median value (p = 0.01). These results provide strong hints for the design of further clinical trials aimed to test TSPP vaccination in mCRC patients.
Collapse
Affiliation(s)
- Pierpaolo Correale
- Unit of Medical Oncology, Grand Metropolitan Hospital Bianchi Melacrino Morelli, Reggio-Calabria, Italy
| | - Cirino Botta
- Medical Oncology Unit, AUO Mater Domini, Magna Graecia University, Catanzaro, Italy
| | - Nicoletta Staropoli
- Department of Experimental and Clinical Medicine, Magna Graecia University , Catanzaro, Italy
| | - Valerio Nardone
- Unit of Radiotherapy, Department of Surgery, Medicine and Neurological Science, Siena University Hospital, Siena, Italy
| | - Pierpaolo Pastina
- Unit of Radiotherapy, Department of Surgery, Medicine and Neurological Science, Siena University Hospital, Siena, Italy
| | | | - Claudia Gandolfo
- Microbiology and Virology Unit, Department of Medical Biotechnology, Siena University, Siena, Italy
| | | | - Stefano Lazzi
- Unit of Pathology, Department of Surgery, Medicine and Neurological Science, Siena University Hospital, Siena, Italy
| | - Domenico Ciliberto
- Department of Experimental and Clinical Medicine, Magna Graecia University , Catanzaro, Italy
| | - Rocco Giannicola
- Unit of Medical Oncology, Grand Metropolitan Hospital Bianchi Melacrino Morelli, Reggio-Calabria, Italy
| | - Antonella Fioravanti
- Unit of Rheumatology, Department of Clinical Medicine and Immunologic Sciences, University of Siena, Siena, Italy
| | - Antonio Giordano
- Department of Biotechnology, Temple University, Sbarro Foundation, Philadelphia, Pennsylvania, USA
| | - Silvia Zappavigna
- Department of Precision Medicine, University of Campania L. Vanvitelli, Naples, Italy
| | - Michele Caraglia
- Department of Biotechnology, Temple University, Sbarro Foundation, Philadelphia, Pennsylvania, USA.,Department of Precision Medicine, University of Campania L. Vanvitelli, Naples, Italy
| | - Pierfrancesco Tassone
- Medical Oncology Unit, AUO Mater Domini, Magna Graecia University, Catanzaro, Italy.,Department of Experimental and Clinical Medicine, Magna Graecia University , Catanzaro, Italy.,Department of Precision Medicine, University of Campania L. Vanvitelli, Naples, Italy
| | - Luigi Pirtoli
- Unit of Radiotherapy, Department of Surgery, Medicine and Neurological Science, Siena University Hospital, Siena, Italy
| | - Maria Grazia Cusi
- Microbiology and Virology Unit, Department of Medical Biotechnology, Siena University, Siena, Italy
| | - Pierosandro Tagliaferri
- Department of Experimental and Clinical Medicine, Magna Graecia University , Catanzaro, Italy
| |
Collapse
|
92
|
Chen Y, Zhang Y, Guo X. Proteasome dysregulation in human cancer: implications for clinical therapies. Cancer Metastasis Rev 2018; 36:703-716. [PMID: 29039081 DOI: 10.1007/s10555-017-9704-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Cancer cells show heightened dependency on the proteasome for their survival, growth, and spread. Proteasome dysregulation is therefore commonly selected in favor of the development of many types of cancer. The vast abnormalities in a cancer cell, on top of the complexity of the proteasome itself, have enabled a plethora of mechanisms gearing the proteasome to the oncogenic process. Here, we use selected examples to highlight some general mechanisms underlying proteasome dysregulation in cancer, including copy number variations, transcriptional control, epigenetic regulation, and post-translational modifications. Research in this field has greatly advanced our understanding of proteasome regulation and will shed new light on proteasome-based combination therapies for cancer treatment.
Collapse
Affiliation(s)
- Yulin Chen
- Life Sciences Institute of Zhejiang University, 866 Yuhangtang Rd, Hangzhou, 310058, China
| | - Yanan Zhang
- Life Sciences Institute of Zhejiang University, 866 Yuhangtang Rd, Hangzhou, 310058, China
| | - Xing Guo
- Life Sciences Institute of Zhejiang University, 866 Yuhangtang Rd, Hangzhou, 310058, China.
| |
Collapse
|
93
|
Oliva J. Proteasome and Organs Ischemia-Reperfusion Injury. Int J Mol Sci 2017; 19:ijms19010106. [PMID: 29301204 PMCID: PMC5796056 DOI: 10.3390/ijms19010106] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 12/12/2017] [Accepted: 12/27/2017] [Indexed: 12/17/2022] Open
Abstract
The treatment of organ failure on patients requires the transplantation of functional organs, from donors. Over time, the methodology of transplantation was improved by the development of organ preservation solutions. The storage of organs in preservation solutions is followed by the ischemia of the organ, resulting in a shortage of oxygen and nutrients, which damage the tissues. When the organ is ready for the transplantation, the reperfusion of the organ induces an increase of the oxidative stress, endoplasmic reticulum stress, and inflammation which causes tissue damage, resulting in a decrease of the transplantation success. However, the addition of proteasome inhibitor in the preservation solution alleviated the injuries due to the ischemia-reperfusion process. The proteasome is a protein structure involved in the regulation the inflammation and the clearance of damaged proteins. The goal of this review is to summarize the role of the proteasome and pharmacological compounds that regulate the proteasome in protecting the organs from the ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Joan Oliva
- Department of Medicine, LA BioMed at Harbor UCLA Medical Center, Torrance, CA 90502, USA.
| |
Collapse
|
94
|
Liu Q, Zhang YL, Hu SP, Ma ZL, Gao SL, Sun B, Xiao F, Zhang Z, Cai XH, He XJ. Expression of immunoproteasome subunits in the porcine lung: Alterations during normal and inflammatory conditions. Vet Microbiol 2017; 210:134-141. [DOI: 10.1016/j.vetmic.2017.07.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Revised: 07/21/2017] [Accepted: 07/21/2017] [Indexed: 01/30/2023]
|
95
|
Abstract
The ubiquitin proteasome pathway was discovered in the 1980s to be a central component of the cellular protein-degradation machinery with essential functions in homeostasis, which include preventing the accumulation of misfolded or deleterious proteins. Cancer cells produce proteins that promote both cell survival and proliferation, and/or inhibit mechanisms of cell death. This notion set the stage for preclinical testing of proteasome inhibitors as a means to shift this fine equilibrium towards cell death. Since the late 1990s, clinical trials have been conducted for a variety of malignancies, leading to regulatory approvals of proteasome inhibitors to treat multiple myeloma and mantle-cell lymphoma. First-generation and second-generation proteasome inhibitors can elicit deep initial responses in patients with myeloma, for whom these drugs have dramatically improved outcomes, but relapses are frequent and acquired resistance to treatment eventually emerges. In addition, promising preclinical data obtained with proteasome inhibitors in models of solid tumours have not been confirmed in the clinic, indicating the importance of primary resistance. Investigation of the mechanisms of resistance is, therefore, essential to further maximize the utility of this class of drugs in the era of personalized medicine. Herein, we discuss the advances and challenges resulting from the introduction of proteasome inhibitors into the clinic.
Collapse
Affiliation(s)
- Elisabet E Manasanch
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 429, Houston, Texas 77030-4009, USA
| | - Robert Z Orlowski
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 429, Houston, Texas 77030-4009, USA
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 429, Houston, Texas 77030-4009, USA
| |
Collapse
|
96
|
Broekaart DWM, van Scheppingen J, Geijtenbeek KW, Zuidberg MRJ, Anink JJ, Baayen JC, Mühlebner A, Aronica E, Gorter JA, van Vliet EA. Increased expression of (immuno)proteasome subunits during epileptogenesis is attenuated by inhibition of the mammalian target of rapamycin pathway. Epilepsia 2017. [DOI: 10.1111/epi.13823] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Diede W. M. Broekaart
- Department of (Neuro)Pathology; Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| | - Jackelien van Scheppingen
- Department of (Neuro)Pathology; Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| | - Karlijne W. Geijtenbeek
- Department of (Neuro)Pathology; Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| | - Mark R. J. Zuidberg
- Department of (Neuro)Pathology; Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| | - Jasper J. Anink
- Department of (Neuro)Pathology; Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| | - Johannes C. Baayen
- Department of Neurosurgery; VU University Medical Center; Vrije Universiteit; Amsterdam The Netherlands
| | - Angelika Mühlebner
- Department of (Neuro)Pathology; Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| | - Eleonora Aronica
- Department of (Neuro)Pathology; Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
- Swammerdam Institute for Life Sciences; Center for Neuroscience; University of Amsterdam; Amsterdam The Netherlands
- Stichting Epilepsie Instellingen Nederland (SEIN); Heemstede The Netherlands
| | - Jan A. Gorter
- Swammerdam Institute for Life Sciences; Center for Neuroscience; University of Amsterdam; Amsterdam The Netherlands
| | - Erwin A. van Vliet
- Department of (Neuro)Pathology; Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| |
Collapse
|
97
|
Wong YY, Johnson B, Friedrich TC, Trepanier LA. Hepatic expression profiles in retroviral infection: relevance to drug hypersensitivity risk. Pharmacol Res Perspect 2017; 5:e00312. [PMID: 28603631 PMCID: PMC5464341 DOI: 10.1002/prp2.312] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 03/13/2017] [Indexed: 12/11/2022] Open
Abstract
HIV‐infected patients show a markedly increased risk of delayed hypersensitivity (HS) reactions to potentiated sulfonamide antibiotics (trimethoprim/sulfamethoxazole or TMP/SMX). Some studies have suggested altered SMX biotransformation in HIV infection, but hepatic biotransformation pathways have not been evaluated directly. Systemic lupus erythematosus (SLE) is another chronic inflammatory disease with a higher incidence of sulfonamide HS, but it is unclear whether retroviral infection and SLE share risk factors for drug HS. We hypothesized that retroviral infection would lead to dysregulation of hepatic pathways of SMX biotransformation, as well as pathway alterations in common with SLE that could contribute to drug HS risk. We characterized hepatic expression profiles and enzymatic activities in an SIV‐infected macaque model of retroviral infection, and found no evidence for dysregulation of sulfonamide drug biotransformation pathways. Specifically, NAT1,NAT2,CYP2C8,CYP2C9,CYB5R3,MARC1/2, and glutathione‐related genes (GCLC,GCLM,GSS,GSTM1, and GSTP1) were not differentially expressed in drug naïve SIVmac239‐infected male macaques compared to age‐matched controls, and activities for SMX N‐acetylation and SMX hydroxylamine reduction were not different. However, multiple genes that are reportedly over‐expressed in SLE patients were also up‐regulated in retroviral infection, to include enhanced immunoproteasomal processing and presentation of antigens as well as up‐regulation of gene clusters that may be permissive to autoimmunity. These findings support the hypothesis that pathways downstream from drug biotransformation may be primarily important in drug HS risk in HIV infection.
Collapse
Affiliation(s)
- Yat Yee Wong
- Department of Medical Sciences School of Veterinary Medicine Madison Wisconsin
| | - Brian Johnson
- Molecular and Environmental Toxicology Center School of Medicine and Public Health University of Wisconsin-Madison Madison Wisconsin
| | - Thomas C Friedrich
- Department of Pathobiological Sciences School of Veterinary Medicine Madison Wisconsin.,AIDS Vaccine Research Laboratory Wisconsin National Primate Research Center Madison Wisconsin
| | - Lauren A Trepanier
- Department of Medical Sciences School of Veterinary Medicine Madison Wisconsin
| |
Collapse
|
98
|
Abstract
In most patients with chronic heart failure (HF), levels of circulating cytokines are elevated and the elevated cytokine levels correlate with the severity of HF and prognosis. Various stresses induce subcellular component abnormalities, such as mitochondrial damage. Damaged mitochondria induce accumulation of reactive oxygen species and apoptogenic proteins, and subcellular inflammation. The vicious cycle of subcellular component abnormalities, inflammatory cell infiltration and neurohumoral activation induces cardiomyocyte injury and death, and cardiac fibrosis, resulting in cardiac dysfunction and HF. Quality control mechanisms at both the protein and organelle levels, such as elimination of apoptogenic proteins and damaged mitochondria, maintain cellular homeostasis. An imbalance between protein synthesis and degradation is likely to result in cellular dysfunction and disease. Three major protein degradation systems have been identified, namely the cysteine protease system, autophagy, and the ubiquitin proteasome system. Autophagy was initially believed to be a non-selective process. However, recent studies have described the process of selective mitochondrial autophagy, known as mitophagy. Elimination of damaged mitochondria by autophagy is important for maintenance of cellular homeostasis. DNA and RNA degradation systems also play a critical role in regulating inflammation and maintaining cellular homeostasis mediated by damaged DNA clearance and post-transcriptional regulation, respectively. This review discusses some recent advances in understanding the role of sterile inflammation and degradation systems in HF.
Collapse
Affiliation(s)
- Kazuhiko Nishida
- Cardiovascular Division, King's College London British Heart Foundation Centre of Excellence
| | - Kinya Otsu
- Cardiovascular Division, King's College London British Heart Foundation Centre of Excellence
| |
Collapse
|
99
|
Bader El Din NG, Farouk S, El-Shenawy R, Elhady MM, Ibrahim MK, Dawood RM, Salem AM, El Awady MK. The Synergistic Effect of TNFα -308 G/A and TGFβ1 -509 C/T Polymorphisms on Hepatic Fibrosis Progression in Hepatitis C Virus Genotype 4 Patients. Viral Immunol 2017; 30:127-135. [PMID: 28151059 DOI: 10.1089/vim.2016.0083] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Tumor necrosis factor-alpha (TNFα) and transforming growth factor-beta (TGFβ1) cytokines are highly implicated in liver fibrosis. Polymorphisms in these cytokines affect their expression, secretion, and activity. This study aimed to evaluate the influence of TNFα -308 G/A and TGFβ1 -509 C/T polymorphism on hepatic fibrosis progression in Egyptian patients with hepatitis C virus (HCV) genotype 4. Genotyping of TNFα -308 G/A and TGFβ1 -509 C/T was performed using polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) analysis in 122 subjects (50 healthy controls and 72 HCV patients). Also, serum TNFα and TGFβ1 levels were detected by enzyme-linked immunosorbent assay (ELISA). The genotyping results of early (F0-F1, n = 36) and late (F2-F4, n = 36) HCV fibrosis patients showed that late fibrosis patients had higher TNFα -308 AA genotype and TGFβ1 -509 TT genotype than early fibrosis patients (p = 0.016, 0.028, respectively). Moreover, the TNFα and TGFβ1 serum levels were significantly higher in HCV patients with TNFα A containing genotypes (GA+AA) (p = 0.004) and patients with TGFβ1 T containing genotypes (CT+TT) (p = 0.001), respectively. The combined unfavorable TNFα (GA/AA) and TGFβ1 (CT/TT) genotypes were highly associated with abnormal liver function parameters and were significantly higher in high activity (A2-A3) and late fibrosis (F2-F4) HCV patients (p = 0.023, 0.029). The multivariate analysis results confirmed that the combined TNFα-308 (AA) and TGFβ1 -509 (TT) unfavorable genotypes increased the risk of hepatic fibrosis progression by 6.4-fold than combined favorable genotypes (odds ratio: 6.417, 95% confidence interval [1.490-27.641], p = 0.013). In conclusion, both TNFα -308 G/A and TGFβ1 -509 C/T polymorphisms synergistically influence the hepatic fibrosis progression and can be used as potential biomarkers to predict hepatic disease progression in chronic hepatitis C patients.
Collapse
Affiliation(s)
- Noha G Bader El Din
- 1 Department of Microbial Biotechnology, National Research Centre , Giza, Egypt
| | - Sally Farouk
- 1 Department of Microbial Biotechnology, National Research Centre , Giza, Egypt
| | - Reem El-Shenawy
- 1 Department of Microbial Biotechnology, National Research Centre , Giza, Egypt
| | - Mostafa M Elhady
- 2 Department of Biochemistry, Faculty of Science, Ain Shams University , Cairo, Egypt
| | - Marwa K Ibrahim
- 1 Department of Microbial Biotechnology, National Research Centre , Giza, Egypt
| | - Reham M Dawood
- 1 Department of Microbial Biotechnology, National Research Centre , Giza, Egypt
| | - Ahmed M Salem
- 2 Department of Biochemistry, Faculty of Science, Ain Shams University , Cairo, Egypt
| | - Mostafa K El Awady
- 1 Department of Microbial Biotechnology, National Research Centre , Giza, Egypt
| |
Collapse
|
100
|
Liu H, Wan C, Ding Y, Han R, He Y, Xiao J, Hao J. PR-957, a selective inhibitor of immunoproteasome subunit low-MW polypeptide 7, attenuates experimental autoimmune neuritis by suppressing T h17-cell differentiation and regulating cytokine production. FASEB J 2017; 31:1756-1766. [PMID: 28096232 DOI: 10.1096/fj.201601147r] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 01/03/2017] [Indexed: 12/23/2022]
Abstract
Experimental autoimmune neuritis (EAN) is a CD4+ T-cell-mediated autoimmune inflammatory demyelinating disease of the peripheral nervous system. It has been replicated in an animal model of human inflammatory demyelinating polyradiculoneuropathy, Guillain-Barré syndrome. In this study, we evaluated the therapeutic efficacy of a selective inhibitor of the immunoproteasome subunit, low-MW polypeptide 7 (PR-957) in rats with EAN. Our results showed that PR-957 significantly delayed onset day, reduced severity and shortened duration of EAN, and alleviated demyelination and inflammatory infiltration in sciatic nerves. In addition to significantly regulating expression of the cytokine profile, PR-957 treatment down-regulated the proportion of proinflammatory T-helper (Th)17 cells in sciatic nerves and spleens of rats with EAN. Data presented show the role of PR-957 in the signal transducer and activator of transcription 3 (STAT3) pathway. PR-957 not only decreased expression of IL-6 and IL-23 but also led to down-regulation of STAT3 phosphorylation in CD4+ T cells. Regulation of the STAT3 pathway led to a reduction in retinoid-related orphan nuclear receptor γ t and IL-17 production. Furthermore, reduction of STAT3 phosphorylation may have directly suppressed Th17-cell differentiation. Therefore, our study demonstrates that PR-957 could potently alleviate inflammation in rats with EAN and that it may be a likely candidate for treating Guillain-Barré syndrome.-Liu, H., Wan, C., Ding, Y., Han, R., He, Y., Xiao, J., Hao, J. PR-957, a selective inhibitor of immunoproteasome subunit low-MW polypeptide 7, attenuates experimental autoimmune neuritis by suppressing Th17-cell differentiation and regulating cytokine production.
Collapse
Affiliation(s)
- Haijie Liu
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China.,Department of Physical Medicine and Rehabilitation, Tianjin Medical University General Hospital, Tianjin, China
| | - Chunxiao Wan
- Department of Physical Medicine and Rehabilitation, Tianjin Medical University General Hospital, Tianjin, China
| | - Yanan Ding
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Ranran Han
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Yating He
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Jinting Xiao
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Junwei Hao
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China;
| |
Collapse
|