1
|
Anton DB, de Lima JC, Dahmer BR, Camini AM, Goettert MI, Timmers LFSM. Taming the storm: potential anti-inflammatory compounds targeting SARS-CoV-2 MPro. Inflammopharmacology 2024:10.1007/s10787-024-01525-9. [PMID: 39048773 DOI: 10.1007/s10787-024-01525-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 07/05/2024] [Indexed: 07/27/2024]
Abstract
In severe COVID-19 cases, an exacerbated inflammatory response triggers a cytokine storm that can worsen the prognosis. Compounds with both antiviral and anti-inflammatory activities show promise as candidates for COVID-19 therapy, as they potentially act against the SARS-CoV-2 infection regardless of the disease stage. One of the most attractive drug targets among coronaviruses is the main protease (MPro). This enzyme is crucial for cleaving polyproteins into non-structural proteins required for viral replication. The aim of this review was to identify SARS-CoV-2 MPro inhibitors with both antiviral and anti-inflammatory properties. The interactions of the compounds within the SARS-CoV-2 MPro binding site were analyzed through molecular docking when data from crystallographic structures were unavailable. 18 compounds were selected and classified into five different superclasses. Five of them exhibit high potency against MPro: GC-376, baicalein, naringenin, heparin, and carmofur, with IC50 values below 0.2 μM. The MPro inhibitors selected have the potential to alleviate lung edema and decrease cytokine release. These molecules mainly target three critical inflammatory pathways: NF-κB, JAK/STAT, and MAPK, all previously associated with COVID-19 pathogenesis. The structures of the compounds occupy the S1/S2 substrate binding subsite of the MPro. They interact with residues from the catalytic dyad (His41 and Cys145) and/or with the oxyanion hole (Gly143, Ser144, and Cys145), which are pivotal for substrate recognition. The MPro SARS-CoV-2 inhibitors with potential anti-inflammatory activities present here could be optimized for maximum efficacy and safety and be explored as potential treatment of both mild and severe COVID-19.
Collapse
Affiliation(s)
- Débora Bublitz Anton
- Biotechnology Graduate Program, Universidade do Vale do Taquari (Univates), Lajeado, CEP 95914-014, Brazil
| | - Jeferson Camargo de Lima
- Biotechnology Graduate Program, Universidade do Vale do Taquari (Univates), Lajeado, CEP 95914-014, Brazil
| | - Bruno Rampanelli Dahmer
- Biotechnology Graduate Program, Universidade do Vale do Taquari (Univates), Lajeado, CEP 95914-014, Brazil
| | - Ana Micaela Camini
- Biotechnology Graduate Program, Universidade do Vale do Taquari (Univates), Lajeado, CEP 95914-014, Brazil
| | - Marcia Inês Goettert
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmaceutical Sciences, Eberhard Karls Universität Tübingen, 72076, Tübingen, Germany
| | - Luis Fernando Saraiva Macedo Timmers
- Biotechnology Graduate Program, Universidade do Vale do Taquari (Univates), Lajeado, CEP 95914-014, Brazil.
- Medical Science Graduate Program, Universidade do Vale do Taquari (Univates), Lajeado, CEP 95914-014, Brazil.
| |
Collapse
|
2
|
Chen X, Chen Y, Ou Y, Min W, Liang S, Hua L, Zhou Y, Zhang C, Chen P, Yang Z, Hu W, Sun P. Bortezomib inhibits NLRP3 inflammasome activation and NF-κB pathway to reduce psoriatic inflammation. Biochem Pharmacol 2022; 206:115326. [DOI: 10.1016/j.bcp.2022.115326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 10/19/2022] [Accepted: 10/19/2022] [Indexed: 11/02/2022]
|
3
|
Chu HS, Peterson C, Jun A, Foster J. Targeting the integrated stress response in ophthalmology. Curr Eye Res 2021; 46:1075-1088. [PMID: 33474991 DOI: 10.1080/02713683.2020.1867748] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Purpose: To summarize the Integrated Stress Response (ISR) in the context of ophthalmology, with special interest on the cornea and anterior segment. Results: The ISR is a powerful and conserved signaling pathway that allows for cells to respond to a diverse array of both intracellular and extracellular stressors. The pathway is classically responsible for coordination of the cellular response to amino acid starvation, ultraviolet light, heme dysregulation, viral infection, and unfolded protein. Under normal circumstances, it is considered pro-survival and a necessary mechanism through which protein translation is controlled. However, in cases of severe or prolonged stress the pathway can promote apoptosis, and loss of normal cellular phenotype. The activation of this pathway culminates in the global inhibition of cap-dependent protein translation and the canonical expression of the activating transcription factor 4 (ATF4). Conclusion:The eye is uniquely exposed to ISR responsive stressors due to its environmental exposure and relative isolation from the circulatory system which are necessary for its function. We will discuss how this pathway is critical for the proper function of the tissue, its role in development, as well as how targeting of the pathway could alleviate key aspects of diverse ophthalmic diseases.
Collapse
Affiliation(s)
- Hsiao-Sang Chu
- Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins University, Baltimore, MD, USA.,Department of Ophthalmology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei City, Taiwan.,Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei City, Taiwan
| | - Cornelia Peterson
- Department of Molecular & Comparative Pathobiology, Johns Hopkins University, Baltimore, MD, USA
| | - Albert Jun
- Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins University, Baltimore, MD, USA
| | - James Foster
- Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
4
|
Protective Role of mTOR in Liver Ischemia/Reperfusion Injury: Involvement of Inflammation and Autophagy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:7861290. [PMID: 31827701 PMCID: PMC6885218 DOI: 10.1155/2019/7861290] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 08/24/2019] [Accepted: 10/14/2019] [Indexed: 02/06/2023]
Abstract
Liver ischemia/reperfusion (IR) injury is a common phenomenon after liver resection and transplantation, which often results in liver graft dysfunction such as delayed graft function and primary nonfunction. The mammalian target of rapamycin (mTOR) is an evolutionarily highly conserved serine/threonine protein kinase, which coordinates cell growth and metabolism through sensing environmental inputs under physiological or pathological conditions, involved in the pathophysiological process of IR injury. In this review, we mainly present current evidence of the beneficial role of mTOR in modulating inflammation and autophagy under liver IR to provide some evidence for the potential therapies for liver IR injury.
Collapse
|
5
|
Retina transduction by rAAV2 after intravitreal injection: comparison between mouse and rat. Gene Ther 2019; 26:479-490. [PMID: 31562387 DOI: 10.1038/s41434-019-0100-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 09/04/2019] [Accepted: 09/09/2019] [Indexed: 12/11/2022]
Abstract
Adeno-associated virus vectors (rAAV) are currently the most common vehicle used in clinical trials of retinal gene therapy, usually delivered through subretinal injections to target cells of the outer retina. However, targeting the inner retina requires intravitreal injections, a simple and safe procedure, which is effective for transducing the rodent retina, but still of low efficiency in the eyes of primates. We investigated whether adjuvant pharmacological agents may enhance rAAV transduction of the retinas of mouse and rat after intravitreal delivery. Tyrosine kinase inhibitors were highly efficient in mice, especially imatinib and genistein, and promoted transduction even of the outer retina. In rats, however, we report that they were not effective. Even with direct proteasomal inhibition in rats, the effects upon transduction were only minimal and restricted to the inner retina. Even tyrosine capsid mutant rAAVs in rats had a transduction profile similar to wtAAV. Thus, the differences between mouse and rat, in both eye size and the inner limiting membrane, compromise the efficiency of AAV vectors penetration from the vitreous into the retina, and impact the efficacy of strategies developed to enhance intravitreal retinal rAAV transduction. Further improvement of strategies, then are required.
Collapse
|
6
|
Cheng Z, Combs M, Zhu Q, Xia P, Opheim Z, Parker J, Mack CP, Taylor JM. Genome-Wide RNAi Screen Identifies Regulators of Cardiomyocyte Necrosis. ACS Pharmacol Transl Sci 2019; 2:361-371. [PMID: 32259070 DOI: 10.1021/acsptsci.9b00052] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Indexed: 12/22/2022]
Abstract
Regulation of cellular death is central to nearly all physiological routines and is dysregulated in virtually all diseases. Cell death occurs by two major processes, necrosis which culminates in a pervasive inflammatory response and apoptosis which is largely immunologically inert. As necrosis has long been considered an accidental, unregulated form of cellular death that occurred in response to a harsh environmental stimulus, it was largely ignored as a clinical target. However, recent elegant studies suggest that certain forms of necrosis can be reprogrammed. However, scant little is known about the molecules and pathways that orchestrate calcium-overload-induced necrosis, a main mediator of ischemia/reperfusion (IR)-induced cardiomyocyte cell death. To rectify this critical gap in our knowledge, we performed a novel genome-wide siRNA screen to identify modulators of calcium-induced necrosis in human muscle cells. Our screen identified multiple molecular circuitries that either enhance or inhibit this process, including lysosomal calcium channel TPCN1, mitophagy mediatorTOMM7, Ran-binding protein RanBP9, Histone deacetylase HDAC2, chemokine CCL11, and the Arp2/3 complex regulator glia maturation factor-γ (GMFG). Notably, a number of druggable enzymes were identified, including the proteasome β5 subunit (encoded by PSMB5 gene), which controls the proteasomal chymotrypsin-like peptidase activity. Such findings open up the possibility for the discovery of pharmacological interventions that could provide therapeutic benefits to patients affected by myriad disorders characterized by excessive (or too little) necrotic cell loss, including but not limited to IR injury in the heart and kidney, chronic neurodegenerative disorders, muscular dystrophies, sepsis, and cancers.
Collapse
Affiliation(s)
- Zhaokang Cheng
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington 99210, United States
| | - Matthew Combs
- Department of Pathology, Department of Genetics, Lineberger Cancer Center, and McAllister Heart Institute, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Qiang Zhu
- Department of Pathology, Department of Genetics, Lineberger Cancer Center, and McAllister Heart Institute, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Peng Xia
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington 99210, United States
| | - Zachary Opheim
- Department of Pathology, Department of Genetics, Lineberger Cancer Center, and McAllister Heart Institute, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Joel Parker
- Department of Pathology, Department of Genetics, Lineberger Cancer Center, and McAllister Heart Institute, University of North Carolina, Chapel Hill, North Carolina 27599, United States.,Department of Pathology, Department of Genetics, Lineberger Cancer Center, and McAllister Heart Institute, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Christopher P Mack
- Department of Pathology, Department of Genetics, Lineberger Cancer Center, and McAllister Heart Institute, University of North Carolina, Chapel Hill, North Carolina 27599, United States.,Department of Pathology, Department of Genetics, Lineberger Cancer Center, and McAllister Heart Institute, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Joan M Taylor
- Department of Pathology, Department of Genetics, Lineberger Cancer Center, and McAllister Heart Institute, University of North Carolina, Chapel Hill, North Carolina 27599, United States.,Department of Pathology, Department of Genetics, Lineberger Cancer Center, and McAllister Heart Institute, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
7
|
Cellular Responses to Proteasome Inhibition: Molecular Mechanisms and Beyond. Int J Mol Sci 2019; 20:ijms20143379. [PMID: 31295808 PMCID: PMC6678303 DOI: 10.3390/ijms20143379] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 06/26/2019] [Accepted: 07/01/2019] [Indexed: 02/06/2023] Open
Abstract
Proteasome inhibitors have been actively tested as potential anticancer drugs and in the treatment of inflammatory and autoimmune diseases. Unfortunately, cells adapt to survive in the presence of proteasome inhibitors activating a variety of cell responses that explain why these therapies have not fulfilled their expected results. In addition, all proteasome inhibitors tested and approved by the FDA have caused a variety of side effects in humans. Here, we describe the different types of proteasome complexes found within cells and the variety of regulators proteins that can modulate their activities, including those that are upregulated in the context of inflammatory processes. We also summarize the adaptive cellular responses activated during proteasome inhibition with special emphasis on the activation of the Autophagic-Lysosomal Pathway (ALP), proteaphagy, p62/SQSTM1 enriched-inclusion bodies, and proteasome biogenesis dependent on Nrf1 and Nrf2 transcription factors. Moreover, we discuss the role of IRE1 and PERK sensors in ALP activation during ER stress and the involvement of two deubiquitinases, Rpn11 and USP14, in these processes. Finally, we discuss the aspects that should be currently considered in the development of novel strategies that use proteasome activity as a therapeutic target for the treatment of human diseases.
Collapse
|
8
|
Kageyama M, Ota T, Sasaoka M, Katsuta O, Shinomiya K. Chemical proteasome inhibition as a novel animal model of inner retinal degeneration in rats. PLoS One 2019; 14:e0217945. [PMID: 31150519 PMCID: PMC6544319 DOI: 10.1371/journal.pone.0217945] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 05/21/2019] [Indexed: 12/14/2022] Open
Abstract
Chemical proteasome inhibition has been a valuable animal model of neurodegeneration to uncover roles for the ubiquitin-proteasome system in the central nervous system. However, little is known about the effects of chemical proteasome inhibitors on retinal integrity. Therefore, we characterized the effects of structurally different chemical proteasome inhibitors on the retinal morphology and the mechanisms of their action in the normal adult rat eyes. Intravitreal injection of MG-262 and other proteasome inhibitors led to inner retinal degeneration. MG-262-induced inner retinal degeneration was accompanied by reduced proteasome activity, increased poly-ubiquitinated protein levels, and increased positive immunostaining of ubiquitin, 20S proteasome subunit and GADD153/CHOP in the retina. Its retinal degenerative effect was also associated with reduced retinal neurofilament light chain gene expression, reflecting retinal ganglion cell death. MG-262-induced neurofilament light chain downregulation was largely resistant to pharmacological modulation including endoplasmic reticulum stress, apoptosis or MAP kinase inhibitors. Thus, this study provides further evidence of roles for the ubiquitin-proteasome system in the maintenance of the retinal structural integrity. Chemical proteasome inhibition may be used as a novel animal model of inner retinal degeneration, including retinal ganglion cell loss, which warrants further analysis of the molecular mechanisms underlying its retinal degenerative effect.
Collapse
Affiliation(s)
- Masaaki Kageyama
- Global Alliances and External Research, Santen Pharmaceutical Co., Ltd., Nara, Japan
- * E-mail:
| | - Takashi Ota
- Global Alliances and External Research, Santen Pharmaceutical Co., Ltd., Nara, Japan
| | - Masaaki Sasaoka
- Global Alliances and External Research, Santen Pharmaceutical Co., Ltd., Nara, Japan
| | - Osamu Katsuta
- Research and Development Center, Santen Pharmaceutical Co., Ltd., Nara, Japan
| | - Katsuhiko Shinomiya
- Research and Development Center, Santen Pharmaceutical Co., Ltd., Nara, Japan
| |
Collapse
|
9
|
Wu X, Chen Z, Yang Y, Dong Y, Liu H, Kuang S, Luo K. Impact of proteasome inhibitor MG-132 on expression of NF-κB, IL-1β and histological remodeling after myocardial infarction. Exp Ther Med 2018; 16:1365-1372. [PMID: 30112065 DOI: 10.3892/etm.2018.6308] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 09/01/2017] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to investigate the impact of carbobenzoxy-Leu-Leu-leucinal (MG-132) on myocardial remodeling in rats with myocardial infarction (MI) and investigate the possible underlying mechanisms. The rat model of MI was established, followed by administration of MG-132 (MG group), pyrrolidine dithiocarbamic acid (PDTC group) or normal saline (MI group) for 28 days. The expression of nuclear factor-κB (NF-κB) p65, interleukin 1β (IL-1β) and matrix metalloproteinase 2 (MMP-2), as well as the total volume of collagen and the ratio of type I/III collagen were then detected. Total collagen, including type I and III collagen, and the ratio of type I/III collagen were significantly increased in MI rats compared with those in the sham group (P<0.01), while it was significantly decreased in the PDTC and MG groups compared with that in the MI group (P<0.01). A similar trend was identified for the expression of NF-κB, IL-1β and MMP-2, which was significantly increased in the MI group compared with that in the sham group (P<0.01), while it was significantly decreased in the MG and PDTC groups compared with that in the MI group (P<0.01). In conclusion, MG-132 was demonstrated to improve post-MI tissue remodeling, and the mechanism may be associated with the inhibition of NF-κB activation and the downregulation of inflammatory cytokines, such as IL-1β.
Collapse
Affiliation(s)
- Xinhua Wu
- Department of Cardiology, The First Affiliated Hospital of Dali University, Dali, Yunnan 671000, P.R. China
| | - Zhangrong Chen
- Department of Cardiology, The First Affiliated Hospital of Dali University, Dali, Yunnan 671000, P.R. China
| | - Ying Yang
- Department of Cardiology, The First Affiliated Hospital of Dali University, Dali, Yunnan 671000, P.R. China
| | - Yu Dong
- Department of Cardiology, The First Affiliated Hospital of Dali University, Dali, Yunnan 671000, P.R. China
| | - Hong Liu
- Department of Cardiology, The First Affiliated Hospital of Dali University, Dali, Yunnan 671000, P.R. China
| | - Shiquan Kuang
- Department of Cardiology, The First Affiliated Hospital of Dali University, Dali, Yunnan 671000, P.R. China
| | - Kailiang Luo
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 404100, P.R. China
| |
Collapse
|
10
|
Moon K, Lee HG, Baek WK, Lee Y, Kim KS, Jun JH, Kim JY, Joo CK. Bortezomib inhibits proliferation, migration, and TGF-β1-induced epithelial-mesenchymal transition of RPE cells. Mol Vis 2017; 23:1029-1038. [PMID: 29386876 PMCID: PMC5757857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 12/27/2017] [Indexed: 11/22/2022] Open
Abstract
PURPOSE Nuclear factor kappa B (NF-κB) plays an important role in the epithelial-mesenchymal transition (EMT) of RPE cells. We investigated the effects of a proteasome inhibitor, bortezomib, on the EMT in RPE cells. In addition, we assessed the influence of bortezomib on regulation of the NF-κB pathway during this process. METHODS After treatment with various concentrations of bortezomib, cell viability was analyzed with the water-soluble tetrazolium salt-8 assay, cell-cycle regulation was evaluated with flow cytometry, and cell migration was monitored with in vitro wound healing and Transwell migration assays. To induce fibroblastoid transformation, the RPE cells were treated with recombinant human transforming growth factor (TGF)-β1 (10 ng/ml), and western blot and immunocytochemical analyses were performed to evaluate altered expression of EMT markers after treatment with bortezomib. To verify the effect of bortezomib on shrinkage by myofibroblastic transformation, a contraction assay of the RPE-collagen gel lattice was performed. RESULTS Treatment with bortezomib decreased RPE viability in a dose-dependent manner, and flow cytometry revealed that these effects were due to arrest of the G2/M phase cell-cycle. In the in vitro wound healing and Transwell migration assays, treatment with 20 nM bortezomib significantly impeded RPE migration. Treatment with bortezomib also significantly inhibited TGF-β1-induced transdifferentiation of the RPE cells. The effects on proliferation, migration, and the EMT were mediated by regulation of the NF-κB signaling pathway. In addition, bortezomib inhibited contraction of the RPE-collagen gel lattices. CONCLUSIONS Bortezomib inhibits myofibroblastic transformation of RPE cells by downregulating NF-κB expression and prevents contraction of the RPE-collagen gel matrix. Thus, bortezomib represents a candidate putative therapeutic agent for management of retinal fibrotic diseases.
Collapse
Affiliation(s)
- Kun Moon
- Balgeunsesang Eye clinic, Seoul, South Korea
| | - Hyun-Gyo Lee
- Department of Ophthalmology, Keimyung University School of Medicine, Dongsan Medical Center, Daegu, South Korea
| | - Won-Ki Baek
- Institute for Cancer Research, Keimyung University, Dongsan Medical Center, Daegu, South Korea,Department of Microbiology, Keimyung University School of Medicine, Daegu, South Korea
| | - Youngkyun Lee
- Department of Oral Biochemistry, School of Dentistry, IHBR, Kyungpook National University, Daegu, South Korea
| | - Kwang Soo Kim
- Department of Ophthalmology, Keimyung University School of Medicine, Dongsan Medical Center, Daegu, South Korea
| | - Jong Hwa Jun
- Department of Ophthalmology, Keimyung University School of Medicine, Dongsan Medical Center, Daegu, South Korea,Institute for Cancer Research, Keimyung University, Dongsan Medical Center, Daegu, South Korea
| | - Jae-Young Kim
- Department of Oral Biochemistry, School of Dentistry, IHBR, Kyungpook National University, Daegu, South Korea
| | - Choun-Ki Joo
- Department of Ophthalmology and Visual Science, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| |
Collapse
|
11
|
Abstract
PURPOSE OF REVIEW Sarcoidosis is a chronic granulomatous disease typically affecting the lung, lymph nodes, and other organ systems. Evidence suggests that the morbidity and mortality rates for sarcoidosis in the USA are rising, despite widespread use of anti-inflammatory therapies. In this review, we survey new therapies that target specific inflammatory pathways in other diseases (such as rheumatoid arthritis, Crohn's disease, and psoriasis) that are similar to pathways relevant to sarcoidosis immunopathogenesis, and therefore, represent potentially new sarcoidosis therapies. RECENT FINDINGS Immunopathogenesis of sarcoidosis has been well elucidated over the past few years. There is abundant evidence for T-cell activation in sarcoidosis leading to activation of both Th1 and Th17 inflammatory cascades. Therapies targeting T-cell activation, Th1 pathways (such as the interleukin-6 inhibitors), Th17 pathway mediators, and others have been Food and Drug Administration approved or under investigation to treat a variety of autoimmune inflammatory diseases, but have not been studied in sarcoidosis. Targeting the p38 mitogen-activated protein kinases and the ubiquitine proteasome system with new agents may also represent a novel therapeutic option for patients with sarcoidosis. SUMMARY Rising morbidity and mortality rates for patients with sarcoidosis strongly support the need to develop more effective anti-inflammatory therapies to treat chronic disease.
Collapse
|
12
|
The Protective Effects of αB-Crystallin on Ischemia-Reperfusion Injury in the Rat Retina. J Ophthalmol 2017; 2017:7205408. [PMID: 29098085 PMCID: PMC5643040 DOI: 10.1155/2017/7205408] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 07/28/2017] [Accepted: 08/06/2017] [Indexed: 12/13/2022] Open
Abstract
To investigate whether αB-crystallin protects against acute retinal ischemic reperfusion injury (I/R) and elucidate the potential antioxidant mechanisms. Retinal I/R injury was made by elevating the intraocular pressure (IOP) 110 mmHg for 60 min, and αB-crystallin (1 × 10−5 g/L) or vehicle solution was administered intravitreously immediately after I/R injury. The animal was sacrificed 24 h, 1 w, and 1 m after the I/R injury. The retina damage was detected by hematoxylin and eosin (HE) staining and electroretinography (ERG). The level of malondialdehyde (MDA), nitric oxide (NO), and the total superoxide dismutase (T-SOD) was determined. An immunohistochemical study was performed to detect the activation of inducible nitric oxide synthase (iNOS) and NF- (nuclear factor-) kappaB (NF-κB) p65. The decrease of retinal thickness and the number of retinal ganglion cells (RGCs) can be suppressed by αB-crystallin. And the amplitudes of a- and b-wave were remarkably greater without αB-crystallin. Similarly, αB-crystallin also significantly decreased the level of MDA and NO and enhanced the activities of T-SOD. The positive expression of iNOS and NF-kappaB p65 was obviously reduced while treated with αB-crystallin. αB-crystallin can inhibit the expression of NF-κB and its antioxidative effect to protect the retina from I/R injury.
Collapse
|
13
|
Moritz KE, McCormack NM, Abera MB, Viollet C, Yauger YJ, Sukumar G, Dalgard CL, Burnett BG. The role of the immunoproteasome in interferon-γ-mediated microglial activation. Sci Rep 2017; 7:9365. [PMID: 28839214 PMCID: PMC5571106 DOI: 10.1038/s41598-017-09715-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 07/14/2017] [Indexed: 11/12/2022] Open
Abstract
Microglia regulate the brain microenvironment by sensing damage and neutralizing potentially harmful insults. Disruption of central nervous system (CNS) homeostasis results in transition of microglia to a reactive state characterized by morphological changes and production of cytokines to prevent further damage to CNS tissue. Immunoproteasome levels are elevated in activated microglia in models of stroke, infection and traumatic brain injury, though the exact role of the immunoproteasome in neuropathology remains poorly defined. Using gene expression analysis and native gel electrophoresis we characterize the expression and assembly of the immunoproteasome in microglia following interferon-gamma exposure. Transcriptome analysis suggests that the immunoproteasome regulates multiple features of microglial activation including nitric oxide production and phagocytosis. We show that inhibiting the immunoproteasome attenuates expression of pro-inflammatory cytokines and suppresses interferon-gamma-dependent priming of microglia. These results imply that targeting immunoproteasome function following CNS injury may attenuate select microglial activity to improve the pathophysiology of neurodegenerative conditions or the progress of inflammation-mediated secondary injury following neurotrauma.
Collapse
Affiliation(s)
- Kasey E Moritz
- Neuroscience Program, Uniformed Services University of the Health Sciences, F. Edward Hébert School of Medicine, Bethesda, MD, USA
| | - Nikki M McCormack
- Neuroscience Program, Uniformed Services University of the Health Sciences, F. Edward Hébert School of Medicine, Bethesda, MD, USA
| | - Mahlet B Abera
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Coralie Viollet
- Collaborative Health Initiative Research Program, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Young J Yauger
- Neuroscience Program, Uniformed Services University of the Health Sciences, F. Edward Hébert School of Medicine, Bethesda, MD, USA
| | - Gauthaman Sukumar
- Collaborative Health Initiative Research Program, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Clifton L Dalgard
- Neuroscience Program, Uniformed Services University of the Health Sciences, F. Edward Hébert School of Medicine, Bethesda, MD, USA.,Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Collaborative Health Initiative Research Program, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,The American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Barrington G Burnett
- Neuroscience Program, Uniformed Services University of the Health Sciences, F. Edward Hébert School of Medicine, Bethesda, MD, USA. .,Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.
| |
Collapse
|
14
|
Burcham PC. Acrolein and Human Disease: Untangling the Knotty Exposure Scenarios Accompanying Several Diverse Disorders. Chem Res Toxicol 2016; 30:145-161. [DOI: 10.1021/acs.chemrestox.6b00310] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Philip C. Burcham
- Pharmacology, Pharmacy & Anaesthesiology Unit, School of Medicine and Pharmacology, The University of Western Australia, Crawley, Western Australia 6007, Australia
| |
Collapse
|
15
|
Witort E, Capaccioli S, Becatti M, Fiorillo C, Batignani G, Pavoni V, Piccini M, Orioli M, Carini M, Aldini G, Lulli M. Albumin Cys34 adducted by acrolein as a marker of oxidative stress in ischemia-reperfusion injury during hepatectomy. Free Radic Res 2016; 50:831-9. [PMID: 27089934 DOI: 10.1080/10715762.2016.1179736] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The aim of this study was to measure and identify the reactive carbonyl species (RCSs) released in the blood of humans subjected to hepatic resection. Pre-anesthesia malondialdehyde (MDA) plasma content (0.36 ± 0.11 nmol/mg protein) remained almost unchanged immediately after anaesthesia, before clamping and at the 10th min after ischemia, while markedly increased (to 0.59 ± 0.07 nmol/mg; p < 0.01, Tukey's post test) at the 10th min of reperfusion. A similar trend was observed for the protein carbonyls (PCs), whose pre-anesthesia levels (0.17 ± 0.13 nmol/mg) did not significantly change during ischemia, while increased more than fourfold at the 10th min of reperfusion (0.75 ± 0.17 nmol/mg; p < 0.01, Tukey's post test). RCSs were then identified as covalent adducts to the albumin Cys34, which we previously found as the most reactive protein nucleophilic site in plasma. By using a mass spectrometry (MS) approach based on precursor ion scanning, we found that acrolein (ACR) is the main RCS adducted to albumin Cys34. In basal conditions, the adducted albumin was 0.6 ± 0.4% of the native form but it increased by almost fourfold at the 10th min of reperfusion (2.3 ± 0.7%; p < 0.01, t-test analysis). Since RCSs are damaging molecules, we propose that RCSs, and ACR in particular, are new targets for novel molecular treatments aimed at reducing the ischemia/reperfusion damage by the use of RCS sequestering agents.
Collapse
Affiliation(s)
- Ewa Witort
- a Department of Experimental and Clinical Biomedical Sciences , University of Florence , Florence , Italy
| | - Sergio Capaccioli
- a Department of Experimental and Clinical Biomedical Sciences , University of Florence , Florence , Italy
| | - Matteo Becatti
- a Department of Experimental and Clinical Biomedical Sciences , University of Florence , Florence , Italy
| | - Claudia Fiorillo
- a Department of Experimental and Clinical Biomedical Sciences , University of Florence , Florence , Italy
| | - Giacomo Batignani
- b Department of Surgery and Translational Medicine , University of Florence , Florence , Italy
| | - Vittorio Pavoni
- c Department of Anesthesia and Intensive Care , University-Hospital Careggi , Florence , Italy
| | - Matteo Piccini
- a Department of Experimental and Clinical Biomedical Sciences , University of Florence , Florence , Italy
| | - Marica Orioli
- d Department of Pharmaceutical Sciences , University of Milan , Milan , Italy
| | - Marina Carini
- d Department of Pharmaceutical Sciences , University of Milan , Milan , Italy
| | - Giancarlo Aldini
- d Department of Pharmaceutical Sciences , University of Milan , Milan , Italy
| | - Matteo Lulli
- a Department of Experimental and Clinical Biomedical Sciences , University of Florence , Florence , Italy
| |
Collapse
|
16
|
Yeh PT, Huang HW, Yang CM, Yang WS, Yang CH. Astaxanthin Inhibits Expression of Retinal Oxidative Stress and Inflammatory Mediators in Streptozotocin-Induced Diabetic Rats. PLoS One 2016; 11:e0146438. [PMID: 26765843 PMCID: PMC4713224 DOI: 10.1371/journal.pone.0146438] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Accepted: 12/17/2015] [Indexed: 01/31/2023] Open
Abstract
PURPOSE We evaluated whether orally administered astaxanthin (AST) protects against oxidative damage in the ocular tissues of streptozotocin (STZ)-induced diabetic rats. METHODS AND RESULTS Fifty 6-week-old female Wistar rats were randomly assigned to receive an injection of STZ to induce diabetes (n = 40) or to remain uninduced (n = 10). The diabetic rats were randomly selected into four groups and they were separately administered normal saline, 0.6 mg/kg AST, 3 mg/kg AST, or 0.5 mg/kg lutein daily for eight weeks. Retinal functions of each group were evaluated by electroretinography. The expression of oxidative stress and inflammatory mediators in the ocular tissues was then assessed by immunohistochemistry, western blot analysis, ELISA, RT-PCR, and electrophoretic mobility shift assay (EMSA). Retinal functions were preserved by AST and lutein in different levels. Ocular tissues from AST- and lutein-treated rats had significantly reduced levels of oxidative stress mediators (8-hydroxy-2'-deoxyguanosine, nitrotyrosine, and acrolein) and inflammatory mediators (intercellular adhesion molecule-1, monocyte chemoattractant protein-1, and fractalkine), increased levels of antioxidant enzymes (heme oxygenase-1 and peroxiredoxin), and reduced activity of the transcription factor nuclear factor-kappaB (NF-κB). CONCLUSION The xanthophyll carotenoids AST and lutein have neuroprotective effects and reduce ocular oxidative stress, and inflammation in the STZ diabetic rat model, which may be mediated by downregulation of NF-κB activity.
Collapse
Affiliation(s)
- Po-Ting Yeh
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan
- Graduate Institute of Pathology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hsin-Wei Huang
- Department of Ophthalmology, Taipei Medical University-Wan Fang Hospital, Taipei, Taiwan
| | - Chung-May Yang
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan
- Department of Ophthalmology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Wei-Shiung Yang
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Chang-Hao Yang
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan
- Department of Ophthalmology, College of Medicine, National Taiwan University, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
17
|
Emoto Y, Yoshizawa K, Kinoshita Y, Yuki M, Yuri T, Tsubura A. Green tea extract attenuates MNU-induced photoreceptor cell apoptosis via suppression of heme oxygenase-1. J Toxicol Pathol 2015; 29:61-5. [PMID: 26989304 PMCID: PMC4766529 DOI: 10.1293/tox.2015-0052] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 09/16/2015] [Indexed: 12/12/2022] Open
Abstract
The effects of green tea extract (GTE) on N-methyl-N-nitrosourea (MNU)-induced photoreceptor cell apoptosis were examined, and the possible mechanisms of action of GTE were assessed. Alterations in the retinal morphological architecture were determined by hematoxylin-eosin staining, vimentin immunoreactivity, and photoreceptor cell apoptosis (TUNEL labeling). Expression of oxidant marker, heme oxygenase (HO)-1, mRNA levels in outer nuclear cells was assessed by laser capture microdissection (LCM). Sprague-Dawley rats were given 40 mg/kg MNU at 7 weeks of age in the absence and presence of 250 mg/kg GTE treatment (once daily from 3 days prior to MNU for a maximum 10 days). Although photoreceptor cell degeneration began 24 hr after MNU, the morphological effects of GTE at the time point were not definitive. However, GTE lowered TUNEL labeling and HO-1 mRNA expression. At 7 days after MNU, photoreceptor damage was attenuated by GTE treatment. Therefore, the ability of GTE to reduce MNU-induced photoreceptor cell apoptosis may be due to its antioxidant properties.
Collapse
Affiliation(s)
- Yuko Emoto
- Department of Pathology II, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka 573-1010, Japan
| | - Katsuhiko Yoshizawa
- Department of Pathology II, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka 573-1010, Japan
| | - Yuichi Kinoshita
- Department of Pathology II, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka 573-1010, Japan
| | - Michiko Yuki
- Department of Pathology II, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka 573-1010, Japan
| | - Takashi Yuri
- Department of Pathology II, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka 573-1010, Japan
| | - Airo Tsubura
- Department of Pathology II, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka 573-1010, Japan
| |
Collapse
|
18
|
Gu RP, Fu LL, Jiang CH, Xu YF, Wang X, Yu J. Retina Is Protected by Neuroserpin from Ischemic/Reperfusion-Induced Injury Independent of Tissue-Type Plasminogen Activator. PLoS One 2015; 10:e0130440. [PMID: 26176694 PMCID: PMC4503687 DOI: 10.1371/journal.pone.0130440] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Accepted: 05/20/2015] [Indexed: 11/18/2022] Open
Abstract
The purpose of the present study was to investigate the potential neuroprotective effect of neuroserpin (NSP) on acute retinal ischemic/reperfusion-induced (IR) injury. An IR injury model was established by elevating intraocular pressure (IOP) for 60 minutes in wild type and tPA-deficient (tPA-/-) mice. Prior to IR injury, 1 μL of 20 μmol/L NSP or an equal volume of bovine serum albumin (BSA) was intravitreally administered. Retinal function was evaluated by electroretinograph (ERG) and the number of apoptotic neurons was determined via TUNEL labeling. Caspase-3, -8, -9,poly (ADP-ribose) polymerase (PARP)and their cleaved forms were subsequently analyzed. It was found that IR injury significantly damaged retinal function, inducing apoptosis in the retina, while NSP attenuated the loss of retinal function and significantly reduced the number of apoptotic neurons in both wild type and tPA-/- mice. The levels of cleaved caspase-3, cleaved PARP (the substrate of caspase-3) and caspase-9 (the modulator of the caspase-3), which had increased following IR injury, were significantly inhibited by NSP in both wild type and tPA-/- mice. NSP increased ischemic tolerance in the retina at least partially by inhibiting the intrinsic cell death signaling pathway of caspase-3. It was therefore concluded that the protective effect of neuroserpin maybe independent from its canonical interaction with a tissue-type plasminogen activator.
Collapse
Affiliation(s)
- R. P. Gu
- Department of Ophthalmology and Vision Sciences and Key Laboratory of Myopia of State Health Ministry, Eye and ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, 200031, China
| | - L. L. Fu
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - C. H. Jiang
- Department of Ophthalmology and Vision Sciences and Key Laboratory of Myopia of State Health Ministry, Eye and ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, 200031, China
- Department of Ophthalmology, No. 5 people’s Hospital of Shanghai, Shanghai, 200240, China
| | - Y. F. Xu
- Department of Ophthalmology and Vision Sciences and Key Laboratory of Myopia of State Health Ministry, Eye and ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, 200031, China
| | - X. Wang
- Department of Ophthalmology and Vision Sciences and Key Laboratory of Myopia of State Health Ministry, Eye and ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, 200031, China
| | - J. Yu
- Department of Ophthalmology and Vision Sciences and Key Laboratory of Myopia of State Health Ministry, Eye and ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, 200031, China
| |
Collapse
|
19
|
Bian Y, Ren L, Wang L, Xu S, Tao J, Zhang X, Huang Y, Qian Y, Zhang X, Song Z, Wu W, Wang Y, Liang G. A novel imidazopyridine derivative, X22, prevents the retinal ischemia-reperfusion injury via inhibition of MAPKs. Exp Eye Res 2015; 135:26-36. [DOI: 10.1016/j.exer.2015.04.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2014] [Revised: 04/13/2015] [Accepted: 04/17/2015] [Indexed: 10/23/2022]
|
20
|
Yazici A, Aksit H, Sari ES, Yay A, Erken HA, Aksit D, Cakmak H, Seyrek K, Ermis SS. Comparison of pre-treatment and post-treatment use of selenium in retinal ischemia reperfusion injury. Int J Ophthalmol 2015; 8:263-8. [PMID: 25938038 DOI: 10.3980/j.issn.2222-3959.2015.02.09] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 08/27/2014] [Indexed: 11/02/2022] Open
Abstract
AIM To investigate the effects of selenium in rat retinal ischemia reperfusion (IR) model and compare pre-treatment and post-treatment use. METHODS Selenium pre-treatment group (n=8) was treated with intraperitoneal (i.p.) selenium 0.5 mg/kg for 7d and terminated 24h after the IR injury. Selenium post-treatment group (n=8) was treated with i.p. selenium 0.5 mg/kg for 7d after the IR injury with termination at the end of the 7d period. Sham group (n=8) received i.p. saline injections identical to the selenium volume for 7d with termination 24h after the IR injury. Control group (n=8) received no intervention. Main outcome measures were retina superoxide dismutase (SOD), glutathione (GSH), total antioxidant status (TAS), malondialdehyde (MDA), DNA fragmentation levels, and immunohistological apoptosis evaluation. RESULTS Compared to the Sham group, selenium pre-treatment had a statistical difference in all parameters except SOD. Post-treatment selenium also resulted in statistical differences in all parameters except the MDA levels. When comparing selenium groups, the pre-treatment selenium group had a statistically higher success in reduction of markers of cell damage such as MDA and DNA fragmentation. In contrast, the post-selenium treatment group had resulted in statistically higher levels of GSH. Histologically both selenium groups succeeded to limit retinal thickening and apoptosis. Pre-treatment use was statistically more successful in decreasing apoptosis in ganglion cell layer compared to post-treatment use. CONCLUSION Selenium was successful in retinal protection in IR injuries. Pre-treatment efficacy was superior in terms of prevention of tissue damage and apoptosis.
Collapse
Affiliation(s)
- Alper Yazici
- Department of Ophthalmology, Faculty of Medicine, Balikesir University, Balikesir 10010, Turkey
| | - Hasan Aksit
- Department of Biochemistry, Faculty of Veterinary, Balikesir University, Balikesir 10010, Turkey
| | - Esin Sogutlu Sari
- Department of Ophthalmology, Faculty of Medicine, Balikesir University, Balikesir 10010, Turkey
| | - Arzu Yay
- Department of Histology and Embryology, Faculty of Medicine, Erciyes University, Kayseri 38010, Turkey
| | - Haydar Ali Erken
- Department of Physiology, Faculty of Medicine, Balikesir University, Balikesir 10010, Turkey
| | - Dilek Aksit
- Department of Pharmacology, Faculty of Veterinary, Balikesir University, Balikesir 10010, Turkey
| | - Harun Cakmak
- Department of Ophthalmology, Faculty of Medicine, Adnan Menderes University, Aydin 09010, Turkey
| | - Kamil Seyrek
- Department of Biochemistry, Faculty of Medicine, Balikesir University, Balikesir 10010, Turkey
| | - Sitki Samet Ermis
- Department of Ophthalmology, Faculty of Medicine, Balikesir University, Balikesir 10010, Turkey
| |
Collapse
|
21
|
Tian H, Wang L, Cai R, Zheng L, Guo L. Identification of protein network alterations upon retinal ischemia-reperfusion injury by quantitative proteomics using a Rattus norvegicus model. PLoS One 2014; 9:e116453. [PMID: 25549249 PMCID: PMC4280217 DOI: 10.1371/journal.pone.0116453] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 12/08/2014] [Indexed: 01/15/2023] Open
Abstract
Retinal ischemia is a common feature associated with several ocular diseases, including diabetic retinopathy. In this study, we investigated the effect of a retinal ischemia and reperfusion (I/R) injury on protein levels via a quantitative shotgun strategy using stable isotope dimethyl labeling combined with LC-MS/MS analysis. Based on the relative quantitation data of 1088 proteins, 234 proteins showed a greater than 1.5-fold change following I/R injury, 194 of which were up-regulated and 40 were down-regulated. Gene ontology analysis revealed that after I/R injury, there was an increase in the metabolic-process related proteins but a decline in cell communication, system process and transport-related proteins. A ribosome protein network and a secreted protein network consisting of many protease inhibitors were identified among the up-regulated proteins, despite a suppression of the mammalian target of rapamycin (mTOR) pathway following the I/R injury. A synaptic-related protein network was found to be significantly down-regulated, implicating a functional reduction of neurons following a retinal I/R injury. Our results provide new systems-biology clues for the study of retinal ischemia.
Collapse
Affiliation(s)
- Han Tian
- College of Life Sciences, Wuhan University, Wuhan, China
| | - Leilei Wang
- College of Life Sciences, Wuhan University, Wuhan, China
| | - Ruiqi Cai
- College of Life Sciences, Wuhan University, Wuhan, China
| | - Ling Zheng
- College of Life Sciences, Wuhan University, Wuhan, China
- * E-mail: (LZ); (LG)
| | - Lin Guo
- College of Life Sciences, Wuhan University, Wuhan, China
- * E-mail: (LZ); (LG)
| |
Collapse
|
22
|
Hong S, Iizuka Y, Lee T, Kim CY, Seong GJ. Neuroprotective and neurite outgrowth effects of maltol on retinal ganglion cells under oxidative stress. Mol Vis 2014; 20:1456-62. [PMID: 25352751 PMCID: PMC4203576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 10/15/2014] [Indexed: 11/04/2022] Open
Abstract
PURPOSE To evaluate the neuroprotective and neurite outgrowth effects of maltol, a natural aroma compound, on retinal ganglion cells (RGCs) under oxidative stress in vitro. METHODS Mouse primary RGCs were isolated using immunopanning-magnetic separation and exposed to H2O2 in the presence of maltol. The cell viability and apoptosis were determined by using adenosine 5'-triphosphate (ATP) assay and terminal deoxynucleotidyl transferase (TdT)-mediated deoxyuridine triphosphate (dUTP) nick end labeling (TUNEL), respectively. Neurite outgrowth was assessed by immunofluorescence for α-tubulin. The activation of nuclear factor-κB (NF-κB) was also evaluated using immunofluorescence. RESULTS When the RGCs were exposed to 20 μM of H2O2 for 16 h, their viability dropped to 40.3±3.4%. However, the maltol treatment restored the cells in a dose-dependent manner. The viability recovered to 73.9±5.1% with 10 μM of maltol and even reached 175.1±11.3% with 2 mM of maltol, as measured by ATP assay. This oxidative stress significantly increased the number of TUNEL-positive RGCs, but the maltol drastically reduced the proportion of those apoptotic cells. The oxidative stress hampered the neurite outgrowth of the RGCs, whereas maltol restored their ability to sprout neurites. Regarding NF-κB, the active form of phosphorylated NF-κB (pNF-κB) increased the oxidative stress level but the maltol treatment again reduced it to an unstressful level. CONCLUSIONS Our data revealed that maltol attenuated the oxidative stress-induced injury in the primary mouse RGCs. Its neuroprotective and neurite outgrowth effects seemed to be related to NF-κB signaling. Maltol has potential as a new neuroprotective therapeutic agent for oxidative stress-related ocular diseases, including glaucoma.
Collapse
Affiliation(s)
- Samin Hong
- Institute of Vision Research, Department of Ophthalmology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yoko Iizuka
- Institute of Vision Research, Department of Ophthalmology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Taekjune Lee
- Institute of Vision Research, Department of Ophthalmology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Chan Yun Kim
- Institute of Vision Research, Department of Ophthalmology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Gong Je Seong
- Institute of Vision Research, Department of Ophthalmology, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
23
|
Abcouwer SF, Lin CM, Shanmugam S, Muthusamy A, Barber AJ, Antonetti DA. Minocycline prevents retinal inflammation and vascular permeability following ischemia-reperfusion injury. J Neuroinflammation 2013; 10:149. [PMID: 24325836 PMCID: PMC3866619 DOI: 10.1186/1742-2094-10-149] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 11/24/2013] [Indexed: 12/21/2022] Open
Abstract
Background Many retinal diseases are associated with vascular dysfunction accompanied by neuroinflammation. We examined the ability of minocycline (Mino), a tetracycline derivative with anti-inflammatory and neuroprotective properties, to prevent vascular permeability and inflammation following retinal ischemia-reperfusion (IR) injury, a model of retinal neurodegeneration with breakdown of the blood-retinal barrier (BRB). Methods Male Sprague–Dawley rats were subjected to 45 min of pressure-induced retinal ischemia, with the contralateral eye serving as control. Rats were treated with Mino prior to and following IR. At 48 h after reperfusion, retinal gene expression, cellular inflammation, Evan’s blue dye leakage, tight junction protein organization, caspase-3 activation, and DNA fragmentation were measured. Cellular inflammation was quantified by flow-cytometric evaluation of retinal tissue using the myeloid marker CD11b and leukocyte common antigen CD45 to differentiate and quantify CD11b+/CD45low microglia, CD11b+/CD45hi myeloid leukocytes and CD11bneg/CD45hi lymphocytes. Major histocompatibility complex class II (MHCII) immunoreactivity was used to determine the inflammatory state of these cells. Results Mino treatment significantly inhibited IR-induced retinal vascular permeability and disruption of tight junction organization. Retinal IR injury significantly altered mRNA expression for 21 of 25 inflammation- and gliosis-related genes examined. Of these, Mino treatment effectively attenuated IR-induced expression of lipocalin 2 (LCN2), serpin peptidase inhibitor clade A member 3 N (SERPINA3N), TNF receptor superfamily member 12A (TNFRSF12A), monocyte chemoattractant-1 (MCP-1, CCL2) and intercellular adhesion molecule-1 (ICAM-1). A marked increase in leukostasis of both myeloid leukocytes and lymphocytes was observed following IR. Mino treatment significantly reduced retinal leukocyte numbers following IR and was particularly effective in decreasing the appearance of MHCII+ inflammatory leukocytes. Surprisingly, Mino did not significantly inhibit retinal cell death in this model. Conclusions IR induces a retinal neuroinflammation within hours of reperfusion characterized by inflammatory gene expression, leukocyte adhesion and invasion, and vascular permeability. Despite Mino significantly inhibiting these responses, it failed to block neurodegeneration.
Collapse
Affiliation(s)
- Steven F Abcouwer
- Department of Ophthalmology and Visual Sciences, University of Michigan Kellogg Eye Center, 1000 Wall Street, Ann Arbor, MI 48105, USA.
| | | | | | | | | | | |
Collapse
|
24
|
Palanca A, Casafont I, Berciano MT, Lafarga M. Reactive nucleolar and Cajal body responses to proteasome inhibition in sensory ganglion neurons. Biochim Biophys Acta Mol Basis Dis 2013; 1842:848-59. [PMID: 24269586 DOI: 10.1016/j.bbadis.2013.11.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Revised: 11/12/2013] [Accepted: 11/13/2013] [Indexed: 12/25/2022]
Abstract
The dysfunction of the ubiquitin proteasome system has been related to a broad array of neurodegenerative disorders in which the accumulation of misfolded protein aggregates causes proteotoxicity. The ability of proteasome inhibitors to induce cell cycle arrest and apoptosis has emerged as a powerful strategy for cancer therapy. Bortezomib is a proteasome inhibitor used as an antineoplastic drug, although its neurotoxicity frequently causes a severe sensory peripheral neuropathy. In this study we used a rat model of bortezomib treatment to study the nucleolar and Cajal body responses to the proteasome inhibition in sensory ganglion neurons that are major targets of bortezomib-induced neurotoxicity. Treatment with bortezomib induced dose-dependent dissociation of protein synthesis machinery (chromatolysis) and nuclear retention of poly(A) RNA granules resulting in neuronal dysfunction. However, as a compensatory response to the proteotoxic stress, both nucleoli and Cajal bodies exhibited reactive changes. These include an increase in the number and size of nucleoli, strong nucleolar incorporation of the RNA precursor 5'-fluorouridine, and increased expression of both 45S rRNA and genes encoding nucleolar proteins UBF, fibrillarin and B23. Taken together, these findings appear to reflect the activation of the nucleolar transcription in response to proteotoxic stress Furthermore, the number of Cajal bodies, a parameter related to transcriptional activity, increases upon proteasome inhibition. We propose that nucleoli and Cajal bodies are important targets in the signaling pathways that are activated by the proteotoxic stress response to proteasome inhibition. The coordinating activity of these two organelles in the production of snRNA, snoRNA and rRNA may contribute to neuronal survival after proteasome inhibition. This article is part of a Special Issue entitled: Role of the Nucleolus in Human Disease.
Collapse
Affiliation(s)
- Ana Palanca
- Department of Anatomy and Cell Biology, University of Cantabria-IFIMAV, Santander, Spain; "Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)", Santander, Spain
| | - Iñigo Casafont
- Department of Anatomy and Cell Biology, University of Cantabria-IFIMAV, Santander, Spain; "Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)", Santander, Spain
| | - María T Berciano
- Department of Anatomy and Cell Biology, University of Cantabria-IFIMAV, Santander, Spain; "Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)", Santander, Spain
| | - Miguel Lafarga
- Department of Anatomy and Cell Biology, University of Cantabria-IFIMAV, Santander, Spain; "Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)", Santander, Spain.
| |
Collapse
|
25
|
Bejaoui M, Zaouali MA, Folch-Puy E, Pantazi E, Bardag-Gorce F, Carbonell T, Oliva J, Rimola A, Abdennebi HB, Roselló-Catafau J. Bortezomib enhances fatty liver preservation in Institut George Lopez-1 solution through adenosine monophosphate activated protein kinase and Akt/mTOR pathways. ACTA ACUST UNITED AC 2013; 66:62-72. [PMID: 24127984 DOI: 10.1111/jphp.12154] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2013] [Accepted: 09/05/2013] [Indexed: 12/29/2022]
Abstract
OBJECTIVES The aim of this study is to investigate the protective mechanisms induced by bortezomib added to Institut George Lopez (IGL)-1 preservation solution to protect steatotic livers against cold ischaemia reperfusion injury and to examine whether these mechanisms occur through the activation of adenosine monophosphate activated protein kinase (AMPK), Akt/mTOR pathways. METHODS Steatotic livers from obese rats were preserved for 24 h (at 4 °C) in IGL-1 solution with or without bortezomib (100 nM) or pretreated with AMPK inhibitor adenine 9-α-D-arabinofuranoside and preserved in IGL-1 + bortezomib. Livers were then perfused for 2 h at 37 °C. Liver injury (alanine aminotransferase/aspartate aminotransferase) and function (bile production and vascular resistance) were measured. Also, Akt/mTOR, phosphorylated AMPK (pAMPK) and apoptosis were determined by Western blot analyses. KEY FINDINGS Bortezomib addition to IGL-1 solution significantly reduced steatotic liver injury, improved graft function and decreased liver apoptosis. These benefits were diminished by the pretreatment of obese rats with AMPK inhibitor Ara. Western blot analyses showed a significant increase in pAMPK after ischaemia and reperfusion. We also observed a significant phosphorylation of Akt in IGL-1 +bortezomib group that, in turn, induced the phosphorylation of mTOR and glycogen synthase kinase 3β. CONCLUSIONS Bortezomib, at low and non toxic concentration, is a promising additive to IGL-1 solution for steatotic liver preservation. Its protective effect is due to the activation of AMPK and Akt/mTOR pathways.
Collapse
Affiliation(s)
- Mohamed Bejaoui
- Experimental Pathology Department, IIBB-CSIC, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), IDIBAPS, Barcelona, Catalonia, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Ramapathiran L, Bernas T, Walter F, Williams L, Düssmann H, Concannon CG, Prehn JHM. Single cell imaging of the heat shock response during proteasome inhibitor-induced apoptosis in colon cancer cells suggests that magnitude and length rather than time of onset determines resistance to apoptosis. J Cell Sci 2013; 127:609-19. [DOI: 10.1242/jcs.137158] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Targeting the proteasome is a valuable approach for cancer therapy, potentially limited by pro-survival pathways induced in parallel to cell death. Whether these pro-survival pathways are activated in all cells, show different activation kinetics in sensitive versus resistant cells, or interact functionally with cell death pathways is unknown. We monitored activation of the heat shock response (HSR), a key survival pathway induced by proteasome inhibition, relative to apoptosis activation in HCT116 colon cancer cells expressing green fluorescent protein (GFP) under the control of the Hsp70 promoter. Single cell and high content time-lapse imaging of epoxomicin treatment revealed that neither basal activity, nor the time of onset of the HSR differed between resistant and sensitive populations. However, resistant cells had significantly higher and prolonged reporter activity than those that succumbed to cell death. p53 deficiency protected against cell death but failed to modulate the HSR. In contrast, inhibition of the HSR significantly increased the cytotoxicity of epoxomicin. Our data provide novel insights into the kinetics and heterogeneity of HSR during proteasome inhibition, suggesting that the HSR modulates cell death signaling unidirectionally.
Collapse
|