1
|
Jakobsen TS, Adsersen RL, Askou AL, Corydon TJ. Functional Roles of Pigment Epithelium-Derived Factor in Retinal Degenerative and Vascular Disorders: A Scoping Review. Invest Ophthalmol Vis Sci 2024; 65:41. [PMID: 39728690 PMCID: PMC11684118 DOI: 10.1167/iovs.65.14.41] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 11/29/2024] [Indexed: 12/28/2024] Open
Abstract
Purpose This review explores the role of pigment epithelium-derived factor (PEDF) in retinal degenerative and vascular disorders and assesses its potential both as an adjunct to established vascular endothelial growth factor inhibiting treatments for retinal vascular diseases and as a neuroprotective therapeutic agent. Methods A comprehensive literature review was conducted, focusing on the neuroprotective and anti-angiogenic properties of PEDF. The review evaluated its effects on retinal health, its dysregulation in ocular disorders, and its therapeutic application in preclinical models. Advances in drug delivery, including gene therapy, were also examined. Results PEDF, initially identified for promoting neuronal differentiation, is also a potent endogenous angiogenesis inhibitor. Strong anti-angiogenic and neuroprotective effects are observed in preclinical studies. It has pro-apoptotic and antiproliferative effects on endothelial cells thereby reducing neovascularization. Although promising, clinical development is limited with only a single conducted phase I clinical trial for macular neovascularization. Development of PEDF-derived peptides enhances potency and specificity, and emerging gene therapy approaches offer sustained PEDF expression for long-term treatment. However, questions regarding dosage, durability, and efficacy remain, particularly in large animal models. Conclusions PEDF shows significant therapeutic potential in preclinical models of retinal degeneration and vascular disorders. Despite inconclusive evidence on PEDF downregulation as a primary disease driver, many studies highlight its therapeutic benefits and favorable safety profile. Advances in gene therapy could enable long-acting PEDF-based treatments, but further research is needed to optimize dosage and durability, potentially leading to clinical trials and expanding treatment options for retinal disorders.
Collapse
Affiliation(s)
- Thomas Stax Jakobsen
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
- Department of Ophthalmology, Aarhus University Hospital, Aarhus N, Denmark
| | | | - Anne Louise Askou
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
- Department of Ophthalmology, Aarhus University Hospital, Aarhus N, Denmark
| | - Thomas J. Corydon
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
- Department of Ophthalmology, Aarhus University Hospital, Aarhus N, Denmark
| |
Collapse
|
2
|
Zhao Z, Liu W, Cheng G, Dong S, Zhao Y, Wu H, Cao Z. Knockdown of DAPK1 inhibits IL-1β-induced inflammation and cartilage degradation in human chondrocytes by modulating the PEDF-mediated NF-κB and NLRP3 inflammasome pathway. Innate Immun 2024; 30:21-30. [PMID: 36412004 PMCID: PMC10720599 DOI: 10.1177/17534259221086837] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 01/23/2022] [Accepted: 02/24/2022] [Indexed: 12/17/2023] Open
Abstract
Osteoarthritis (OA) is a common joint disease that is characterized by inflammation and cartilage degradation. Death-associated protein kinase 1 (DAPK1) is a multi-domain serine/threonine kinase and has been reported to be involved in the progression of OA. However, its role and mechanism in OA remain unclear. Here, we found the expression of DAPK1 in OA cartilage tissues was higher than that in normal cartilage tissues. The expression of DAPK1 in chondrocytes was up-regulated by IL-1β. Knockdown of DAPK1 promoted cell viability and anti-apoptotic protein expression, while it inhibited the apoptosis rate and pro-apoptotic protein expressions in IL-1β-induced chondrocytes. In addition, DAPK1 inhibition reduced the levels of inflammatory cytokines and expressions of matrix metalloproteinases (MMPs), and increased the expressions of collagen II and aggrecan. The data of mechanistic investigation indicated that the expression of pigment epithelium-derived factor (PEDF) was positively regulated by DAPK1. Overexpression of PEDF attenuated the effects of DAPK1 knockdown on IL-1β-induced cell viability, apoptosis, inflammation, and cartilage degradation. Furthermore, PEDF overexpression restored the activity of the NF-κB pathway and NLRP3 inflammasome after DAPK1 knockdown. Collectively, down-regulation of DAPK1 inhibited IL-1β-induced inflammation and cartilage degradation via the PEDF-mediated NF-κB and NLRP3 inflammasome pathways.
Collapse
Affiliation(s)
- Zhongyuan Zhao
- Department of Articulation Surgery, Yantaishan Hospital, Yantai, Shandong Province, China
| | - Wei Liu
- Department of Pathophysiology, Binzhou Medical University, Yantai, Shandong Province, China
| | - Gong Cheng
- Department of Sports Medicine, Yantaishan Hospital, Yantai, Shandong Province, China
| | - Shengjie Dong
- Department of Articulation Surgery, Yantaishan Hospital, Yantai, Shandong Province, China
| | - Yuchi Zhao
- Department of Articulation Surgery, Yantaishan Hospital, Yantai, Shandong Province, China
| | - Hao Wu
- Department of Sports Medicine, Yantaishan Hospital, Yantai, Shandong Province, China
| | - Zhilin Cao
- Department of Sports Medicine, Yantaishan Hospital, Yantai, Shandong Province, China
| |
Collapse
|
3
|
Kvivik I, Grimstad T, Bårdsen K, Jonsson G, Kvaløy JT, Omdal R. High mobility group box 1 and a network of other biomolecules influence fatigue in patients with Crohn's disease. Mol Med 2023; 29:81. [PMID: 37365509 DOI: 10.1186/s10020-023-00679-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 06/05/2023] [Indexed: 06/28/2023] Open
Abstract
BACKGROUND Fatigue is common in patients with chronic inflammatory and autoimmune diseases, often with a severe impact on the patient's daily life. From a biological point of view, fatigue can be regarded as an element of the sickness behavior response, a coordinated set of responses induced by pathogens to enhance survival during an infection and immunological danger. The mechanisms are not fully understood but involve activation of the innate immune system, with pro-inflammatory cytokines, in particular interleukin (IL)-1β, acting on cerebral neurons. These mechanisms are also active during chronic inflammatory conditions. High mobility group box 1 (HMGB1) protein has interleukin-1 like properties and is a strong inducer of innate immune responses. Its role in generation of fatigue is not clarified. Emerging evidence indicates that also other biomolecules may influence sickness behavior. We aimed to elucidate how HMGB1 influences fatigue in patients with Crohn's disease, and how the protein interacts with other candidate biomarkers of fatigue. METHODS In 56 patients with newly diagnosed Crohn's disease, fatigue was evaluated using three different fatigue instruments: the fatigue visual analog scale (fVAS), Fatigue Severity Scale (FSS), and the vitality subscale of Medical Outcomes Study Short-Form Health Survey (SF-36vs). The biochemical markers IL-1 receptor antagonist (RA), soluble IL-1 receptor type 2 (sIL-RII), heat shock protein 90 alpha (HSP90α), HMGB1, anti-fully reduced (fr)HMGB1 antibodies (abs), hemopexin (HPX), and pigment epithelium-derived factor (PEDF) were measured in plasma. Multivariable regression and principal component analyses (PCA) were applied. RESULTS Multivariable regression analyses revealed significant contributions to fatigue severity for HMGB1 in the FSS model, HSP90α in the fVAS model and IL-1RA in the SF-36vs model. Depression and pain scores contributed to all three models. In PCA, two components described 53.3% of the variation. The "inflammation and cellular stress dimension" was dominated by IL-1RA, sIL-1RII, HSP90α, HPX, and PEDF scores, where the "HMGB1 dimension" was dominated by HMGB1, anti-frHMGB1 abs, and fVAS scores. CONCLUSION This study supports the hypothesis that HMGB1 and a network of other biomolecules influence fatigue severity in chronic inflammatory conditions. The well-known association with depression and pain is also acknowledged.
Collapse
Affiliation(s)
- Ingeborg Kvivik
- Research Department, Stavanger University Hospital, P.O. Box 8100, 4068, Stavanger, Norway
| | - Tore Grimstad
- Unit of Gastroenterology, Department of Internal Medicine, Stavanger University Hospital, Stavanger, Norway
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Kjetil Bårdsen
- Research Department, Stavanger University Hospital, P.O. Box 8100, 4068, Stavanger, Norway
| | - Grete Jonsson
- Department of Medical Biochemistry, Stavanger University Hospital, Stavanger, Norway
| | - Jan Terje Kvaløy
- Research Department, Stavanger University Hospital, P.O. Box 8100, 4068, Stavanger, Norway
- Department of Mathematics and Physics, University of Stavanger, Stavanger, Norway
| | - Roald Omdal
- Research Department, Stavanger University Hospital, P.O. Box 8100, 4068, Stavanger, Norway.
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway.
- Department of Rheumatology, Stavanger University Hospital, Stavanger, Norway.
| |
Collapse
|
4
|
Bernardo-Colón A, Lerner M, Becerra SP. Pigment epithelium-derived factor is an interleukin-6 antagonist in the RPE: Insight of structure-function relationships. Front Physiol 2022; 13:1045613. [PMID: 36467689 PMCID: PMC9709256 DOI: 10.3389/fphys.2022.1045613] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 11/03/2022] [Indexed: 07/04/2024] Open
Abstract
Retinal and choroidal inflammatory lesions increase the levels of the pro-inflammatory cytokine interleukin-6 (IL-6). Pigment epithelium-derived factor (PEDF) has anti-inflammatory properties, but it is not known if it can prevent the production of IL-6 by the retinal pigment epithelium. To investigate the anti-inflammatory effects of PEDF in the RPE, we used human ARPE-19 cells stimulated with human recombinant tumor necrosis factor-alpha (TNF-α) to induce overexpression of the IL6 gene. We found that the viability of ARPE-19 cells decreased by 22% with TNF-α at 10 ng/ml, being drastically decreased at ≥50 ng/ml. TNF-α at 5-100 ng/ml elevated the production and secretion of IL-6 protein, as measured by ELISA. To challenge the TNF-α-mediated stimulation of IL-6, we used recombinant human PEDF protein. PEDF at 100 nM recovered the TNF-α-mediated loss of cell viability and repressed IL-6 gene expression as determined by RT-PCR. PEDF at 10-100 nM attenuated the IL-6 protein secretion in a dose dependent fashion (IC50 = 65 nM), being abolished with 100 nM PEDF. To map the region that confers the IL-6 blocking effect to the PEDF polypeptide, we used chemically synthesized peptides designed from its biologically active domains, pro-death 34-mer, and pro-survival 44-mer and 17-mer (H105A), to challenge the IL-6 overproduction. The pro-survival peptides recovered the TNF-α-mediated cell viability loss, and inhibited IL-6 secretion, while the 34-mer did not have an effect, suggesting a role for the pro-survival domain in blocking TNF-α-mediated cell death and IL-6 stimulation. Our findings position PEDF as a novel antagonistic agent of IL-6 production in RPE cells, underscoring its use for the management of retinal disease-related inflammation.
Collapse
Affiliation(s)
| | | | - S. Patricia Becerra
- Laboratory of Retinal Cell and Molecular Biology, Section of Protein Structure and Function, National Eye Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
5
|
Wang Y, Liu X, Quan X, Qin X, Zhou Y, Liu Z, Chao Z, Jia C, Qin H, Zhang H. Pigment epithelium-derived factor and its role in microvascular-related diseases. Biochimie 2022; 200:153-171. [DOI: 10.1016/j.biochi.2022.05.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 05/19/2022] [Accepted: 05/30/2022] [Indexed: 01/02/2023]
|
6
|
Morenikeji OB, Bernard K, Strutton E, Wallace M, Thomas BN. Evolutionarily Conserved Long Non-coding RNA Regulates Gene Expression in Cytokine Storm During COVID-19. Front Bioeng Biotechnol 2021; 8:582953. [PMID: 33520952 PMCID: PMC7844208 DOI: 10.3389/fbioe.2020.582953] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 10/27/2020] [Indexed: 01/08/2023] Open
Abstract
Coronavirus is a family of viruses including alpha-, beta-, gamma-, delta-coronaviruses. Only alpha- and betacoronaviruses have been observed to infect humans. Past outbreaks of SARS-CoV and MERS-CoV, both betacoronavirus, are the result of a spillover from animals. Recently, a new strain termed SARS-CoV-2 emerged in December 2019 in Wuhan, China. Severe cases of COVID-19, the disease caused by SARS-CoV-2, lead to acute respiratory distress syndrome (ARDS). One contributor to the development of ARDS is cytokine storm, an overwhelming inflammatory immune response. Long non-coding RNAs (lncRNAs) are genetic regulatory elements that, among many functions, alter gene expression and cellular processes. lncRNAs identified to be pertinent in COVID-19 cytokine storm have the potential to serve as disease markers or drug targets. This project aims to computationally identify conserved lncRNAs potentially regulating gene expression in cytokine storm during COVID-19. We found 22 lncRNAs that can target 10 cytokines overexpressed in COVID-19 cytokine storm, 8 of which targeted two or more cytokine storm cytokines. In particular, the lncRNA non-coding RNA activated by DNA damage (NORAD), targeted five out of the ten identified cytokine storm cytokines, and is evolutionarily conserved across multiple species. These lncRNAs are ideal candidates for further in vitro and in vivo analysis.
Collapse
Affiliation(s)
| | - Kahleel Bernard
- Department of Biology, Hamilton College, Clinton, NY, United States
| | - Ellis Strutton
- Department of Biology, Hamilton College, Clinton, NY, United States
| | | | - Bolaji N. Thomas
- Department of Biomedical Sciences, Rochester Institute of Technology, Rochester, NY, United States
| |
Collapse
|
7
|
Mugahid DA, Sengul TG, You X, Wang Y, Steil L, Bergmann N, Radke MH, Ofenbauer A, Gesell-Salazar M, Balogh A, Kempa S, Tursun B, Robbins CT, Völker U, Chen W, Nelson L, Gotthardt M. Proteomic and Transcriptomic Changes in Hibernating Grizzly Bears Reveal Metabolic and Signaling Pathways that Protect against Muscle Atrophy. Sci Rep 2019; 9:19976. [PMID: 31882638 PMCID: PMC6934745 DOI: 10.1038/s41598-019-56007-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 12/05/2019] [Indexed: 12/31/2022] Open
Abstract
Muscle atrophy is a physiological response to disuse and malnutrition, but hibernating bears are largely resistant to this phenomenon. Unlike other mammals, they efficiently reabsorb amino acids from urine, periodically activate muscle contraction, and their adipocytes differentially responds to insulin. The contribution of myocytes to the reduced atrophy remains largely unknown. Here we show how metabolism and atrophy signaling are regulated in skeletal muscle of hibernating grizzly bear. Metabolic modeling of proteomic changes suggests an autonomous increase of non-essential amino acids (NEAA) in muscle and treatment of differentiated myoblasts with NEAA is sufficient to induce hypertrophy. Our comparison of gene expression in hibernation versus muscle atrophy identified several genes differentially regulated during hibernation, including Pdk4 and Serpinf1. Their trophic effects extend to myoblasts from non-hibernating species (including C. elegans), as documented by a knockdown approach. Together, these changes reflect evolutionary favored adaptations that, once translated to the clinics, could help improve atrophy treatment.
Collapse
Affiliation(s)
- D A Mugahid
- Neuromuscular and Cardiovascular Cell Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - T G Sengul
- Neuromuscular and Cardiovascular Cell Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - X You
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Y Wang
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - L Steil
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - N Bergmann
- Neuromuscular and Cardiovascular Cell Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - M H Radke
- Neuromuscular and Cardiovascular Cell Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - A Ofenbauer
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - M Gesell-Salazar
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - A Balogh
- Experimental and Clinical Research Center, Charité & Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - S Kempa
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - B Tursun
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - C T Robbins
- School of the Environment and School of Biological Sciences, Washington State University, Pullman, Washington, USA
| | - U Völker
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
| | - W Chen
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - L Nelson
- College of Veterinary Medicine and Department of Veterinary Clinical Science, Washington State University, Pullman, Washington, USA
| | - M Gotthardt
- Neuromuscular and Cardiovascular Cell Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany. .,Charité Universitätsmedizin Berlin, Berlin, Germany. .,DZHK (German Center for Cardiovascular Research), partner site Berlin, Berlin, Germany.
| |
Collapse
|
8
|
Brook N, Brook E, Dharmarajan A, Chan A, Dass CR. The role of pigment epithelium-derived factor in protecting against cellular stress. Free Radic Res 2019; 53:1166-1180. [PMID: 31760841 DOI: 10.1080/10715762.2019.1697809] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Since its discovery as a neurotrophic factor in retinal pigmented epithelium cells in the late 1980s, there has been an increase in understanding of the role that pigment epithelium-derived factor (PEDF) plays in cellular functions. PEDF plays an important role in mediating cellular protection during exposure to oxidative stress and inflammation by preventing stress-induced angiogenesis and apoptosis. PEDF acts to reduce oxidative stress by promoting mitochondrial stability and by regulating the expression of enzymes involved in ROS accumulation and clearance. PEDF protects against the negative effects of oxidative stress by regulating cell survival pathways and the expression of inflammatory and proangiogenic mediators. PEDF-mediated cellular protection may be of clinical importance in diseases characterised by oxidative stress, chronic inflammation and pathological neovascularization, indicating that targeting PEDF may be a potential focus for therapeutic interventions in chronic diseases. In this review, we provide a historical perspective on the discoveries of PEDF interactions and functions, and discuss recent in vitro, in vivo and clinical findings to provide a current summary of the important protective effects following cellular exposure to stress stimuli and future clinical potential of PEDF.
Collapse
Affiliation(s)
- Naomi Brook
- School of Pharmacy and Biomedical Science, Curtin University, Bentley, Australia.,Curtin Health Innovation Research Institute, Bentley, Australia
| | - Emily Brook
- School of Pharmacy and Biomedical Science, Curtin University, Bentley, Australia.,Curtin Health Innovation Research Institute, Bentley, Australia
| | - Arun Dharmarajan
- School of Pharmacy and Biomedical Science, Curtin University, Bentley, Australia.,Curtin Health Innovation Research Institute, Bentley, Australia.,Department of Biomedical Sciences, Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| | - Arlene Chan
- Curtin Medical School, Curtin University, Bentley, Australia.,Hollywood Private Hospital, Breast Clinical Trials Unit, Breast Cancer Research Centre-Western Australia, Nedlands, Australia
| | - Crispin R Dass
- School of Pharmacy and Biomedical Science, Curtin University, Bentley, Australia.,Curtin Health Innovation Research Institute, Bentley, Australia
| |
Collapse
|
9
|
Hou J, Wang S, Jia M, Li D, Liu Y, Li Z, Zhu H, Xu H, Sun M, Lu L, Zhou Z, Peng H, Zhang Q, Fu S, Liang G, Yao L, Yu X, Carpp LN, Huang Y, McElrath J, Self S, Shao Y. A Systems Vaccinology Approach Reveals Temporal Transcriptomic Changes of Immune Responses to the Yellow Fever 17D Vaccine. THE JOURNAL OF IMMUNOLOGY 2017; 199:1476-1489. [PMID: 28687661 DOI: 10.4049/jimmunol.1700083] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 06/09/2017] [Indexed: 01/10/2023]
Abstract
In this study, we used a systems vaccinology approach to identify temporal changes in immune response signatures to the yellow fever (YF)-17D vaccine, with the aim of comprehensively characterizing immune responses associated with protective immunity. We conducted a cohort study in which 21 healthy subjects in China were administered one dose of the YF-17D vaccine; PBMCs were collected at 0 h and then at 4 h and days 1, 2, 3, 5, 7, 14, 28, 84, and 168 postvaccination, and analyzed by transcriptional profiling and immunological assays. At 4 h postvaccination, genes associated with innate cell differentiation and cytokine pathways were dramatically downregulated, whereas receptor genes were upregulated, compared with their baseline levels at 0 h. Immune response pathways were primarily upregulated on days 5 and 7, accompanied by the upregulation of the transcriptional factors JUP, STAT1, and EIF2AK2. We also observed robust activation of innate immunity within 2 d postvaccination and a durable adaptive response, as assessed by transcriptional profiling. Coexpression network analysis indicated that lysosome activity and lymphocyte proliferation were associated with dendritic cell (DC) and CD4+ T cell responses; FGL2, NFAM1, CCR1, and TNFSF13B were involved in these associations. Moreover, individuals who were baseline-seropositive for Abs against another flavivirus exhibited significantly impaired DC, NK cell, and T cell function in response to YF-17D vaccination. Overall, our findings indicate that YF-17D vaccination induces a prompt innate immune response and DC activation, a robust Ag-specific T cell response, and a persistent B cell/memory B cell response.
Collapse
Affiliation(s)
- Jue Hou
- State Key Laboratory of Infectious Disease Prevention and Control, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Shuhui Wang
- State Key Laboratory of Infectious Disease Prevention and Control, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Manxue Jia
- State Key Laboratory of Infectious Disease Prevention and Control, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Dan Li
- State Key Laboratory of Infectious Disease Prevention and Control, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Ying Liu
- State Key Laboratory of Infectious Disease Prevention and Control, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Zhengpeng Li
- State Key Laboratory of Infectious Disease Prevention and Control, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Hong Zhu
- Beijing Entry-Exit Inspection and Quarantine Bureau, Beijing 102206, China
| | - Huifang Xu
- Beijing Entry-Exit Inspection and Quarantine Bureau, Beijing 102206, China
| | - Meiping Sun
- Beijing Center for Disease Control and Prevention, Beijing 102206, China
| | - Li Lu
- Beijing Center for Disease Control and Prevention, Beijing 102206, China
| | - Zhinan Zhou
- Beijing Center for Disease Control and Prevention, Beijing 102206, China
| | - Hong Peng
- State Key Laboratory of Infectious Disease Prevention and Control, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Qichen Zhang
- State Key Laboratory of Infectious Disease Prevention and Control, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Shihong Fu
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou 310058, China
| | - Guodong Liang
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou 310058, China
| | - Lena Yao
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109; and
| | - Xuesong Yu
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109; and
| | - Lindsay N Carpp
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109; and
| | - Yunda Huang
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109; and
| | - Julie McElrath
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109; and
| | - Steve Self
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109; and
| | - Yiming Shao
- State Key Laboratory of Infectious Disease Prevention and Control, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China; .,Health Science Center, Peking University, Haidian District, Beijing 100191, China
| |
Collapse
|
10
|
Nishimon S, Ohnuma T, Takebayashi Y, Katsuta N, Takeda M, Nakamura T, Sannohe T, Higashiyama R, Kimoto A, Shibata N, Gohda T, Suzuki Y, Yamagishi SI, Tomino Y, Arai H. High serum soluble tumor necrosis factor receptor 1 predicts poor treatment response in acute-stage schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry 2017; 76:145-154. [PMID: 28341443 DOI: 10.1016/j.pnpbp.2017.03.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 03/08/2017] [Accepted: 03/17/2017] [Indexed: 10/19/2022]
Abstract
Inflammation may be involved in the pathophysiology of schizophrenia. However, few cross-sectional or longitudinal studies have examined changes in biomarker expression to evaluate diagnostic and prognostic efficacy in acute-stage schizophrenia. We compared serum inflammatory biomarker concentrations in 87 patients with acute-stage schizophrenia on admission to 105 age-, sex-, and body mass index (BMI)-matched healthy controls. The measured biomarkers were soluble tumor necrosis factor receptor 1 (sTNFR1) and adiponectin, which are associated with inflammatory responses, and pigment epithelium-derived factor (PEDF), which has anti-inflammatory properties. We then investigated biomarker concentrations and associations with clinical factors in 213 patients (including 42 medication-free patients) and 110 unmatched healthy controls to model conditions typical of clinical practice. Clinical symptoms were assessed using the Brief Psychiatric Rating Scale and Global Assessment of Function. In 121 patients, biomarker levels and clinical status were evaluated at both admission and discharge. Serum sTNFR1 was significantly higher in patients with acute-stage schizophrenia compared to matched controls while no significant group differences were observed for the other markers. Serum sTNFR1 was also significantly higher in the 213 patients compared to unmatched controls. The 42 unmedicated patients had significantly lower PEDF levels compared to controls. Between admission and discharge, sTNFR1 levels decreased significantly; however, biomarker changes did not correlate with clinical symptoms. The discriminant accuracy of sTNFR1 was 93.2% between controls and patients, showing no symptom improvement during care. Inflammation and a low-level anti-inflammatory state may be involved in both schizophrenia pathogenesis and acute-stage onset. High serum sTNFR1 in the acute stage could be a useful prognostic biomarker for treatment response in clinical practice.
Collapse
Affiliation(s)
- Shohei Nishimon
- Juntendo University Schizophrenia Projects (JUSP), Department of Psychiatry, Juntendo University, Faculty of Medicine, Tokyo, Japan
| | - Tohru Ohnuma
- Juntendo University Schizophrenia Projects (JUSP), Department of Psychiatry, Juntendo University, Faculty of Medicine, Tokyo, Japan.
| | - Yuto Takebayashi
- Juntendo University Schizophrenia Projects (JUSP), Department of Psychiatry, Juntendo University, Faculty of Medicine, Tokyo, Japan
| | - Narimasa Katsuta
- Juntendo University Schizophrenia Projects (JUSP), Department of Psychiatry, Juntendo University, Faculty of Medicine, Tokyo, Japan
| | - Mayu Takeda
- Juntendo University Schizophrenia Projects (JUSP), Department of Psychiatry, Juntendo University, Faculty of Medicine, Tokyo, Japan
| | - Toru Nakamura
- Juntendo University Schizophrenia Projects (JUSP), Department of Psychiatry, Juntendo University, Faculty of Medicine, Tokyo, Japan
| | - Takahiro Sannohe
- Juntendo University Schizophrenia Projects (JUSP), Department of Psychiatry, Juntendo University, Faculty of Medicine, Tokyo, Japan
| | - Ryoko Higashiyama
- Juntendo University Schizophrenia Projects (JUSP), Department of Psychiatry, Juntendo University, Faculty of Medicine, Tokyo, Japan
| | - Ayako Kimoto
- Juntendo University Schizophrenia Projects (JUSP), Department of Psychiatry, Juntendo University, Faculty of Medicine, Tokyo, Japan
| | - Nobuto Shibata
- Juntendo University Schizophrenia Projects (JUSP), Department of Psychiatry, Juntendo University, Faculty of Medicine, Tokyo, Japan
| | - Tomohito Gohda
- Division of Nephrology, Department of Internal Medicine, Juntendo University, Faculty of Medicine, Tokyo, Japan
| | - Yusuke Suzuki
- Division of Nephrology, Department of Internal Medicine, Juntendo University, Faculty of Medicine, Tokyo, Japan
| | - Sho-Ichi Yamagishi
- Department of Pathophysiology and Therapeutics of Diabetic Vascular Complications, Kurume University School of Medicine, Kurume, Japan
| | - Yasuhiko Tomino
- Division of Nephrology, Department of Internal Medicine, Juntendo University, Faculty of Medicine, Tokyo, Japan
| | - Heii Arai
- Juntendo University Schizophrenia Projects (JUSP), Department of Psychiatry, Juntendo University, Faculty of Medicine, Tokyo, Japan
| |
Collapse
|
11
|
Ryan KM, Glaviano A, O'Donovan SM, Kolshus E, Dunne R, Kavanagh A, Jelovac A, Noone M, Tucker GM, Dunn MJ, McLoughlin DM. Electroconvulsive therapy modulates plasma pigment epithelium-derived factor in depression: a proteomics study. Transl Psychiatry 2017; 7:e1073. [PMID: 28350398 PMCID: PMC5404616 DOI: 10.1038/tp.2017.51] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 01/15/2017] [Accepted: 02/01/2017] [Indexed: 12/12/2022] Open
Abstract
Electroconvulsive therapy (ECT) is the most effective treatment for severe depression, yet its mechanism of action is not fully understood. Peripheral blood proteomic analyses may offer insights into the molecular mechanisms of ECT. Patients with a major depressive episode were recruited as part of the EFFECT-Dep trial (enhancing the effectiveness of electroconvulsive therapy in severe depression; ISRCTN23577151) along with healthy controls. As a discovery-phase study, patient plasma pre-/post-ECT (n=30) was analyzed using 2-dimensional difference in gel electrophoresis and mass spectrometry. Identified proteins were selected for confirmation studies using immunodetection methods. Samples from a separate group of patients (pre-/post-ECT; n=57) and matched healthy controls (n=43) were then used to validate confirmed changes. Target protein mRNA levels were also assessed in rat brain and blood following electroconvulsive stimulation (ECS), the animal model of ECT. We found that ECT significantly altered 121 protein spots with 36 proteins identified by mass spectrometry. Confirmation studies identified a post-ECT increase (P<0.01) in the antiangiogenic and neuroprotective mediator pigment epithelium-derived factor (PEDF). Validation work showed an increase (P<0.001) in plasma PEDF in depressed patients compared with the controls that was further increased post-ECT (P=0.03). PEDF levels were not associated with mood scores. Chronic, but not acute, ECS increased PEDF mRNA in rat hippocampus (P=0.02) and dentate gyrus (P=0.03). This study identified alterations in blood levels of PEDF in depressed patients and further alterations following ECT, as well as in an animal model of ECT. These findings implicate PEDF in the biological response to ECT for depression.
Collapse
Affiliation(s)
- K M Ryan
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland,Department of Psychiatry, Trinity College Dublin, St. Patrick's University Hospital, Dublin, Ireland
| | - A Glaviano
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - S M O'Donovan
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - E Kolshus
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland,Department of Psychiatry, Trinity College Dublin, St. Patrick's University Hospital, Dublin, Ireland
| | - R Dunne
- Department of Psychiatry, Trinity College Dublin, St. Patrick's University Hospital, Dublin, Ireland
| | - A Kavanagh
- Department of Psychiatry, Trinity College Dublin, St. Patrick's University Hospital, Dublin, Ireland
| | - A Jelovac
- Department of Psychiatry, Trinity College Dublin, St. Patrick's University Hospital, Dublin, Ireland
| | - M Noone
- Department of Psychiatry, Trinity College Dublin, St. Patrick's University Hospital, Dublin, Ireland
| | - G M Tucker
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - M J Dunn
- Conway Institute of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - D M McLoughlin
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland,Department of Psychiatry, Trinity College Dublin, St. Patrick's University Hospital, Dublin, Ireland,Department of Psychiatry, Trinity College Dublin, St. Patrick's University Hospital, James's Street, Dublin 8, Ireland. E-mail:
| |
Collapse
|
12
|
Nakamura DS, Hollander JM, Uchimura T, Nielsen HC, Zeng L. Pigment Epithelium-Derived Factor (PEDF) mediates cartilage matrix loss in an age-dependent manner under inflammatory conditions. BMC Musculoskelet Disord 2017; 18:39. [PMID: 28122611 PMCID: PMC5264335 DOI: 10.1186/s12891-017-1410-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 01/16/2017] [Indexed: 02/04/2023] Open
Abstract
Background Inflammation is a major cause of cartilage destruction and leads to the imbalance of metabolic activities in the arthritic joint. Pigment epithelium-derived factor (PEDF) has been reported to have both pro- and anti-inflammatory activities in various cell types and to be upregulated in the arthritic joint, but its role in joint destruction is unclear. Our aim was to investigate the role of PEDF in cartilage degeneration under inflammatory conditions. Methods PEDF was ectopically expressed in primary human articular chondrocytes, and catabolic gene expression and protein secretion in response to the pro-inflammatory cytokine interleukin 1 beta (IL-1β) were evaluated. Metatarsal bones from PEDF-deficient and wild type mice were cultured in the presence or absence of IL-1β. Cartilage matrix integrity and matrix metalloproteinases MMP-1, MMP-3, and MMP-13 were evaluated. PEDF-deficient and wild type mice were evaluated in the monosodium iodoacetate (MIA) inflammatory joint destruction animal model to determine the role of PEDF in inflammatory arthritis in vivo. Student’s t-tests and Mann–Whitney tests were employed where appropriate, for parametric and non-parametric data, respectively. Results We showed that PEDF protein levels were higher in human osteoarthritis samples compared to normal samples. We demonstrated that ectopic PEDF expression in primary human articular chondrocytes exacerbated catabolic gene expression in the presence of IL-1β. In whole bone organ cultures, IL-1β induced MMP-1, MMP-3 and MMP-13 protein production, and caused significant cartilage matrix loss. Interestingly, Toluidine Blue staining showed that PEDF-deficient bones from 29 week old animals, but not 10 week old animals, had reduced matrix loss in response to IL-1β compared to their wild type counterparts. In addition, PEDF-deficiency in 29 week old animals preserved matrix integrity and protected against cell loss in the MIA joint destruction model in vivo. Conclusion We conclude that PEDF exacerbates cartilage degeneration in an age-dependent manner under an inflammatory setting. This is the first study identifying a specific role for PEDF in joint inflammation and highlights the multi-faceted activities of PEDF. Electronic supplementary material The online version of this article (doi:10.1186/s12891-017-1410-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Daisy S Nakamura
- Program in Cell, Molecular and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA, USA
| | - Judith M Hollander
- Program in Cell, Molecular and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA, USA
| | - Tomoya Uchimura
- Program in Cell, Molecular and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA, USA
| | - Heber C Nielsen
- Program in Cell, Molecular and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA, USA. .,Department of Pediatrics, Tufts Medical Center, Boston, MA, USA.
| | - Li Zeng
- Program in Cell, Molecular and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA, USA. .,Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, Boston, MA, USA. .,Department of Orthopaedics, Tufts Medical Center, Boston, MA, USA.
| |
Collapse
|
13
|
Chen L, Liu X, Wang H, Qu M. Gastrodin Attenuates Pentylenetetrazole-Induced Seizures by Modulating the Mitogen-Activated Protein Kinase-Associated Inflammatory Responses in Mice. Neurosci Bull 2016; 33:264-272. [PMID: 27909971 DOI: 10.1007/s12264-016-0084-z] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Accepted: 10/14/2016] [Indexed: 01/21/2023] Open
Abstract
Gastrodin, the major component isolated from the rhizome of the Chinese traditional medicinal herb Gastrodia elata ("Tianma"), has a long history in the treatment of epilepsy and other neurological disorders. However, the molecular mechanisms are not clear. Here, we found that gastrodin ameliorated pentylenetetrazole (PTZ)-induced epileptic seizures with improvement of the electroencephalographic pattern in mice. Further studies demonstrated that gastrodin decreased the levels of the pro-inflammatory cytokines interleukin-1β and tumor necrosis factor-α while increasing interleukin-10, an anti-inflammatory cytokine in the brain. Furthermore, gastrodin attenuated the PTZ-induced microglial activation along with inhibition of mitogen-activated protein kinases, cAMP response element binding protein, and NF-κB. Our data suggest that gastrodin attenuates seizures by modulating the mitogen-activated protein kinase-associated inflammatory responses.
Collapse
Affiliation(s)
- Liming Chen
- Neurology Department, The First People's Hospital of Jingzhou, The First Affiliated Hospital of Yangtze University, Jingzhou, 434000, China
| | - Xinan Liu
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Hua Wang
- Neurology Department, The First People's Hospital of Jingzhou, The First Affiliated Hospital of Yangtze University, Jingzhou, 434000, China.
| | - Min Qu
- Hubei Provincial Key Laboratory for Applied Toxicology, Hubei Provincial Center for Disease Control and Prevention, Wuhan, 430079, China.
| |
Collapse
|
14
|
Daniele S, Zappelli E, Martini C. Trazodone regulates neurotrophic/growth factors, mitogen-activated protein kinases and lactate release in human primary astrocytes. J Neuroinflammation 2015; 12:225. [PMID: 26627476 PMCID: PMC4666178 DOI: 10.1186/s12974-015-0446-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 11/25/2015] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND In the central nervous system, glial cells provide metabolic and trophic support to neurons and respond to protracted stress and insults by up-regulating inflammatory processes. Reactive astrocytes and microglia are associated with the pathophysiology of neuronal injury, neurodegenerative diseases and major depression, in both animal models and human brains. Several studies have reported clear anti-inflammatory effects of anti-depressant treatment on astrocytes, especially in models of neurological disorders. Trazodone (TDZ) is a triazolopyridine derivative that is structurally unrelated to other major classes of antidepressants. Although the molecular mechanisms of TDZ in neurons have been investigated, it is unclear whether astrocytes are also a TDZ target. METHODS The effects of TDZ on human astrocytes were investigated in physiological conditions and following inflammatory insult with lipopolysaccharide (LPS) and tumour necrosis factor-α (TNF-α). Astrocytes were assessed for their responses to pro-inflammatory mediators and cytokines, and the receptors and signalling pathways involved in TDZ-mediated effects were evaluated. RESULTS TDZ had no effect on cell proliferation, but it decreased pro-inflammatory mediator release and modulated trophic and transcription factor mRNA expression. Following TDZ treatment, the AKT pathway was activated, whereas extracellular signal-regulated kinase and c-Jun NH2-terminal kinase were inhibited. Most importantly, a 72-h TDZ pre-treatment before inflammatory insult completely reversed the anti-proliferative effects induced by LPS-TNF-α. The expression or the activity of inflammatory mediators, including interleukin-6, c-Jun NH2-terminal kinase and nuclear factor κB, were also reduced. Furthermore, TDZ affected astrocyte metabolic support to neurons by counteracting the inflammation-mediated lactate decrease. Finally, TDZ protected neuronal-like cells against neurotoxicity mediated by activated astrocytes. These effects mainly involved an activation of 5-HT1A and an antagonism at 5-HT2A/C serotonin receptors. Fluoxetine, used in parallel, showed similar final effects nevertheless it activates different receptors/intracellular pathways. CONCLUSIONS Altogether, our results demonstrated that TDZ directly acts on astrocytes by regulating intracellular signalling pathways and increasing specific astrocyte-derived neurotrophic factor expression and lactate release. TDZ may contribute to neuronal support by normalizing trophic and metabolic support during neuroinflammation, which is associated with neurological diseases, including major depression.
Collapse
Affiliation(s)
- Simona Daniele
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano, 6, Pisa, 56126, PI, Italy.
| | - Elisa Zappelli
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano, 6, Pisa, 56126, PI, Italy.
| | - Claudia Martini
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano, 6, Pisa, 56126, PI, Italy.
| |
Collapse
|
15
|
PEDF and its roles in physiological and pathological conditions: implication in diabetic and hypoxia-induced angiogenic diseases. Clin Sci (Lond) 2015; 128:805-23. [PMID: 25881671 PMCID: PMC4557399 DOI: 10.1042/cs20130463] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Pigment epithelium-derived factor (PEDF) is a broadly expressed multifunctional member of the serine proteinase inhibitor (serpin) family. This widely studied protein plays critical roles in many physiological and pathophysiological processes, including neuroprotection, angiogenesis, fibrogenesis and inflammation. The present review summarizes the temporal and spatial distribution patterns of PEDF in a variety of developing and adult organs, and discusses its functions in maintaining physiological homoeostasis. The major focus of the present review is to discuss the implication of PEDF in diabetic and hypoxia-induced angiogenesis, and the pathways mediating PEDF's effects under these conditions. Furthermore, the regulatory mechanisms of PEDF expression, function and degradation are also reviewed. Finally, the therapeutic potential of PEDF as an anti-angiogenic drug is briefly summarized.
Collapse
|
16
|
Carnagarin R, Dharmarajan AM, Dass CR. PEDF-induced alteration of metabolism leading to insulin resistance. Mol Cell Endocrinol 2015; 401:98-104. [PMID: 25462587 DOI: 10.1016/j.mce.2014.11.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2014] [Revised: 10/21/2014] [Accepted: 11/10/2014] [Indexed: 02/06/2023]
Abstract
Pigment epithelium-derived factor (PEDF) is an anti-angiogenic, immunomodulatory, and neurotrophic serine protease inhibitor protein. PEDF is evolving as a novel metabolic regulatory protein that plays a causal role in insulin resistance. Insulin resistance is the central pathogenesis of metabolic disorders such as obesity, type 2 diabetes mellitus, polycystic ovarian disease, and metabolic syndrome, and PEDF is associated with them. The current evidence suggests that PEDF administration to animals induces insulin resistance, whereas neutralisation improves insulin sensitivity. Inflammation, lipolytic free fatty acid mobilisation, and mitochondrial dysfunction are the proposed mechanism of PEDF-mediated insulin resistance. This review summarises the probable mechanisms adopted by PEDF to induce insulin resistance, and identifies PEDF as a potential therapeutic target in ameliorating insulin resistance.
Collapse
Affiliation(s)
- Revathy Carnagarin
- Curtin Biosciences Research Precinct, Bentley 6102, Australia; School of Pharmacy, Curtin University, Bentley 6102, Australia
| | - Arunasalam M Dharmarajan
- Curtin Biosciences Research Precinct, Bentley 6102, Australia; School of Biomedical Science, Curtin University, Bentley 6102, Australia
| | - Crispin R Dass
- Curtin Biosciences Research Precinct, Bentley 6102, Australia; School of Pharmacy, Curtin University, Bentley 6102, Australia.
| |
Collapse
|
17
|
Lu Y, He M, Zhang Y, Xu S, Zhang L, He Y, Chen C, Liu C, Pi H, Yu Z, Zhou Z. Differential pro-inflammatory responses of astrocytes and microglia involve STAT3 activation in response to 1800 MHz radiofrequency fields. PLoS One 2014; 9:e108318. [PMID: 25275372 PMCID: PMC4183530 DOI: 10.1371/journal.pone.0108318] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 08/19/2014] [Indexed: 12/23/2022] Open
Abstract
Microglia and astrocytes play important role in maintaining the homeostasis of central nervous system (CNS). Several CNS impacts have been postulated to be associated with radiofrequency (RF) electromagnetic fields exposure. Given the important role of inflammation in neural physiopathologic processes, we investigated the pro-inflammatory responses of microglia and astrocytes and the involved mechanism in response to RF fields. Microglial N9 and astroglial C8-D1A cells were exposed to 1800 MHz RF for different time with or without pretreatment with STAT3 inhibitor. Microglia and astrocytes were activated by RF exposure indicated by up-regulated CD11b and glial fibrillary acidic protein (GFAP). However, RF exposure induced differential pro-inflammatory responses in astrocytes and microglia, characterized by different expression and release profiles of IL-1β, TNF-α, IL-6, PGE2, nitric oxide (NO), inducible nitric oxide synthase (iNOS) and cyclooxygenase 2 (COX2). Moreover, the RF exposure activated STAT3 in microglia but not in astrocytes. Furthermore, the STAT3 inhibitor Stattic ameliorated the RF-induced release of pro-inflammatory cytokines in microglia but not in astrocytes. Our results demonstrated that RF exposure differentially induced pro-inflammatory responses in microglia and astrocytes, which involved differential activation of STAT3 in microglia and astrocytes. Our data provide novel insights into the potential mechanisms of the reported CNS impacts associated with mobile phone use and present STAT3 as a promising target to protect humans against increasing RF exposure.
Collapse
Affiliation(s)
- Yonghui Lu
- Department of Occupational Health, Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Third Military Medical University, Chongqing, China
| | - Mindi He
- Department of Occupational Health, Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Third Military Medical University, Chongqing, China
| | - Yang Zhang
- Department of Laboratory Medicine, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Shangcheng Xu
- Department of Occupational Health, Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Third Military Medical University, Chongqing, China
| | - Lei Zhang
- Department of Occupational Health, Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Third Military Medical University, Chongqing, China
| | - Yue He
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chunhai Chen
- Department of Occupational Health, Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Third Military Medical University, Chongqing, China
| | - Chuan Liu
- Department of Occupational Health, Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Third Military Medical University, Chongqing, China
| | - Huifeng Pi
- Department of Occupational Health, Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Third Military Medical University, Chongqing, China
| | - Zhengping Yu
- Department of Occupational Health, Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Third Military Medical University, Chongqing, China
| | - Zhou Zhou
- Department of Occupational Health, Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Third Military Medical University, Chongqing, China
| |
Collapse
|
18
|
Proapoptotic PEDF functional peptides inhibit prostate tumor growth--a mechanistic study. Biochem Pharmacol 2014; 92:425-37. [PMID: 25261795 DOI: 10.1016/j.bcp.2014.09.012] [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: 06/14/2014] [Revised: 09/13/2014] [Accepted: 09/17/2014] [Indexed: 11/22/2022]
Abstract
PEDF inhibits tumor growth via anti-angiogenic activity; however, the direct effect of PEDF on prostate carcinoma and its functional epitope as well as the underlying mechanism regulating the pathway from extracellular receptors to nuclear transcription factors has not been fully elucidated. This study investigates the ability and mechanism by which the functional PEDF peptides PEDF34 and PEDF44 suppress tumor growth. The results showed that death receptor pathway was activated by PEDF34 through up-regulation of FasL and activation of caspase-8 in both xenograft tumor tissues and PC-3 cells. FasL knockdown by siRNA or JNK-p inhibition attenuated apoptosis induced by PEDF34. NF-κB and PPARγ are crucial transcription factors for FasL expression. PEDF34 up-regulated PPARγ but did not affect NF-κB. PEDF34-induced up-regulation of FasL was abolished by siRNA-mediated PPARγ knockdown or using PPARγ inhibitor GW9662, whereas inhibition of NF-κB by the inhibitor PDTC or by siRNA had no effect. Furthermore, activation of JNK is necessary for PEDF34-induced up-regulation of FasL. PEDF34 has stronger hydropathicity and more interactions with laminin receptor than PEDF44. Blocking the laminin receptor abolished the up-regulation of FasL and PPARγ by PEDF34. Moreover, PEDF34 uses a similar mechanism to induce apoptosis in both endothelial and cancer cells. This study provides evidence that PEDF34, not PEDF44, serves as the proapoptotic epitope and exerts proapoptotic activity in both cancer and endothelial cells through activation of the extrinsic death receptor pathway. The dual anti-tumor and anti-angiogenic activities of PEDF34 suggest that it may be a promising agent for the treatment of prostate cancer.
Collapse
|
19
|
Elahy M, Baindur-Hudson S, Cruzat VF, Newsholme P, Dass CR. Mechanisms of PEDF-mediated protection against reactive oxygen species damage in diabetic retinopathy and neuropathy. J Endocrinol 2014; 222:R129-39. [PMID: 24928938 DOI: 10.1530/joe-14-0065] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Pigment epithelium-derived factor (PEDF) is a pluripotent glycoprotein belonging to the serpin family. PEDF can stimulate several physiological processes such as angiogenesis, cell proliferation, and survival. Oxidative stress plays an important role in the occurrence of diabetic retinopathy (DR), which is the major cause of blindness in young diabetic adults. PEDF plays a protective role in DR and there is accumulating evidence of the neuroprotective effect of PEDF. In this paper, we review the role of PEDF and the mechanisms involved in its antioxidative, anti-inflammatory, and neuroprotective properties.
Collapse
Affiliation(s)
- Mina Elahy
- College of Health and BiomedicineVictoria University, St Albans, Victoria 3021, AustraliaSchool of Biomedical SciencesBiosciences Research PrecinctSchool of PharmacyCurtin University, Bentley, Perth, Western Australia 6102, Australia
| | - Swati Baindur-Hudson
- College of Health and BiomedicineVictoria University, St Albans, Victoria 3021, AustraliaSchool of Biomedical SciencesBiosciences Research PrecinctSchool of PharmacyCurtin University, Bentley, Perth, Western Australia 6102, Australia
| | - Vinicius F Cruzat
- College of Health and BiomedicineVictoria University, St Albans, Victoria 3021, AustraliaSchool of Biomedical SciencesBiosciences Research PrecinctSchool of PharmacyCurtin University, Bentley, Perth, Western Australia 6102, AustraliaCollege of Health and BiomedicineVictoria University, St Albans, Victoria 3021, AustraliaSchool of Biomedical SciencesBiosciences Research PrecinctSchool of PharmacyCurtin University, Bentley, Perth, Western Australia 6102, Australia
| | - Philip Newsholme
- College of Health and BiomedicineVictoria University, St Albans, Victoria 3021, AustraliaSchool of Biomedical SciencesBiosciences Research PrecinctSchool of PharmacyCurtin University, Bentley, Perth, Western Australia 6102, AustraliaCollege of Health and BiomedicineVictoria University, St Albans, Victoria 3021, AustraliaSchool of Biomedical SciencesBiosciences Research PrecinctSchool of PharmacyCurtin University, Bentley, Perth, Western Australia 6102, Australia
| | - Crispin R Dass
- College of Health and BiomedicineVictoria University, St Albans, Victoria 3021, AustraliaSchool of Biomedical SciencesBiosciences Research PrecinctSchool of PharmacyCurtin University, Bentley, Perth, Western Australia 6102, AustraliaCollege of Health and BiomedicineVictoria University, St Albans, Victoria 3021, AustraliaSchool of Biomedical SciencesBiosciences Research PrecinctSchool of PharmacyCurtin University, Bentley, Perth, Western Australia 6102, Australia
| |
Collapse
|
20
|
Nelius T, Martinez-Marin D, Hirsch J, Miller B, Rinard K, Lopez J, de Riese W, Filleur S. Pigment epithelium-derived factor expression prolongs survival and enhances the cytotoxicity of low-dose chemotherapy in castration-refractory prostate cancer. Cell Death Dis 2014; 5:e1210. [PMID: 24810046 PMCID: PMC4047872 DOI: 10.1038/cddis.2014.180] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2013] [Revised: 02/02/2014] [Accepted: 02/03/2014] [Indexed: 01/24/2023]
Abstract
There is currently no cure for advanced castration-refractory prostate cancer (CRPC) despite the recent approval of several new therapeutic agents. We report here the anti-tumor effect of the angio-inhibitory pigment epithelium-derived factor (PEDF) in the metastatic LNCaP-derivative CRPC CL1 model and explore PEDF anti-neoplasic efficacy in combination with low-dose chemotherapy. Androgen-sensitive LNCaP and CRPC PC3 cell lines were examined as comparison. Using a retroviral expression system, we showed that PEDF limited the proliferation of all prostatic cell lines tested; an effect attributed to interleukin 8 (IL8)-CXCR1/IL8RA inhibition. PEDF also reduced the number and size of 3D tumor spheroids in vitro, but only induced cell differentiation in CRPC spheroids. Similarly, PEDF inhibited the migration of CRPC cells suggesting both anti-proliferative and anti-migratory functions. In vivo, PEDF decreased by 85% and 65% the growth of subcutaneous (s.c.) PC3 and CL1 tumors, respectively. In the CL1 orthotopic model, tumor intake with lethal metastases was found in all animals; nevertheless, PEDF prolonged the median survival of tumor-bearing mice (95% confidence interval: 53±0.001 to 57±1 days). Accordingly, PEDF delayed the emergence of skeletal-related event in intra-tibial xenografts. Next, we evaluated low-dose docetaxel (DTX; 5, 1, 0.5 mg/kg) or cyclophosphamide (CTX; 10–20 mg/kg) on established s.c. PC3 tumors that conditionally express PEDF anti-tumoral epitope/NT3. Although NT3–DTX-5 mg/kg combination was inefficient, NT3–DTX-1 mg/kg and -0.5 mg/kg inhibited by 95% and 87.8%, respectively, tumor growth compared with control and induced tumor stasis. Both NT3–CTX combinations were advantageous. Inversely, PEDF–DTX-5 mg/kg and PEDF–CTX-10 mg/kg delayed the most CL1 tumor growth (15, 11 and 5 days for PEDF–DTX-5 mg/kg, PEDF–CTX-10 mg/kg and single treatments, respectively) with elevated apoptosis and serum thrombospondin-1 as possible mechanism and marker, respectively. As well, both PEDF–CTX-10 mg/kg and PEDF–DTX-5 mg/kg prolonged significantly the survival of tumor-bearing mice compared with single treatments. Metastases were reduced in PEDF–DTX-5 mg/kg compared with other treatments, suggesting that PEDF–DTX delayed metastases formation. Our results advocate that PEDF/low-dose chemotherapy may represent a new therapeutic alternative for CRPC.
Collapse
Affiliation(s)
- T Nelius
- Department of Urology, Texas Tech University-Health Sciences Center, 3601 4th Street, Lubbock, TX, USA
| | - D Martinez-Marin
- Department of Urology, Texas Tech University-Health Sciences Center, 3601 4th Street, Lubbock, TX, USA
| | - J Hirsch
- Department of Urology, Texas Tech University-Health Sciences Center, 3601 4th Street, Lubbock, TX, USA
| | - B Miller
- Department of Pathology, Texas Tech University-Health Sciences Center, 3601 4th Street, Lubbock, TX, USA
| | - K Rinard
- Department of Urology, Texas Tech University-Health Sciences Center, 3601 4th Street, Lubbock, TX, USA
| | - J Lopez
- Department of Urology, Texas Tech University-Health Sciences Center, 3601 4th Street, Lubbock, TX, USA
| | - W de Riese
- Department of Urology, Texas Tech University-Health Sciences Center, 3601 4th Street, Lubbock, TX, USA
| | - S Filleur
- 1] Department of Urology, Texas Tech University-Health Sciences Center, 3601 4th Street, Lubbock, TX, USA [2] Department of Immunology and Molecular Microbiology, Texas Tech University-Health Sciences Center, 3601 4th Street, Lubbock, TX, USA
| |
Collapse
|
21
|
Smith SB, Magid-Slav M, Brown JR. Host response to respiratory bacterial pathogens as identified by integrated analysis of human gene expression data. PLoS One 2013; 8:e75607. [PMID: 24086587 PMCID: PMC3785471 DOI: 10.1371/journal.pone.0075607] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Accepted: 08/20/2013] [Indexed: 01/24/2023] Open
Abstract
Respiratory bacterial pathogens are one of the leading causes of infectious death in the world and a major health concern complicated by the rise of multi-antibiotic resistant strains. Therapeutics that modulate host genes essential for pathogen infectivity could potentially avoid multi-drug resistance and provide a wider scope of treatment options. Here, we perform an integrative analysis of published human gene expression data generated under challenges from the gram-negative and Gram-positive bacteria pathogens, Pseudomonas aeruginosa and Streptococcus pneumoniae, respectively. We applied a previously described differential gene and pathway enrichment analysis pipeline to publicly available host mRNA GEO datasets resulting from exposure to bacterial infection. We found 72 canonical human pathways common between four GEO datasets, representing P. aeruginosa and S. pneumoniae. Although the majority of these pathways are known to be involved with immune response, we found several interesting new interactions such as the SUMO1 pathway that might have a role in bacterial infections. Furthermore, 36 host-bacterial pathways were also shared with our previous results for respiratory virus host gene expression. Based on our pathway analysis we propose several drug-repurposing opportunities supported by the literature.
Collapse
Affiliation(s)
- Steven B. Smith
- Computational Biology, Quantitative Sciences, GlaxoSmithKline, Collegeville, Pennsylvania, United States of America
- Institute for Genome Science, University of Maryland, Baltimore, Maryland, United States of America
| | - Michal Magid-Slav
- Computational Biology, Quantitative Sciences, GlaxoSmithKline, Collegeville, Pennsylvania, United States of America
| | - James R. Brown
- Computational Biology, Quantitative Sciences, GlaxoSmithKline, Collegeville, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
22
|
Cheng Q, Xia W, Yang S, Ye P, Mei M, Song Y, Luo M, Li Q. Association of serum pigment epithelium-derived factor with high-sensitivity C-reactive protein in women with polycystic ovary syndrome. J Endocrinol Invest 2013; 36:632-5. [PMID: 24105071 DOI: 10.1007/bf03346755] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
BACKGROUND The basic studies showed that Pigment epithelium-derived factor (PEDF) plays an important role in inflammation. AIM This study aims to investigate the association of serum PEDF with high-sensitivity C-reactive protein (hs-CRP) in women with polycystic ovary syndrome (PCOS). METHODS Ninety-six PCOS women and 63 healthy, age-matched controls were enrolled in this study. Serum levels of PEDF, hs-CRP, Complement C3 (C3), glucose, insulin, lipids, and total testosterone were measured. Ninety-six PCOS women and 20 controls underwent hyperinsulinemiceuglycemic clamp to assess their insulin sensitivity, which was expressed as M value. RESULTS Serum PEDF levels and hs- CRP levels were much higher in PCOS women than in controls. In PCOS women, PEDF positively correlated with body mass index (BMI), hs-CRP, C3, LDL cholesterol (LDL-c), and systolic blood pressure (SBP), and negatively correlated with M value and HDL cholesterol (HDL-c). After controlling for BMI, PEDF was still positively correlated with hs-CRP. Multiple linear regression analysis revealed that in PCOS women, hs-CRP, SBP, LDL-c and M value were independent determinants of serum PEDF. CONCLUSION The serum PEDF levels are closely associated with hs-CRP in women with PCOS. PEDF may play a role in the development of chronic inflammation in PCOS.
Collapse
Affiliation(s)
- Q Cheng
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Li CM, Li W, Man XY, Liu ZG, Zheng M. Expression of pigment epithelium-derived factor in human cutaneous appendages. Clin Exp Dermatol 2013; 38:652-8. [PMID: 23675974 DOI: 10.1111/ced.12066] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/22/2012] [Indexed: 01/10/2023]
Abstract
BACKGROUND Pigment epithelium-derived factor (PEDF), a 50-kDa glycoprotein and a member of the serine protease inhibitor gene family, is well known as a potent endogenous inhibitor of angiogenesis. However, the expression of PEDF in human cutaneous appendages has not yet been determined. AIM To investigate the expression of PEDF in human cutaneous appendages. METHODS Immunohistochemical staining was used to detect the expression of PEDF in human cutaneous appendages. Reverse transcriptase PCR, western blotting and indirect immunofluorescence were used to determine the mRNA and protein expression of PEDF on cells of the outer root sheath (ORS). A wound-healing assay was used to determine the effect of different concentrations of PEDF on the migration of ORS cells. RESULTS PEDF was expressed in the hair follicle (including epidermal matrix, inner root sheath, ORS and fibrous root sheath), sebaceous glands and eccrine sweat glands. Both protein and RNA expression of PEDF was detected, and expression was localized to both cytoplasm and nucleus of ORS cells. Both interleukin (IL)-4 and IL-17 at 25 ng/mL upregulated the expression of PEDF of ORS cells, with IL-4 having the greater effect. PEDF 50 ng/mL decreased migration of ORS cells. CONCLUSIONS PEDF is expressed in human cutaneous appendages and may play a modulatory role in the physiology of ORS cells.
Collapse
Affiliation(s)
- C-M Li
- Department of Dermatology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | | | | | | | | |
Collapse
|
24
|
Chavan SS, Hudson LK, Li JH, Ochani M, Harris Y, Patel NB, Katz D, Scheinerman JA, Pavlov VA, Tracey KJ. Identification of pigment epithelium-derived factor as an adipocyte-derived inflammatory factor. Mol Med 2012; 18:1161-8. [PMID: 22714715 DOI: 10.2119/molmed.2012.00156] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2012] [Accepted: 06/14/2012] [Indexed: 12/31/2022] Open
Abstract
Obesity is a major risk factor for insulin resistance, type 2 diabetes mellitus and cardiovascular disease. The pathophysiology of obesity is associated with chronic low-grade inflammation. Adipose tissue in obesity is significantly infiltrated by macrophages that secrete cytokines. The mechanisms of interaction between macrophages and adipocytes, leading to macrophage activation and increased cytokine release, remain to be elucidated. We reasoned that an adipocyte-derived factor might stimulate activation of macrophages. We have identified pigment epithelium-derived factor (PEDF) as a mediator of inflammation that is secreted by adipocytes and mediates macrophage activation. Recombinant PEDF activates macrophages to release tumor necrosis factor (TNF) and interleukin-1 (IL-1). The PEDF receptor adipose triglyceride lipase (ATGL) is required for PEDF-mediated macrophage activation. Selective inhibition of ATGL on macrophages attenuates PEDF-induced TNF production, and PEDF enhances the phosphorylation of p38 and extracellular signal-regulated kinase 1/2 mitogen-activated protein kinases. PEDF administration to rats results in increased serum TNF levels, and insulin resistance. Together, these findings suggest that PEDF secreted by adipocytes contributes to the onset and maintenance of chronic inflammation in obesity, and may be a therapeutic target in ameliorating insulin resistance.
Collapse
Affiliation(s)
- Sangeeta S Chavan
- Laboratory of Biomedical Science, The Feinstein Institute for Medical Research, Manhasset, New York 11030, United States of America.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Ma W, St-Jacques B, Cruz Duarte P. Targeting pain mediators induced by injured nerve-derived COX2 and PGE2 to treat neuropathic pain. Expert Opin Ther Targets 2012; 16:527-40. [DOI: 10.1517/14728222.2012.680955] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
26
|
Ahmed AS, Li J, Erlandsson-Harris H, Stark A, Bakalkin G, Ahmed M. Suppression of pain and joint destruction by inhibition of the proteasome system in experimental osteoarthritis. Pain 2011; 153:18-26. [PMID: 22018973 DOI: 10.1016/j.pain.2011.08.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2010] [Revised: 06/30/2011] [Accepted: 08/01/2011] [Indexed: 12/28/2022]
Abstract
Osteoarthritis is a degenerative joint disease with pain and loss of joint function as major pathological features. Recent studies show that proteasome inhibitors reduce pain in various pathological conditions. We evaluated the effects of MG132, a reversible proteasome inhibitor on pain and joint destruction in a rat model of osteoarthritis. Osteoarthritis was induced by intraarticular injection of monosodium iodoacetate into the rat knee. Knee joint stiffness was scored and nociception was evaluated by mechanical pressure applied to the respective hind paw. Knee joint destruction was assessed by radiological and histological analyses. Expression of matrix metalloproteinase-3 (MMP-3) was analyzed by quantitative reverse transcription polymerase chain reaction in the knee articular cartilage. Expression of substance P (SP) and calcitonin gene-related peptide (CGRP) was studied in the dorsal root ganglia (L4-L6) by quantitative reverse transcription polymerase chain reaction and in the knee joints by immunohistochemistry. Our results indicate that daily treatment of osteoarthritic rats with MG132 significantly increases their mobility while the swelling, pain thresholds, and pathological features of the affected joints were reduced. Furthermore, the upregulated expression of MMP-3, SP, and CGRP in the arthritic rats was normalized by MG132 administration. We conclude that the proteasome inhibitor MG132 reduces pain and joint destruction, probably by involving the peripheral nervous system, and that changes in SP and CGRP expression correlate with alterations in behavioural responses. Our findings suggest that nontoxic proteasome inhibitors may represent a novel pharmacotherapy for osteoarthritis.
Collapse
Affiliation(s)
- Aisha Siddiqah Ahmed
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Karolinska University Hospital, Solna, Stockholm 17176, Sweden Department of Medicine, Centre for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Solna, Stockholm 17176, Sweden Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm 18288, Sweden Department of Pharmaceutical Biosciences, Uppsala University, Uppsala 75105, Sweden Department of Neurobiology, Care Sciences and Society, Center for Family and Community Medicine, Karolinska Institutet, Huddinge 14183, Sweden
| | | | | | | | | | | |
Collapse
|
27
|
St-Jacques B, Ma W. Role of prostaglandin E2 in the synthesis of the pro-inflammatory cytokine interleukin-6 in primary sensory neurons: an in vivo and in vitro study. J Neurochem 2011; 118:841-54. [PMID: 21371033 DOI: 10.1111/j.1471-4159.2011.07230.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Following various types of nerve injury, cyclooxygenase 2 and prostaglandin E2 (PGE2) are universally and chronically up-regulated in injured nerves and contribute to the genesis of neuropathic pain. Persistent high levels of PGE2 likely exert chronic effects on nociceptive dorsal root ganglion (DRG) neurons. In the present study, we tested the hypothesis that injured nerve-derived PGE2 contributes to the up-regulation of the pro-inflammatory cytokine interleukin-6 (IL-6) in DRG neurons following partial sciatic nerve ligation. In naive adult rats, IL-6 was expressed in only a few small size DRG neurons which all co-expressed EP4 receptors. Partial sciatic nerve ligation increased and shifted IL-6 expression from small to medium and large size damaged DRG neurons. Perineural injection of a selective cyclooxygenase 2 inhibitor or a selective EP4 receptor antagonist significantly suppressed the up-regulation of IL-6 in DRG, suggesting that injured nerve derived PGE2 contributes to the de novo synthesis of IL-6 in DRG neurons through EP4 receptors. In cultured sensory ganglion explants, a stabilized PGE2 analog increased IL-6 mRNA and protein levels through the activation of EP4, protein kinase A, protein kinase C, extracellular regulated protein kinase/MAPK, cAMP response element binding protein and NFκB signalling pathways. Taken together, these data indicate that facilitating the de novo synthesis of pain-related cytokines in injured medium and large size DRG neurons is a novel mechanism underlying the role of injured nerve derived PGE2 in the genesis of neuropathic pain.
Collapse
Affiliation(s)
- Bruno St-Jacques
- Douglas Mental Health University Institute, McGill University, Montreal, Quebec, Canada
| | | |
Collapse
|
28
|
Dai JN, Zong Y, Zhong LM, Li YM, Zhang W, Bian LG, Ai QL, Liu YD, Sun J, Lu D. Gastrodin inhibits expression of inducible NO synthase, cyclooxygenase-2 and proinflammatory cytokines in cultured LPS-stimulated microglia via MAPK pathways. PLoS One 2011; 6:e21891. [PMID: 21765922 PMCID: PMC3134470 DOI: 10.1371/journal.pone.0021891] [Citation(s) in RCA: 180] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2011] [Accepted: 06/08/2011] [Indexed: 01/10/2023] Open
Abstract
Background Microglial activation plays an important role in neurodegenerative diseases by producing several proinflammatory enzymes and proinflammatory cytokines. The phenolic glucoside gastrodin, a main constituent of a Chinese herbal medicine, has been known to display anti-inflammatory properties. The current study investigates the potential mechanisms whereby gastrodin affects the expression of potentially pro-inflammatory proteins by cultured murine microglial BV-2 cells stimulated with lipopolysaccharide (LPS). Methodology/Principal Findings BV-2 cells were pretreated with gastrodin (30, 40, and 60 µM) for 1 h and then stimulated with LPS (1 µg/ml) for another 4 h. The effects on proinflammatory enzymes, inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2), and proinflammatory cytokines, tumor necrosis factor-α (TNF-α), and interleukin-1β (IL-1β), are analysed by double-immunofluorescence labeling and RT-PCR assay. To reveal the mechanisms of action of gastrodin we investigated the involvement of mitogen-activated protein kinases (MAPKs) cascades and their downstream transcription factors, nuclear factor-κB (NF-κB) and cyclic AMP-responsive element (CRE)-binding protein (CREB). Gastrodin significantly reduced the LPS-induced protein and mRNA expression levels of iNOS, COX-2, TNF-α, IL-1β and NF-κB. LPS (1 µg/ml, 30 min)-induced phosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2), c-Jun N-terminal protein kinase (JNK) and p38 mitogen-activated protein kinase (p38 MAPK) and this was inhibited by pretreatment of BV-2 cells with different concentrations of gastrodin (30, 40, and 60 µM). In addition, gastrodin blocked LPS-induced phosphorylation of inhibitor κB-α (IκB-α) (and hence the activation of NF-κB) and of CREB, respectively. Conclusion and Implications This study indicates that gastrodin significantly attenuate levels of neurotoxic proinflammatory mediators and proinflammatory cytokines by inhibition of the NF-κB signaling pathway and phosphorylation of MAPKs in LPS-stimulated microglial cells. Arising from the above, we suggest that gastrodin has a potential as an anti-inflammatory drug candidate in neurodegenerative diseases.
Collapse
Affiliation(s)
- Ji-Nan Dai
- Department of Anatomy, Kunming Medical University, Kunming, Yunnan, China
| | - Yi Zong
- Department of Anatomy, Kunming Medical University, Kunming, Yunnan, China
| | - Lian-Mei Zhong
- Department of Neurology, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Yue-Min Li
- Department of Anatomy, Kunming Medical University, Kunming, Yunnan, China
| | - Wei Zhang
- Department of Neurology, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Li-Gong Bian
- Department of Anatomy, Kunming Medical University, Kunming, Yunnan, China
| | - Qing-Long Ai
- Department of Neurology, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Yi-Dan Liu
- Kunming Pharmaceutical Corporation, Kunming, Yunnan, China
| | - Jun Sun
- Department of Anatomy, Kunming Medical University, Kunming, Yunnan, China
- * E-mail: (JS); (DL)
| | - Di Lu
- Department of Anatomy, Kunming Medical University, Kunming, Yunnan, China
- Rehabilitation Engineering Research Laboratory, Biomedicine Engineering Research Centre, Kunming Medical University, Kunming, Yunnan, China
- * E-mail: (JS); (DL)
| |
Collapse
|
29
|
Roet KCD, Bossers K, Franssen EHP, Ruitenberg MJ, Verhaagen J. A meta-analysis of microarray-based gene expression studies of olfactory bulb-derived olfactory ensheathing cells. Exp Neurol 2011; 229:10-45. [PMID: 21396936 DOI: 10.1016/j.expneurol.2011.03.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2010] [Revised: 12/28/2010] [Accepted: 03/02/2011] [Indexed: 12/23/2022]
Abstract
Genome wide transcriptional profiling and large scale proteomics have emerged as two powerful methods to dissect the molecular properties of specific neural tissues or cell types on a global scale. Several genome-wide transcriptional profiling and proteomics studies have been published on cultured olfactory ensheathing cells (OEC). In this article we present a meta-analysis of all five published and publicly available micro-array gene expression datasets of cultured early-passage-OB-OEC with other cell types (Schwann cells, late-passage-OB-OEC, mucosa-OEC, an OEC cell line, and acutely dissected OEC). The aim of this meta-analysis is to identify genes and molecular pathways that are found in multiple instead of one isolated study. 454 Genes were detected in at least three out of five microarray datasets. In this "Top-list", genes involved in the biological processes "growth of neurites", "blood vessel development", "migration of cells" and "immune response" were strongly overrepresented. By applying network analysis tools, molecular networks were constructed and Hub-genes were identified that may function as key genes in the above mentioned interrelated processes. We also identified 7 genes (ENTPD2, MATN2, CTSC, PTHLH, GLRX1, COL27A1 and ID2) with uniformly higher or lower expression in early-passage-OB-OEC in all five microarray comparisons. These genes have diverse but intriguing roles in neuroprotection, neurite extension and/or tissue repair. Our meta-analysis provides novel insights into the molecular basis of OB-OEC-mediated neural repair and can serve as a repository for investigators interested in the molecular biology of OEC. This article is part of a Special Issue entitled: Understanding olfactory ensheathing glia and their prospect for nervous system repair.
Collapse
Affiliation(s)
- Kasper C D Roet
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, Meibergdreef 47, Amsterdam, The Netherlands.
| | | | | | | | | |
Collapse
|
30
|
Kim JM, Song JS, Cho HH, Shin KK, Bae YC, Lee BJ, Jung JS. Effect of the modulation of leucine zipper tumor suppressor 2 expression on proliferation of various cancer cells functions as a tumor suppressor. Mol Cell Biochem 2010; 346:125-36. [PMID: 20890637 DOI: 10.1007/s11010-010-0599-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2010] [Accepted: 09/18/2010] [Indexed: 11/30/2022]
Abstract
β-catenin is a component of the adhesion complex linking cadherin and actin cytoskeleton, as well as a major mediator of the Wnt pathway, which is a critical signal cascade regulating embryonic development, cell polarity, carcinogenesis, and stem cell function. NF-κB functions as a key regulator of immune responses and apoptosis, and mutations in NF-κB signaling can lead to immune diseases and cancers. We previously showed that NF-κB-mediated modulation of β-catenin/Tcf signaling is mediated by leucine zipper tumor suppressor 2 (Lzts2) and that lzts2 expression is differentially regulated in various cancer cells. Its functional significances, however, are poorly understood. We showed that NF-κB-induced modulation of β-catenin/Tcf pathway is regulated by lzts2 expression in mesenchymal stem cells (MSCs) and several cancer cells, and that NF-κB-induced lzts2 expression is differentially regulated among cancer cell types. Here, using a promoter-reporter assay and EMSA, we demonstrate that NF-κB regulates lzts2 transcription by directly binding to the lzts2 promoter, and that NF-κB-induced lzts2 transcription differs by cell types. Modulation of lzts2 expression by lentiviral techniques affected proliferation and tumorigenicity of several cancer cell lines such as breast, colon, prostate cancer, and glioma, but did not affect cisplatin sensitivity or cell migration. Our data indicate that lzts2 expression is transcriptionally regulated by NF-κB activities, and the modulation of lzts2 expression affects cell proliferation and tumor growth through the Wnt/β-catenin pathway in various cancer cell lines.
Collapse
Affiliation(s)
- Jong Myung Kim
- MD-PhD Program, School of Medicine, Pusan National University, Yangsan, Korea
| | | | | | | | | | | | | |
Collapse
|
31
|
Pigment epithelium-derived factor stimulates tumor macrophage recruitment and is downregulated by the prostate tumor microenvironment. Neoplasia 2010; 12:336-45. [PMID: 20360944 DOI: 10.1593/neo.92046] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2009] [Revised: 02/08/2010] [Accepted: 02/08/2010] [Indexed: 01/07/2023] Open
Abstract
Pigment epithelium-derived factor (PEDF) is a potent inhibitor of angiogenesis but whether it has additional effects on the tumor microenvironment is largely unexplored. We show that overexpression of PEDF in orthotopic MatLyLu rat prostate tumors increased tumor macrophage recruitment. The fraction of macrophages expressing inducible nitric oxide synthase, a marker of cytotoxic M1 macrophages, was increased, suggesting that PEDF could enhance antitumor immunity. In addition, PEDF overexpression reduced vascular growth both in the tumor and in the surrounding normal tissue, slowed tumor growth, and decreased lymph node metastasis. Contrary, extratumoral lymphangiogenesis was increased. PEDF expression is, for reasons unknown, often decreased or lost during prostate tumor progression. When AT-1 rat prostate tumor cells, expressing high levels of PEDF messenger RNA (mRNA) and protein, were injected into the prostate, PEDF is markedly downregulated, suggesting that factors in the microenvironment suppressed its expression. One such factor could be macrophage-derived tumor necrosis factor alpha (TNFalpha). A fraction of the accumulating macrophages expressed TNFalpha, and TNFalpha treatment downregulated the expression of PEDF protein and mRNA in prostate AT-1 tumor cells in vitro and in the rat ventral prostate in vivo. PEDF apparently has multiple effects in prostate tumors: it suppresses angiogenesis and metastasis, but it also causes macrophage accumulation. Accumulating macrophages may inhibit tumor growth, but they may also suppress PEDF and enhance lymph angiogenesis and, in this way, eventually enhance tumor growth.
Collapse
|
32
|
Adenoviral Gene Delivery of Pigment Epithelium-Derived Factor Protects Striatal Neurons from Quinolinic Acid-Induced Excitotoxicity. J Neuropathol Exp Neurol 2010; 69:224-33. [DOI: 10.1097/nen.0b013e3181cfc46f] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
|
33
|
Liu Y, Teng X, Yang X, Song Q, Lu R, Xiong J, Liu B, Zeng N, Zeng Y, Long J, Cao R, Lin Y, He Q, Chen P, Lu M, Liang S. Shotgun Proteomics and Network Analysis between Plasma Membrane and Extracellular Matrix Proteins from Rat Olfactory Ensheathing Cells. Cell Transplant 2010; 19:133-46. [PMID: 20350363 DOI: 10.3727/096368910x492607] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Olfactory ensheathing cells (OECs) are a special type of glial cells that have characteristics of both astrocytes and Schwann cells. Evidence suggests that the regenerative capacity of OECs is induced by soluble, secreted factors that influence their microenvironment. These factors may regulate OECs self-renewal and/or induce their capacity to augment spinal cord regeneration. Profiling of plasma membrane and extracellular matrix through a high-throughput expression proteomics approach was undertaken to identify plasma membrane and extracellular matrix proteins of OECs under serum-free conditions. 1D-shotgun proteomics followed with gene ontology (GO) analysis was used to screen proteins from primary culture rat OECs. Four hundred and seventy nonredundant plasma membrane proteins and 168 extracellular matrix proteins were identified, the majority of which were never before reported to be produced by OECs. Furthermore, plasma membrane and extracellular proteins were classified based on their protein–protein interaction predicted by STRING quantitatively integrates interaction data. The proteomic profiling of the OECs plasma membrane proteins and their connection with the secretome in serum-free culture conditions provides new insights into the nature of their in vivo microenvironmental niche. Proteomic analysis for the discovery of clinical biomarkers of OECs mechanism warrants further study.
Collapse
Affiliation(s)
- Yisong Liu
- Key laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, P.R. China
| | - Xiaohua Teng
- Department of Neurosurgery, Second Affiliated Hospital of Hunan Normal University (163 Hospital of PLA), Changsha, P.R. China
| | - Xiaoxu Yang
- Key laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, P.R. China
| | - Qing Song
- Key laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, P.R. China
| | - Rong Lu
- Key laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, P.R. China
| | - Jixian Xiong
- Key laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, P.R. China
| | - Bo Liu
- Department of Neurosurgery, Second Affiliated Hospital of Hunan Normal University (163 Hospital of PLA), Changsha, P.R. China
| | - Nianju Zeng
- Department of Neurosurgery, Second Affiliated Hospital of Hunan Normal University (163 Hospital of PLA), Changsha, P.R. China
| | - Yu Zeng
- Department of Neurosurgery, Second Affiliated Hospital of Hunan Normal University (163 Hospital of PLA), Changsha, P.R. China
| | - Jia Long
- Key laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, P.R. China
| | - Rui Cao
- Key laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, P.R. China
| | - Yong Lin
- Key laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, P.R. China
| | - Quanze He
- Key laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, P.R. China
| | - Ping Chen
- Key laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, P.R. China
| | - Ming Lu
- Department of Neurosurgery, Second Affiliated Hospital of Hunan Normal University (163 Hospital of PLA), Changsha, P.R. China
| | - Songping Liang
- Key laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, P.R. China
| |
Collapse
|
34
|
Andreu-Agulló C, Morante-Redolat JM, Delgado AC, Fariñas I. Vascular niche factor PEDF modulates Notch-dependent stemness in the adult subependymal zone. Nat Neurosci 2009; 12:1514-23. [PMID: 19898467 DOI: 10.1038/nn.2437] [Citation(s) in RCA: 159] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2009] [Accepted: 09/17/2009] [Indexed: 12/28/2022]
Abstract
We sought to address the fundamental question of how stem cell microenvironments can regulate self-renewal. We found that Notch was active in astroglia-like neural stem cells (NSCs), but not in transit-amplifying progenitors of the murine subependymal zone, and that the level of Notch transcriptional activity correlated with self-renewal and multipotency. Moreover, dividing NSCs appeared to balance renewal with commitment via controlled segregation of Notch activity, leading to biased expression of known (Hes1) and previously unknown (Egfr) Notch target genes in daughter cells. Pigment epithelium-derived factor (PEDF) enhanced Notch-dependent transcription in cells with low Notch signaling, thereby subverting the output of an asymmetrical division to the production of two highly self-renewing cells. Mechanistically, PEDF induced a non-canonical activation of the NF-kappaB pathway, leading to the dismissal of the transcriptional co-repressor N-CoR from specific Notch-responsive promoters. Our data provide a basis for stemness regulation in vascular niches and indicate that Notch and PEDF cooperate to regulate self-renewal.
Collapse
Affiliation(s)
- Celia Andreu-Agulló
- Departamento de Biología Celular and CIBER en Enfermedades Neurodegenerativas, Universidad de Valencia, Burjassot, Spain
| | | | | | | |
Collapse
|
35
|
Zhang P, Katz J, Michalek SM. Glycogen synthase kinase-3beta (GSK3beta) inhibition suppresses the inflammatory response to Francisella infection and protects against tularemia in mice. Mol Immunol 2009; 46:677-87. [PMID: 18929413 PMCID: PMC3033759 DOI: 10.1016/j.molimm.2008.08.281] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2008] [Accepted: 08/27/2008] [Indexed: 12/01/2022]
Abstract
Francisella tularensis, the causative agent of tularemia, is currently considered a category A bioterrorism agent due to its high virulence. Infection with F. tularensis results in an inflammatory response that plays an important role in the pathogenesis of the disease; however, the cellular mechanisms regulating this response are poorly understood. Glycogen synthase kinase-3beta (GSK3beta) is a serine/threonine protein kinase that has recently emerged as a key regulatory switch in the modulation of the inflammatory response. In this study, we investigated the effect of GSK3beta inhibition in regulating F. tularensis LVS-induced inflammatory responses. F. tularensis LVS infection of murine peritoneal macrophages induced a TLR2 dependent phosphorylation of GSK3beta. Inhibition of GSK3beta resulted in a significant decrease in the production of pro-inflammatory cytokine IL-6, IL-12p40 and TNF-alpha, as well as a significant increase in the production of the anti-inflammatory cytokine IL-10. GSK3beta regulated the F. tularensis LVS-induced cytokine response by differentially affecting the activation of transcription factors NF-kappaB and CREB. Inhibition of GSK3beta by lithium in vivo suppressed the inflammatory response in mice infected with F. tularensis LVS and conferred a survival advantage. In addition, we show that the production of IFN-gamma contributed to the development of tularemia and to the fatal outcome of the infected animals, depending on the timing and the relative level of the IFN-gamma produced. IFN-gamma potentiated F. tularensis LVS-induced cytokine production by increasing GSK3beta activity and the nuclear translocation of NF-kappaB. Taken together, these results demonstrate a regulatory function of GSK3beta in modulating inflammatory responses that can be detrimental to the host during an F. tularensis LVS infection, and suggest that inhibition of GSK3beta may represent a novel therapeutic approach in the treatment of tularemia.
Collapse
Affiliation(s)
- Ping Zhang
- Department of Pediatric Dentistry, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Jenny Katz
- Department of Pediatric Dentistry, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Suzanne M. Michalek
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| |
Collapse
|
36
|
Sanagi T, Yabe T, Yamada H. Gene transfer of PEDF attenuates ischemic brain damage in the rat middle cerebral artery occlusion model. J Neurochem 2008; 106:1841-54. [DOI: 10.1111/j.1471-4159.2008.05529.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
37
|
Glutamate-induced c-Jun expression in neuronal PC12 cells: the effects of ketamine and propofol. J Neurosurg Anesthesiol 2008; 20:124-30. [PMID: 18362774 DOI: 10.1097/ana.0b013e3181667c27] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Transcription factor c-Jun affects neuronal cell death and survival in mammalian brain. As general anesthetics, such as ketamine and propofol, are thought to provide some degree of neuroprotection, this study was intended to test whether the protection of injured neuronal PC12 cells by ketamine and propofol is related to the inhibition of phospho-c-Jun. Using neuronal PC12 cells from rat pheochromocytoma cells differentiated with nerve growth factor, we found that 24 hours of exposure to glutamate (1 to 100 mM) induced concentration-dependent cell death as determined by an ability to reduce the tetrazolium derivative, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) into a blue formazan salt. Neuronal PC12 cells were exposed to ketamine (0.1, 1.0 mM) or propofol (0.5, 5.0 microM) and glutamate (0, 20 mM) for 24 hours. Cell injury was assessed using MTT, in situ terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate-biotin nick end labeling, and c-Jun activity assay. Glutamate, 20 mM, induced about 70% of cell death as determined by MTT and terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate-biotin nick end labeling staining. Glutamate-induced cell death was related to an increase in expression of phospho-c-Jun. Glutamate-induced cell death was reduced by ketamine (0.1, 1.0 mM) in a dose-dependent manner and also by propofol (0.5, 5.0 microM). In addition, the expression of phospho-c-Jun was substantially reduced by ketamine (0.1, 1.0 mM) and propofol (0.5, 5.0 microM), respectively, as determined by Western blot assay. These results suggest that inhibition of c-Jun activity is involved in the neuroprotective effects of ketamine and propofol on glutamate-induced injury in neuronal PC12 cells.
Collapse
|
38
|
Sanagi T, Yabe T, Yamada H. Changes in pigment epithelium-derived factor expression following kainic acid induced cerebellar lesion in rat. Neurosci Lett 2007; 424:66-71. [PMID: 17709187 DOI: 10.1016/j.neulet.2007.07.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2007] [Revised: 06/16/2007] [Accepted: 07/15/2007] [Indexed: 11/16/2022]
Abstract
Pigment epithelium-derived factor (PEDF) is a potent and broad-acting neurotrophic factor that protects various types of cultured neurons against glutamate excitotoxicity and induced apoptosis. The expression pattern and functions of PEDF in the central nervous system (CNS) remain largely undetermined. In this study, we analyzed the spatial and temporal expression of PEDF in normal and kainic acid (KA)-induced lesioned rat cerebellum using immunoblotting, immunohistochemistry and fluorescent in situ hybridization techniques. In normal rat cerebellum, PEDF protein and mRNA were mostly confined and co-localized with calbindin-positive cells in the Purkinje cell layer of the cerebellum, but not with glial fibrillary acidic protein (GFAP)-, 2', 3'-cyclic nucleotide 3'-phosphodiesterase (CNPase)-, and isolectin B4-positive cells. Injection of KA into the right cellebellum caused severe loss of calbindin-positive Purkinje neurons, and an increased number of GFAP-positive astrocytes and isolectin B4-positive microglia was observed on the ipsilateral side of the lesioned cerebellum. Although the PEDF level on the ipsilateral side of the cerebellum was dramatically decreased 2 days after KA treatment, significantly elevation of PEDF levels was observed at 7 days. In agreement with these results, PEDF protein and PEDF mRNA expression were co-localized with GFAP-positive reactive astrocytes in the ipsilateral side 7 days after KA treatment. Although the mechanism by which PEDF is induced in reactive astrocytes remains unclear, the increase in PEDF expression in injured brain may form part of a compensation mechanism against neuronal degeneration.
Collapse
Affiliation(s)
- Tomomi Sanagi
- Kitasato Institute for Life Sciences, Kitasato University, Tokyo, Japan
| | | | | |
Collapse
|
39
|
Bagnato C, Thumar J, Mayya V, Hwang SI, Zebroski H, Claffey KP, Haudenschild C, Eng JK, Lundgren DH, Han DK. Proteomics analysis of human coronary atherosclerotic plaque: a feasibility study of direct tissue proteomics by liquid chromatography and tandem mass spectrometry. Mol Cell Proteomics 2007; 6:1088-102. [PMID: 17339633 DOI: 10.1074/mcp.m600259-mcp200] [Citation(s) in RCA: 130] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cardiovascular disease presents significant variations in human populations with respect to the atherosclerotic plaque progression, inflammation, thrombosis, and rupture. To gain a more comprehensive picture of the pathogenic mechanism of atherosclerosis and the variations seen in patients, efficient methods to identify proteins from the normal and diseased arteries need to be developed. To accomplish this goal, we tested the feasibility and efficiency of protein identification by a recently developed method, termed direct tissue proteomics (DTP). We analyzed frozen and paraformaldehyde-fixed archival coronary arteries with the DTP method. We also validated the distinct expression of four proteins by immunohistochemistry. In addition, we demonstrated the compatibility of the DTP method with laser capture microdissection and the possibility of monitoring specific cytokines and growth factors by the absolute quantification of abundance method. Major findings from this feasibility study are that 1) DTP can be used to efficiently identify proteins from paraformaldehyde-fixed, paraffin-embedded, and frozen coronary arteries; 2) approximately twice the number of proteins were identified from the frozen sections when compared with the paraformaldehyde-fixed sections; 3) laser capture microdissection is compatible with DTP; and 4) detection of low abundance cytokines and growth factors in the coronary arteries required selective reaction monitoring experiments coupled to absolute quantification of abundance. The analysis of 35 human coronary atherosclerotic samples allowed identification of a total of 806 proteins. The present study provides the first large scale proteomics map of human coronary atherosclerotic plaques.
Collapse
Affiliation(s)
- Carolina Bagnato
- Department of Cell Biology, Center for Vascular Biology, University of Connecticut School of Medicine, Farmington, Connecticut 06030, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Mhyre AJ, Shapiro RA, Dorsa DM. Estradiol reduces nonclassical transcription at cyclic adenosine 3',5'-monophosphate response elements in glioma cells expressing estrogen receptor alpha. Endocrinology 2006; 147:1796-804. [PMID: 16439453 DOI: 10.1210/en.2005-1316] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Estradiol can protect the brain from a variety of insults by activating membrane-initiated signaling pathways, and thereby modulate gene expression and lead to functional changes in neurons. These direct neuronal effects of the hormone have been well documented; however, it is less understood what effects estradiol may have on nonneuronal cells of the central nervous system. There is evidence that estradiol levels can induce the release of glial-derived growth factors and other cytokines, suggesting that estradiol may both directly and indirectly protect neurons. To determine whether 17beta-estradiol (E2) can activate rapid signaling and modulate nonclassical transcription in astrocytes, we stably transfected the C6 rat glioblastoma cell line with human estrogen receptor (ER) alpha (C6ERalpha) or rat ERbeta (C6ERbeta). Introduction of a cAMP response element-luciferase reporter gene into C6, C6ERalpha, and C6ERbeta cells leads to the observation that E2 treatment reduced isoproterenol-stimulated luciferase activity by 35% in C6ERalpha but had no effect on reporter gene expression in C6ERbeta or untransfected C6 cells. A similar effect was seen with a membrane-impermeable estrogen (E2-BSA), suggesting the modulation of nonclassical transcription by estradiol treatment is mediated by the activation of a membrane-initiated signaling pathway. Furthermore, pretreatment with wortmannin (phosphatidylinsositol 3-kinase) or U73122 (phospholipase C) attenuated the E2-induced reduction in nonclassical transcription. We conclude that E2 treatment reduces cAMP response element-mediated transcription in glioma cells expressing ERalpha and that this reduction is dependent on the activation of membrane-initiated signaling. These findings suggest a novel model of estrogen rapid signaling in astrocytes that leads to modulation of nonclassical transcription.
Collapse
Affiliation(s)
- Andrew J Mhyre
- Department of Pharmacology, University of Washington School of Medicine, Seattle, 98195, USA.
| | | | | |
Collapse
|
41
|
Chen YD, Zhang YB, Zhu R, Zhang FT, Jiang J, Shi Y, Zhang QY, Chen SL, Gui JF. Inductive expression and characterization analysis of Paralichthys olivaceus pigment epithelium-derived factor in a virally infected cell line. Biochem Biophys Res Commun 2005; 335:799-809. [PMID: 16098479 DOI: 10.1016/j.bbrc.2005.07.143] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2005] [Accepted: 07/22/2005] [Indexed: 11/17/2022]
Abstract
Pigment epithelium-derived factor (PEDF) is acknowledged to be a non-inhibitory member of the serine protease inhibitor (serpin) superfamily, with antiangiogenesis, and neuroprotective and immunoregulatory function, mainly in the tissues of nervous system. Here, A PEDF gene homolog, Paralichthys olivaceus PEDF (PoPEDF), was isolated from flounder embryonic cells (FEC) treated with UV-inactivated Grass carp hemorrhage virus (GCHV) and subsequently identified as a differentially expressed gene. The full length of PoPEDF cDNA is 1803bp with an open reading frame of 1212bp encoding a 403-amino-acid protein. This deduced protein contains an N-terminal signal peptide, a glycosylation site, a consensus serpin motif, and a 34-mer and a 44-mer fragment, all of which are very conserved in the PEDF family. PoPEDF gene exhibits a conserved exon-intron arrangement with 8 exons and 7 introns. This conserved evolutionary relationship was further confirmed by a phylogenetic analysis, where fish PEDFs and mammalian members formed a well-supported clade. Constitutive expression of PoPEDF was widely detected in many tissues. In response to UV-inactivated GCHV or poly(I:C), PEDF mRNA was upregulated in FEC cells with time. This is the first report on the transcriptional induction of PEDF in virally infected cells.
Collapse
Affiliation(s)
- Yu-Dong Chen
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Graduate School of Chinese Academy of Sciences, Wuhan 430072, China
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Yabe T, Kanemitsu K, Sanagi T, Schwartz JP, Yamada H. Pigment epithelium-derived factor induces pro-survival genes through cyclic AMP-responsive element binding protein and nuclear factor kappa B activation in rat cultured cerebellar granule cells: Implication for its neuroprotective effect. Neuroscience 2005; 133:691-700. [PMID: 15893882 DOI: 10.1016/j.neuroscience.2005.03.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2004] [Revised: 02/28/2005] [Accepted: 03/09/2005] [Indexed: 11/21/2022]
Abstract
Pigment epithelium-derived factor (PEDF) protects immature cerebellar granule cell neurons (CGCs) against apoptosis induced by K+ and serum deprivation. However, the precise mechanism of this protection remains unknown. We recently reported that the transcription factor nuclear factor kappa B (NF-kappaB) is activated in PEDF-treated CGCs. Although it is well known that NF-kappaB blocks apoptotic cell death through the induction of pro-survival factors, the effects of PEDF on the expression of these factors are not fully understood. In this study, we employed the use of reverse transcriptase-polymerase chain reaction to analyze the gene expression of certain pro-survival genes and found that genes such as c-IAP1, c-IAP2, FLIPs, A1/Bfl-1 and Mn-SOD were induced in PEDF-treated neurons. On the other hand, no induction was observed of the pro-apoptotic Bcl-2 family members Bax and Bid at any time from 3 to 24 h following PEDF addition. Furthermore, phosphorylation of cyclic AMP-responsive element binding protein (CREB) and increment of nuclear cyclic AMP-response element (CRE)-like DNA binding were observed in PEDF-treated CGCs. The anti-apoptotic effect of PEDF was blocked by overexpression of dominant negative CREB or a mutated form of IkappaBalpha. These results suggested that induction of both CRE- and NF-kappaB-dependent genes is required for the observed neuroprotective effects of PEDF on CGCs.
Collapse
Affiliation(s)
- T Yabe
- Kitasato Institute for Life Sciences, Kitasato University, 5-9-1, Shirokane, Tokyo 108-8641, Japan.
| | | | | | | | | |
Collapse
|