101
|
Jiang X, Wang X, Tuo M, Ma J, Xie A. RAGE and its emerging role in the pathogenesis of Parkinson’s disease. Neurosci Lett 2018; 672:65-69. [DOI: 10.1016/j.neulet.2018.02.049] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 02/20/2018] [Accepted: 02/21/2018] [Indexed: 01/10/2023]
|
102
|
Fardell C, Zettergren A, Ran C, Carmine Belin A, Ekman A, Sydow O, Bäckman L, Holmberg B, Dizdar N, Söderkvist P, Nissbrandt H. S100B polymorphisms are associated with age of onset of Parkinson's disease. BMC MEDICAL GENETICS 2018. [PMID: 29529989 PMCID: PMC5848451 DOI: 10.1186/s12881-018-0547-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Background In this study we investigated the association between SNPs in the S100B gene and Parkinson’s disease (PD) in two independent Swedish cohorts. The SNP rs9722 has previously been shown to be associated with higher S100B concentrations in serum and frontal cortex in humans. S100B is widely expressed in the central nervous system and has many functions such as regulating calcium homeostasis, inflammatory processes, cytoskeleton assembly/disassembly, protein phosphorylation and degradation, and cell proliferation and differentiation. Several of these functions have been suggested to be of importance for the pathophysiology of PD. Methods The SNPs rs9722, rs2239574, rs881827, rs9984765, and rs1051169 of the S100B gene were genotyped using the KASPar® PCR SNP genotyping system in a case-control study of two populations (431 PD patients and 465 controls, 195 PD patients and 378 controls, respectively). The association between the genotype and allelic distributions and PD risk was evaluated using Chi-Square and Cox proportional hazards test, as well as logistic regression. Linear regression and Cox proportional hazards tests were applied to assess the effect of the rs9722 genotypes on age of disease onset. Results The S100B SNPs tested were not associated with the risk of PD. However, in both cohorts, the T allele of rs9722 was significantly more common in early onset PD patients compared to late onset PD patients. The SNP rs9722 was significantly related to age of onset, and each T allele lowered disease onset with 4.9 years. In addition, allelic variants of rs881827, rs9984765, and rs1051169, were significantly more common in early-onset PD compared to late-onset PD in the pooled population. Conclusions rs9722, a functional SNP in the 3’-UTR of the S100B gene, was strongly associated with age of onset of PD.
Collapse
Affiliation(s)
- Camilla Fardell
- Department of Pharmacology, Sahlgrenska Academy at the University of Gothenburg, P.O. Box 431, 405 30, Gothenburg, Sweden.
| | - Anna Zettergren
- Department of Pharmacology, Sahlgrenska Academy at the University of Gothenburg, P.O. Box 431, 405 30, Gothenburg, Sweden
| | - Caroline Ran
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | | - Agneta Ekman
- Department of Pharmacology, Sahlgrenska Academy at the University of Gothenburg, P.O. Box 431, 405 30, Gothenburg, Sweden
| | - Olof Sydow
- Department of Clinical Neuroscience, Karolinska University Hospital, Stockholm, Sweden
| | - Lars Bäckman
- Aging Research Center, Karolinska Institutet, Stockholm, Sweden
| | - Björn Holmberg
- Department of Clinical Neuroscience and Rehabilitation, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Nil Dizdar
- Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden.,Department of Neurology, Linköping University Hospital, Linköping University, Linköping, Sweden
| | - Peter Söderkvist
- Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden
| | - Hans Nissbrandt
- Department of Pharmacology, Sahlgrenska Academy at the University of Gothenburg, P.O. Box 431, 405 30, Gothenburg, Sweden
| |
Collapse
|
103
|
Mohammadzadeh F, Tsoporis JN, Izhar S, Desjardins JF, Parker TG. Deficiency of S100B confers resistance to experimental diabetes in mice. Exp Cell Res 2018; 365:129-137. [PMID: 29499206 DOI: 10.1016/j.yexcr.2018.02.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 02/07/2018] [Accepted: 02/23/2018] [Indexed: 11/16/2022]
Abstract
The calcium binding protein S100B has been implicated in diabetic neuronal and vascular complications but has not been examined in the development of diabetes. S100B knock out (S100B KO) and wild-type (WT) mice were injected with 40 mg/kg body weight streptozotocin (STZ) for 5 days. Blood and pancreatic tissue samples were obtained to examine islet structure and function, the profile of glucose and insulin and expression of glucose transporter 2 (Glut2), S100B and its receptor, the receptor for advanced glycation end products (RAGE). Primary islet β-cells cultures from WT mice were used to test the apoptotic potential of S100B. S100B KO mice were resistant to STZ induced-diabetes with lower urine volume, food and water intake compared to WT mice. S100B increased in the WT islet following diabetes but did not co-localize with beta or peri-islet Schwann cells but with CD3 + T lymphocytes. S100B KO mice exhibited enhanced glucose tolerance, insulin sensitivity, prevented β-cell destruction and functional impairment in response to STZ treatment. S100B deficiency was associated with decreased Glut2 and RAGE. In primary β-cell cultures from WT mice, S100B induced reactive oxygen species (ROS) and RAGE-dependent apoptosis. In the STZ diabetic animal model, abrogation of S100B enhances insulin sensitivity and reduces pancreatic islet, and β-cell destruction. S100B may be a promising target for pharmacological interventions aimed at repressing diabetes.
Collapse
Affiliation(s)
- Forough Mohammadzadeh
- Division of Cardiology, Department of Medicine, Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael's Hospital, University of Toronto, Ontario, Canada
| | - James N Tsoporis
- Division of Cardiology, Department of Medicine, Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael's Hospital, University of Toronto, Ontario, Canada.
| | - Shehla Izhar
- Division of Cardiology, Department of Medicine, Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael's Hospital, University of Toronto, Ontario, Canada
| | - Jean-Francois Desjardins
- Division of Cardiology, Department of Medicine, Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael's Hospital, University of Toronto, Ontario, Canada
| | - Thomas G Parker
- Division of Cardiology, Department of Medicine, Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael's Hospital, University of Toronto, Ontario, Canada
| |
Collapse
|
104
|
Khan MI, Su YK, Zou J, Yang LW, Chou RH, Yu C. S100B as an antagonist to block the interaction between S100A1 and the RAGE V domain. PLoS One 2018; 13:e0190545. [PMID: 29444082 PMCID: PMC5812564 DOI: 10.1371/journal.pone.0190545] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 12/15/2017] [Indexed: 11/23/2022] Open
Abstract
Ca2+-binding human S100A1 protein is a type of S100 protein. S100A1 is a significant mediator during inflammation when Ca2+ binds to its EF-hand motifs. Receptors for advanced glycation end products (RAGE) correspond to 5 domains: the cytoplasmic, transmembrane, C2, C1, and V domains. The V domain of RAGE is one of the most important target proteins for S100A1. It binds to the hydrophobic surface and triggers signaling transduction cascades that induce cell growth, cell proliferation, and tumorigenesis. We used nuclear magnetic resonance (NMR) spectroscopy to characterize the interaction between S100A1 and the RAGE V domain. We found that S100B could interact with S100A1 via NMR 1H-15N HSQC titrations. We used the HADDOCK program to generate the following two binary complexes based on the NMR titration results: S100A1-RAGE V domain and S100A1-S100B. After overlapping these two complex structures, we found that S100B plays a crucial role in blocking the interaction site between RAGE V domain and S100A1. A cell proliferation assay WST-1 also supported our results. This report could potentially be useful for new protein development for cancer treatment.
Collapse
Affiliation(s)
- Md. Imran Khan
- National Tsing Hua University, Chemistry Department, Hsinchu, Taiwan
| | - Yu-Kai Su
- National Tsing Hua University, Chemistry Department, Hsinchu, Taiwan
| | - Jinhao Zou
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Lee-Wei Yang
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan
- Physics Division, National Center for Theoretical Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Ruey-Hwang Chou
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Center for Molecular Medicine, China Medical University Hospital, Taichung, Taiwan
- Department of Biotechnology, Asia University, Taichung, Taiwan
| | - Chin Yu
- National Tsing Hua University, Chemistry Department, Hsinchu, Taiwan
| |
Collapse
|
105
|
Impaired oligodendrogenesis and myelination by elevated S100B levels during neurodevelopment. Neuropharmacology 2018; 129:69-83. [DOI: 10.1016/j.neuropharm.2017.11.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 10/22/2017] [Accepted: 11/03/2017] [Indexed: 11/23/2022]
|
106
|
Hagmeyer S, Cristóvão JS, Mulvihill JJE, Boeckers TM, Gomes CM, Grabrucker AM. Zinc Binding to S100B Affords Regulation of Trace Metal Homeostasis and Excitotoxicity in the Brain. Front Mol Neurosci 2018; 10:456. [PMID: 29386995 PMCID: PMC5776125 DOI: 10.3389/fnmol.2017.00456] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 12/26/2017] [Indexed: 12/16/2022] Open
Abstract
Neuronal metal ions such as zinc are essential for brain function. In particular synaptic processes are tightly related to metal and protein homeostasis, for example through extracellular metal-binding proteins. One such protein is neuronal S100B, a calcium and zinc binding damage-associated molecular pattern (DAMP), whose chronic upregulation is associated with aging, Alzheimer’s disease (AD), motor neuron disease and traumatic brain injury (TBI). Despite gained insights on the structure of S100B, it remains unclear how its calcium and zinc binding properties regulate its function on cellular level. Here we report a novel role of S100B in trace metal homeostasis, in particular the regulation of zinc levels in the brain. Our results show that S100B at increased extracellular levels is not toxic, persists at high levels, and is taken up into neurons, as shown by cell culture and biochemical analysis. Combining protein bioimaging and zinc quantitation, along with a zinc-binding impaired S100B variant, we conclude that S100B effectively scavenges zinc ions through specific binding, resulting in a redistribution of the intracellular zinc pool. Our results indicate that scavenging of zinc by increased levels of S100B affects calcium levels in vitro. Thereby S100B is able to mediate the cross talk between calcium and zinc homeostasis. Further, we investigated a possible new neuro-protective role of S100B in excitotoxicity via its effects on calcium and zinc homeostasis. Exposure of cells to zinc-S100B but not the zinc-binding impaired S100B results in an inhibition of excitotoxicity. We conclude that in addition to its known functions, S100B acts as sensor and regulator of elevated zinc levels in the brain and this metal-buffering activity is tied to a neuroprotective role.
Collapse
Affiliation(s)
- Simone Hagmeyer
- WG Molecular Analysis of Synaptopathies, Department of Neurology, Neurocenter of Ulm University, Ulm, Germany.,Cellular Neurobiology and Neuro-Nanotechnology Lab, Department of Biological Sciences, University of Limerick, Limerick, Ireland.,Bernal Institute, University of Limerick, Limerick, Ireland
| | - Joana S Cristóvão
- Biosystems and Integrative Sciences Institute, Faculdade de Ciências, Universidade de Lisboa, and Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - John J E Mulvihill
- Bernal Institute, University of Limerick, Limerick, Ireland.,Health Research Institute (HRI), University of Limerick, Limerick, Ireland
| | - Tobias M Boeckers
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | - Cláudio M Gomes
- Biosystems and Integrative Sciences Institute, Faculdade de Ciências, Universidade de Lisboa, and Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - Andreas M Grabrucker
- Cellular Neurobiology and Neuro-Nanotechnology Lab, Department of Biological Sciences, University of Limerick, Limerick, Ireland.,Bernal Institute, University of Limerick, Limerick, Ireland.,Health Research Institute (HRI), University of Limerick, Limerick, Ireland
| |
Collapse
|
107
|
Kim ID, Lee H, Jin YC, Lee JK. Osteopontin Peptide Icosamer Containing RGD and SLAYGLR Motifs Enhances the Motility and Phagocytic Activity of Microglia. Exp Neurobiol 2017; 26:339-349. [PMID: 29302201 PMCID: PMC5746499 DOI: 10.5607/en.2017.26.6.339] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 11/16/2017] [Accepted: 12/01/2017] [Indexed: 11/19/2022] Open
Abstract
Osteopontin (OPN) is a secreted glycoprotein that is expressed in various tissues, including brain, and mediates a wide range of cellular activities. In a previous study, the authors observed the robust neuroprotective effects of recombinant OPN and of RGD and SLAYGLR-containing OPN-peptide icosamer (OPNpt20) in an animal model of transient focal ischemia, and demonstrated anti-inflammatory and pro-angiogenic effects of OPNpt20 in the postischemic brain. In the present study, we investigated the effects of OPNpt20 on the motility and phagocytic activity of BV2 cells (a microglia cell line). F-actin polymerization and cell motility were significantly enhanced in OPNpt20-treated BV2 cells, and numbers of filopodia-like processes increased and lamellipodia-like structures enlarged and thickened. In addition, treatment of cells with either of three mutant OPN icosamers containing mutation within RGD, SLAY, or RGDSLAY showed that the RGD and SLAY motifs of OPNpt20 play critical roles in the enhancement of cell motility, and the interaction between exogenous OPNpt20 and endogenous αv and α4 integrin and the activations of FAK, Erk, and Akt signaling pathways were found to be involved in the OPNpt20-mediated induction of cell motility. Furthermore, phagocytic activity of microglia was also significantly enhanced by OPNpt20 in a RGD and SLAY dependent manner. These results indicate OPNpt20 containing RGD and SLAY motifs triggers microglial motility and phagocytic activity and OPNpt20-integrin mediated signaling plays a critical role in these activities.
Collapse
Affiliation(s)
- Il-Doo Kim
- Department of Anatomy, Inha University School of Medicine, Incheon, Korea.,Medical Research Center, Inha University School of Medicine, Incheon, Korea
| | - Hahnbie Lee
- Department of Anatomy, Inha University School of Medicine, Incheon, Korea.,Medical Research Center, Inha University School of Medicine, Incheon, Korea
| | - Yin-Chuan Jin
- Department of Histology and Embryology, Binzhou Medical University, Yantai 264000, China
| | - Ja-Kyeong Lee
- Department of Anatomy, Inha University School of Medicine, Incheon, Korea.,Medical Research Center, Inha University School of Medicine, Incheon, Korea
| |
Collapse
|
108
|
Maguire G. Amyotrophic lateral sclerosis as a protein level, non-genomic disease: Therapy with S2RM exosome released molecules. World J Stem Cells 2017; 9:187-202. [PMID: 29312526 PMCID: PMC5745587 DOI: 10.4252/wjsc.v9.i11.187] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 08/10/2017] [Accepted: 09/04/2017] [Indexed: 02/06/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a rapidly progressing neurodegenerative disease that leads to death. No effective treatments are currently available. Based on data from epidemiological, etiological, laboratory, and clinical studies, I offer a new way of thinking about ALS and its treatment. This paper describes a host of extrinsic factors, including the exposome, that disrupt the extracellular matrix and protein function such that a spreading, prion-like disease leads to neurodegeneration in the motor tracts. A treatment regimen is described using the stem cell released molecules from a number of types of adult stem cells to provide tissue dependent molecules that restore homeostasis, including proteostasis, in the ALS patient. Because stem cells themselves as a therapeutic are cumbersome and expensive, and when implanted in a host cause aging of the host tissue and often fail to engraft or remain viable, only the S2RM molecules are used. Rebuilding of the extracellular matrix and repair of the dysfunctional proteins in the ALS patient ensues.
Collapse
Affiliation(s)
- Greg Maguire
- BioRegenerative Sciences, Inc., La Jolla, CA 92037, United States
| |
Collapse
|
109
|
Skvarc DR, Berk M, Byrne LK, Dean OM, Dodd S, Lewis M, Marriott A, Moore EM, Morris G, Page RS, Gray L. Post-Operative Cognitive Dysfunction: An exploration of the inflammatory hypothesis and novel therapies. Neurosci Biobehav Rev 2017; 84:116-133. [PMID: 29180259 DOI: 10.1016/j.neubiorev.2017.11.011] [Citation(s) in RCA: 217] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 10/16/2017] [Accepted: 11/20/2017] [Indexed: 12/11/2022]
Abstract
Post-Operative Cognitive Dysfunction (POCD) is a highly prevalent condition with significant clinical, social and financial impacts for patients and their communities. The underlying pathophysiology is becoming increasingly understood, with the role of neuroinflammation and oxidative stress secondary to surgery and anaesthesia strongly implicated. This review aims to describe the putative mechanisms by which surgery-induced inflammation produces cognitive sequelae, with a focus on identifying potential novel therapies based upon their ability to modify these pathways.
Collapse
Affiliation(s)
- David R Skvarc
- School of Psychology, Deakin University, Melbourne, Australia; Deakin University, Innovations in Mental and Physical Health and Clinical Treatment (IMPACT) Strategic Research Centre, Barwon Health, Geelong, Australia.
| | - Michael Berk
- Deakin University, Innovations in Mental and Physical Health and Clinical Treatment (IMPACT) Strategic Research Centre, Barwon Health, Geelong, Australia; Deakin University, School of Medicine, Geelong, Australia; Orygen, The National Centre of Excellence in Youth Mental Health, The Department of Psychiatry and the Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Australia.
| | - Linda K Byrne
- School of Psychology, Deakin University, Melbourne, Australia.
| | - Olivia M Dean
- Deakin University, Innovations in Mental and Physical Health and Clinical Treatment (IMPACT) Strategic Research Centre, Barwon Health, Geelong, Australia; Deakin University, School of Medicine, Geelong, Australia; Orygen, The National Centre of Excellence in Youth Mental Health, The Department of Psychiatry and the Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Australia
| | - Seetal Dodd
- Deakin University, Innovations in Mental and Physical Health and Clinical Treatment (IMPACT) Strategic Research Centre, Barwon Health, Geelong, Australia; Deakin University, School of Medicine, Geelong, Australia
| | - Matthew Lewis
- School of Psychology, Deakin University, Melbourne, Australia; Aged Psychiatry Service, Caulfield Hospital, Alfred Health, Caulfield, Australia
| | - Andrew Marriott
- Department of Anaesthesia, Perioperative Medicine & Pain Management, Barwon Health, Geelong, Australia; Deakin University, Innovations in Mental and Physical Health and Clinical Treatment (IMPACT) Strategic Research Centre, Barwon Health, Geelong, Australia; Deakin University, School of Medicine, Geelong, Australia; Orygen, The National Centre of Excellence in Youth Mental Health, The Department of Psychiatry and the Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Australia
| | - Eileen M Moore
- Department of Anaesthesia, Perioperative Medicine & Pain Management, Barwon Health, Geelong, Australia; Deakin University, Innovations in Mental and Physical Health and Clinical Treatment (IMPACT) Strategic Research Centre, Barwon Health, Geelong, Australia
| | | | - Richard S Page
- Deakin University, School of Medicine, Geelong, Australia; Department of Orthopaedics, Barwon Health, Geelong, Australia
| | - Laura Gray
- Deakin University, School of Medicine, Geelong, Australia.
| |
Collapse
|
110
|
Gouveia A, Bajwa E, Klegeris A. Extracellular cytochrome c as an intercellular signaling molecule regulating microglial functions. Biochim Biophys Acta Gen Subj 2017; 1861:2274-2281. [DOI: 10.1016/j.bbagen.2017.06.017] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Revised: 06/05/2017] [Accepted: 06/22/2017] [Indexed: 01/13/2023]
|
111
|
Amri J, Sadegh M, Moulaei N, Palizvan MR. Transgenerational modification of hippocampus TNF-α and S100B levels in the offspring of rats chronically exposed to morphine during adolescence. THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 2017; 44:95-102. [PMID: 28750172 DOI: 10.1080/00952990.2017.1348509] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
BACKGROUND TNF-α and S100B are important signaling factors that are involved in many aberrant conditions of the brain. Chronic morphine exposure causes aberrant modifications in the brain. OBJECTIVES We examined the consequences of chronic morphine consumption by parents before mating on hippocampus TNF-α and S100B levels in the parents and their offspring. METHODS A total of 12 adult female and 12 adult male Wistar rats were used as parents. Each gender was divided randomly into two groups: control and morphine consumer. Morphine consumer groups received morphine sulfate dissolved in drinking water (0.4 mg/ml) for 60 days. Control groups received water. Thirty days before mating, morphine was replaced with water. All offspring also received water. The hippocampus of both parental and offspring groups was extracted to measure TNF-α and S100B levels using an ELISA. RESULTS Hippocampus TNF-α levels were significantly increased due to chronic morphine use in both male and female parents compared to those of control parents (P < 0.01). Moreover, both male and female offspring of morphine-exposed parents showed a significant increase in hippocampus TNF-α levels compared to those of control offspring (P < 0.01). Hippocampus levels of S100B were significantly decreased in male (P < 0.05) but not female morphine consumer parents relative to control parents. Both male and female offspring of morphine-exposed parents showed significant decreases in hippocampus S100B levels (P < 0.05) compared to those of control offspring. CONCLUSIONS The consequences of chronic morphine use by parents, even when it is stopped long before mating and pregnancy, could induce modifications in the hippocampus of the next generation.
Collapse
Affiliation(s)
- Jamal Amri
- a Department of Biochemistry and Genetics, Faculty of Medicine , Arak University of Medical Sciences , Arak , Iran
| | - Mehdi Sadegh
- b Department of Physiology, Faculty of Medicine , Arak University of Medical Sciences , Arak , Iran
| | - Neda Moulaei
- b Department of Physiology, Faculty of Medicine , Arak University of Medical Sciences , Arak , Iran
| | - Mohammad Reza Palizvan
- b Department of Physiology, Faculty of Medicine , Arak University of Medical Sciences , Arak , Iran
| |
Collapse
|
112
|
Rezaei O, Pakdaman H, Gharehgozli K, Simani L, Vahedian-Azimi A, Asaadi S, Sahraei Z, Hajiesmaeili M. S100 B: A new concept in neurocritical care. IRANIAN JOURNAL OF NEUROLOGY 2017; 16:83-89. [PMID: 28761630 PMCID: PMC5526782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
After brain injuries, concentrations of some brain markers such as S100B protein in serum and cerebrospinal fluid (CSF) are correlated with the severity and outcome of brain damage. To perform an updated review of S100B roles in human neurocritical care domain, an electronic literature search was carried among articles published in English prior to March 2017. They were retrieved from PubMed, Scopus, EMBSCO, CINAHL, ISC and the Cochrane Library using keywords including "brain", "neurobiochemical marker", "neurocritical care", and "S100B protein". The integrative review included 48 studies until March 2017. S100B protein can be considered as a marker for blood brain barrier damage. The marker has an important role in the development and recovery of normal central nervous system (CNS) after injury. In addition to extra cerebral sources of S100B, the marker is principally built in the astroglial and Schwann cells. The neurobiochemical marker, S100B, has a pathognomonic role in the diagnosis of a broad spectrum of brain damage including traumatic brain injury (TBI), brain tumor, and stroke. Moreover, a potential predicting role for the neurobiochemical marker has been presumed in the efficiency of brain damage treatment and prognosis. However further animal and human studies are required before widespread routine clinical introduction of S100 protein.
Collapse
Affiliation(s)
- Omidvar Rezaei
- Skull Base Research Center, Loghman Hakim Medical Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein Pakdaman
- Brain Mapping Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kurosh Gharehgozli
- Brain Mapping Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leila Simani
- Clinical Research Development Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Vahedian-Azimi
- Trauma Research Center, School of Nursing, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Sina Asaadi
- Clinical Research Development Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Sahraei
- Department of Clinical Pharmacy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Hajiesmaeili
- Anesthesiology Research Center, Loghman Hakim Medical Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
113
|
Tsubokawa D, Hatta T, Kikuchi T, Maeda H, Mikami F, Alim MA, Maruyama H, Tsuji N. Venestatin, a Ca ++-binding protein from the parasitic nematode Strongyloides venezuelensis, is involved in the larval migration process. Int J Parasitol 2017; 47:501-509. [PMID: 28347664 DOI: 10.1016/j.ijpara.2017.01.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 01/17/2017] [Accepted: 01/18/2017] [Indexed: 11/16/2022]
Abstract
The secretory EF-hand Ca++-binding proteins act as calcium signaling molecules for control of cell functions, but those proteins from parasitic helminths are poorly understood. Here, we have identified and characterized an EF-hand Ca++-binding protein from the rodent nematode, Strongyloides venezuelensis, termed 'venestatin', which is highly conserved in Strongyloides spp. Canonical two EF-hand domains and a signal peptide are present in venestatin. A gel mobility shift assay and Ruthenium red staining indicated that the recombinant venestatin possesses binding ability with Ca++ ions. Endogenous venestatin was seemingly localized in the hypodermis and gut of the worms and was found in the excretory-secretory products. Quantitative reverse transcription-PCR data showed that venestatin-specific transcript was upregulated in the parasitic stages of S. venezuelensis, and the upregulation occurred promptly after larval invasion through the host's skin, but not in the case of in vitro incubation. Immunization of mice with recombinant venestatin caused a 55% reduction in larval migration to the lungs, and lung hemorrhaging was mild compared with non-immunized groups, suggesting that anti-venestatin sera may interfere with larval migration from skin to lung. Our results suggest that venestatin is secreted from the hypodermis and gut of S. venezuelensis, and has pivotal roles in larval migration.
Collapse
Affiliation(s)
- Daigo Tsubokawa
- Department of Molecular and Cellular Parasitology, Kitasato University Graduate School of Medical Sciences, 1-15-1 Kitasato, Minami, Sagamihara, Kanagawa 252-0373, Japan; Department of Parasitology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami, Sagamihara, Kanagawa 252-0374, Japan
| | - Takeshi Hatta
- Department of Molecular and Cellular Parasitology, Kitasato University Graduate School of Medical Sciences, 1-15-1 Kitasato, Minami, Sagamihara, Kanagawa 252-0373, Japan; Department of Parasitology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami, Sagamihara, Kanagawa 252-0374, Japan
| | - Taisei Kikuchi
- Division of Parasitology, Department of Infectious Diseases, Faculty of Medicine, University of Miyazaki, 5200 Kihara Kiyotake, Miyazaki 899-1692, Japan
| | - Hiroki Maeda
- Department of Parasitology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami, Sagamihara, Kanagawa 252-0374, Japan; Department of Pathological and Preventive Veterinary Science, The United Graduate School of Veterinary Science, Yamaguchi University, Yoshida, Yamaguchi 753-8515, Japan
| | - Fusako Mikami
- Department of Parasitology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami, Sagamihara, Kanagawa 252-0374, Japan
| | - M Abdul Alim
- Department of Parasitology, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh
| | - Haruhiko Maruyama
- Division of Parasitology, Department of Infectious Diseases, Faculty of Medicine, University of Miyazaki, 5200 Kihara Kiyotake, Miyazaki 899-1692, Japan
| | - Naotoshi Tsuji
- Department of Molecular and Cellular Parasitology, Kitasato University Graduate School of Medical Sciences, 1-15-1 Kitasato, Minami, Sagamihara, Kanagawa 252-0373, Japan; Department of Parasitology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami, Sagamihara, Kanagawa 252-0374, Japan.
| |
Collapse
|
114
|
RAGE-TLR Crosstalk Sustains Chronic Inflammation in Neurodegeneration. Mol Neurobiol 2017; 55:1463-1476. [PMID: 28168427 DOI: 10.1007/s12035-017-0419-4] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 01/24/2017] [Indexed: 01/10/2023]
Abstract
Chronic inflammatory reactions are consistenly present in neurodegeneration of Alzheimer type and are considered important factors that accelerate progression of the disease. Receptors of innate immunity participate in triggering and driving inflammatory reactions. For example, Toll-like receptors (TLRs) and receptor for advanced glycation end product (RAGE), major receptors of innate immunity, play a central role in perpetuation of inflammation. RAGE activation should be perceived as a primary mechanism which determines self-perpetuated chronic inflammation, and RAGE cooperation with TLRs amplifies inflammatory signaling. In this review, we highlight and discuss that RAGE-TLR crosstalk emerges as an important driving force of chronic inflammation in Alzheimer's disease.
Collapse
|
115
|
Qiao H, He X, Zhang Q, Zhang N, Li L, Hui Y, Li W, Wang D, Wu Z. Α-synuclein induces microglial cell migration through stimulating HIF-1α accumulation. J Neurosci Res 2017; 95:1809-1817. [PMID: 28134982 DOI: 10.1002/jnr.24012] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Revised: 11/28/2016] [Accepted: 12/12/2016] [Indexed: 01/09/2023]
Affiliation(s)
- Hongfei Qiao
- Departments of Rehabilitation Medicine; the Second Affiliated Hospital of Xi’an Jiaotong University; Xi’an China
| | - Xijing He
- Departments of Orthopedics; the Second Affiliated Hospital of Xi’an Jiaotong University; Xi’an China
| | - Qiaojun Zhang
- Departments of Rehabilitation Medicine; the Second Affiliated Hospital of Xi’an Jiaotong University; Xi’an China
| | - Ni Zhang
- Departments of Rehabilitation Medicine; the Second Affiliated Hospital of Xi’an Jiaotong University; Xi’an China
| | - Libo Li
- Departments of Rehabilitation Medicine; the Second Affiliated Hospital of Xi’an Jiaotong University; Xi’an China
| | - Yanping Hui
- Departments of Rehabilitation Medicine; the Second Affiliated Hospital of Xi’an Jiaotong University; Xi’an China
| | - Wenjuan Li
- Departments of Rehabilitation Medicine; the Second Affiliated Hospital of Xi’an Jiaotong University; Xi’an China
| | - Dong Wang
- Departments of Orthopedics; the Second Affiliated Hospital of Xi’an Jiaotong University; Xi’an China
| | - Zhonghen Wu
- Departments of Rehabilitation Medicine; the Second Affiliated Hospital of Xi’an Jiaotong University; Xi’an China
| |
Collapse
|
116
|
Guloksuz SA, Abali O, Aktas Cetin E, Bilgic Gazioglu S, Deniz G, Yildirim A, Kawikova I, Guloksuz S, Leckman JF. Elevated plasma concentrations of S100 calcium-binding protein B and tumor necrosis factor alpha in children with autism spectrum disorders. ACTA ACUST UNITED AC 2017; 39:195-200. [PMID: 28099628 PMCID: PMC7111377 DOI: 10.1590/1516-4446-2015-1843] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 09/18/2016] [Indexed: 02/08/2023]
Abstract
Objective: To investigate plasma concentrations of S100B (a calcium-binding protein derived primarily from the glia) and inflammatory cytokines in children with autism and the relationship between S100B and cytokine concentrations. Methods: Plasma levels of S100B, tumor necrosis factor alpha (TNF-α), interferon gamma, interleukin (IL)-1β, IL-4, IL-6, IL-10, and IL-17A were measured in 40 unmedicated children with autism and 35 normally developing healthy children. The severity of autism was assessed using the Childhood Autism Rating Scale (CARS). Results: Concentrations of both S100B and TNF-α were higher in children with autism before and after adjusting for a priori-selected confounders (age, sex, and body mass index). S100B concentrations were higher in children with severe autism compared to children with mild-moderate autism. However, this association remained as a trend after adjusting for confounders. S100B concentrations correlated positively with TNF-α concentrations. Conclusion: Our findings showing an increase in peripheral concentrations of S100B and TNF-α provide limited support to the hypothesis about the roles of altered immune function and S100B in autism spectrum disorder (ASD). Studies of larger numbers of well-characterized individuals with ASD are needed to clarify the potential role of the immune system in the pathophysiology of this disorder.
Collapse
Affiliation(s)
| | - Osman Abali
- Department of Child and Adolescent Psychiatry, Istanbul University School of Medicine, Istanbul, Turkey
| | - Esin Aktas Cetin
- Department of Immunology, Institute of Experimental Medicine (DETAE), Istanbul University, Istanbul, Turkey
| | - Sema Bilgic Gazioglu
- Department of Immunology, Institute of Experimental Medicine (DETAE), Istanbul University, Istanbul, Turkey
| | - Gunnur Deniz
- Department of Immunology, Institute of Experimental Medicine (DETAE), Istanbul University, Istanbul, Turkey
| | | | - Ivana Kawikova
- Department of Pediatrics, School of Medicine, Yale University, New Haven, CT, USA
| | - Sinan Guloksuz
- Department of Psychiatry, School of Medicine, Yale University, New Haven, CT, USA.,Department of Psychiatry and Neuropsychology, South Limburg Mental Health Research and Teaching Network, European Graduate School of Neuroscience (EURON), School for Mental Health and Neuroscience (MHeNS), Maastricht University, Maastricht, The Netherlands
| | - James F Leckman
- Child Study Center, School of Medicine, Yale University, New Haven, CT, USA
| |
Collapse
|
117
|
Shekhtman A, Ramasamy R, Schmidt AM. Glycation & the RAGE axis: targeting signal transduction through DIAPH1. Expert Rev Proteomics 2016; 14:147-156. [PMID: 27967251 DOI: 10.1080/14789450.2017.1271719] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
INTRODUCTION The consequences of chronic disease are vast and unremitting; hence, understanding the pathogenic mechanisms mediating such disorders holds promise to identify therapeutics and diminish the consequences. The ligands of the receptor for advanced glycation end products (RAGE) accumulate in chronic diseases, particularly those characterized by inflammation and metabolic dysfunction. Although first discovered and reported as a receptor for advanced glycation end products (AGEs), the expansion of the repertoire of RAGE ligands implicates the receptor in diverse milieus, such as autoimmunity, chronic inflammation, obesity, diabetes, and neurodegeneration. Areas covered: This review summarizes current knowledge regarding the ligand families of RAGE and data from human subjects and animal models on the role of the RAGE axis in chronic diseases. The recent discovery that the cytoplasmic domain of RAGE binds to the formin homology 1 (FH1) domain, DIAPH1, and that this interaction is essential for RAGE ligand-stimulated signal transduction, is discussed. Finally, we review therapeutic opportunities targeting the RAGE axis as a means to mitigate chronic diseases. Expert commentary: With the aging of the population and the epidemic of cardiometabolic disease, therapeutic strategies to target molecular pathways that contribute to the sequelae of these chronic diseases are urgently needed. In this review, we propose that the ligand/RAGE axis and its signaling nexus is a key factor in the pathogenesis of chronic disease and that therapeutic interruption of this pathway may improve quality and duration of life.
Collapse
Affiliation(s)
- Alexander Shekhtman
- a Department of Chemistry , University at Albany, State University of New York , Albany , NY , 12222 , USA
| | - Ravichandran Ramasamy
- b Diabetes Research Program, Division of Endocrinology, Department of Medicine , NYU Langone Medical Center , New York , NY , 10016 , USA
| | - Ann Marie Schmidt
- b Diabetes Research Program, Division of Endocrinology, Department of Medicine , NYU Langone Medical Center , New York , NY , 10016 , USA
| |
Collapse
|
118
|
S100B raises the alert in subarachnoid hemorrhage. Rev Neurosci 2016; 27:745-759. [DOI: 10.1515/revneuro-2016-0021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 05/26/2016] [Indexed: 12/19/2022]
Abstract
AbstractSubarachnoid hemorrhage (SAH) is a devastating disease with high mortality and mobility, the novel therapeutic strategies of which are essentially required. The calcium binding protein S100B has emerged as a brain injury biomarker that is implicated in pathogenic process of SAH. S100B is mainly expressed in astrocytes of the central nervous system and functions through initiating intracellular signaling or via interacting with cell surface receptor, such as the receptor of advanced glycation end products. The biological roles of S100B in neurons have been closely associated with its concentrations, resulting in either neuroprotection or neurotoxicity. The levels of S100B in the blood have been suggested as a biomarker to predict the progress or the prognosis of SAH. The role of S100B in the development of cerebral vasospasm and brain damage may result from the induction of oxidative stress and neuroinflammation after SAH. To get further insight into mechanisms underlying the role of S100B in SAH based on this review might help us to find novel therapeutic targets for SAH.
Collapse
|
119
|
Lasič E, Galland F, Vardjan N, Šribar J, Križaj I, Leite MC, Zorec R, Stenovec M. Time-dependent uptake and trafficking of vesicles capturing extracellular S100B in cultured rat astrocytes. J Neurochem 2016; 139:309-323. [PMID: 27488079 DOI: 10.1111/jnc.13754] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 07/21/2016] [Accepted: 07/26/2016] [Indexed: 01/16/2023]
Abstract
Astrocytes, the most heterogeneous glial cells in the central nervous system, contribute to brain homeostasis, by regulating a myriad of functions, including the clearance of extracellular debris. When cells are damaged, cytoplasmic proteins may exit into the extracellular space. One such protein is S100B, which may exert toxic effects on neighboring cells unless it is removed from the extracellular space, but the mechanisms of this clearance are poorly understood. By using time-lapse confocal microscopy and fluorescently labeled S100B (S100B-Alexa488 ) and fluorescent dextran (Dextran546 ), a fluid phase uptake marker, we examined the uptake of fluorescently labeled S100B-Alexa488 from extracellular space and monitored trafficking of vesicles that internalized S100B-Alexa488 . Initially, S100B-Alexa488 and Dextran546 internalized with distinct rates into different endocytotic vesicles; S100B-Alexa488 internalized into smaller vesicles than Dextran546 . At a later stage, S100B-Alexa488 -positive vesicles substantially co-localized with Dextran546 -positive endolysosomes and with acidic LysoTracker-positive vesicles. Cell treatment with anti-receptor for advanced glycation end products (RAGE) antibody, which binds to RAGE, a 'scavenger receptor', partially inhibited uptake of S100B-Alexa488 , but not of Dextran546 . The dynamin inhibitor dynole 34-2 inhibited internalization of both fluorescent probes. Directional mobility of S100B-Alexa488 -positive vesicles increased over time and was inhibited by ATP stimulation, an agent that increases cytosolic free calcium concentration ([Ca2+ ]i ). We conclude that astrocytes exhibit RAGE- and dynamin-dependent vesicular mechanism to efficiently remove S100B from the extracellular space. If a similar process occurs in vivo, astroglia may mitigate the toxic effects of extracellular S100B by this process under pathophysiologic conditions. This study reveals the vesicular clearance mechanism of extracellular S100B in astrocytes. Initially, fluorescent S100B internalizes into smaller endocytotic vesicles than dextran molecules. At a later stage, both probes co-localize within endolysosomes. S100B internalization is both dynamin- and RAGE-dependent, whereas dextran internalization is dependent on dynamin. Vesicle internalization likely mitigates the toxic effects of extracellular S100B and other waste products.
Collapse
Affiliation(s)
- Eva Lasič
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Fabiana Galland
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia.,Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Nina Vardjan
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia.,Celica Biomedical, Ljubljana, Slovenia
| | - Jernej Šribar
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Igor Križaj
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Ljubljana, Slovenia.,Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Marina Concli Leite
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Robert Zorec
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia. .,Celica Biomedical, Ljubljana, Slovenia.
| | - Matjaž Stenovec
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia. .,Celica Biomedical, Ljubljana, Slovenia.
| |
Collapse
|
120
|
Xue J, Manigrasso M, Scalabrin M, Rai V, Reverdatto S, Burz DS, Fabris D, Schmidt AM, Shekhtman A. Change in the Molecular Dimension of a RAGE-Ligand Complex Triggers RAGE Signaling. Structure 2016; 24:1509-22. [PMID: 27524199 DOI: 10.1016/j.str.2016.06.021] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 05/20/2016] [Accepted: 06/16/2016] [Indexed: 01/13/2023]
Abstract
The weak oligomerization exhibited by many transmembrane receptors has a profound effect on signal transduction. The phenomenon is difficult to characterize structurally due to the large sizes of and transient interactions between monomers. The receptor for advanced glycation end products (RAGE), a signaling molecule central to the induction and perpetuation of inflammatory responses, is a weak constitutive oligomer. The RAGE domain interaction surfaces that mediate homo-dimerization were identified by combining segmental isotopic labeling of extracellular soluble RAGE (sRAGE) and nuclear magnetic resonance spectroscopy with chemical cross-linking and mass spectrometry. Molecular modeling suggests that two sRAGE monomers orient head to head forming an asymmetric dimer with the C termini directed toward the cell membrane. Ligand-induced association of RAGE homo-dimers on the cell surface increases the molecular dimension of the receptor, recruiting Diaphanous 1 (DIAPH1) and activating signaling pathways.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/chemistry
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- Amino Acid Sequence
- Animals
- Antigens, Neoplasm/chemistry
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/metabolism
- Bacterial Proteins/genetics
- Bacterial Proteins/metabolism
- Binding Sites
- Cross-Linking Reagents/chemistry
- Formins
- Gene Expression
- Genes, Reporter
- Green Fluorescent Proteins/genetics
- Green Fluorescent Proteins/metabolism
- HEK293 Cells
- Humans
- Ligands
- Luminescent Proteins/genetics
- Luminescent Proteins/metabolism
- Maleimides/chemistry
- Mitogen-Activated Protein Kinases/chemistry
- Mitogen-Activated Protein Kinases/genetics
- Mitogen-Activated Protein Kinases/metabolism
- Molecular Docking Simulation
- Nuclear Magnetic Resonance, Biomolecular
- Protein Binding
- Protein Interaction Domains and Motifs
- Protein Structure, Secondary
- Recombinant Proteins/chemistry
- Recombinant Proteins/genetics
- Recombinant Proteins/metabolism
- Sequence Alignment
- Sequence Homology, Amino Acid
- Signal Transduction
- Thermodynamics
Collapse
Affiliation(s)
- Jing Xue
- Department of Chemistry, State University of New York at Albany, Albany, NY 12222, USA
| | | | - Matteo Scalabrin
- Department of Chemistry, State University of New York at Albany, Albany, NY 12222, USA
| | - Vivek Rai
- Institute of Life Sciences, Bhubaneswar, Odisha 751023, India
| | - Sergey Reverdatto
- Department of Chemistry, State University of New York at Albany, Albany, NY 12222, USA
| | - David S Burz
- Department of Chemistry, State University of New York at Albany, Albany, NY 12222, USA
| | - Daniele Fabris
- Department of Chemistry, State University of New York at Albany, Albany, NY 12222, USA
| | - Ann Marie Schmidt
- New York University, Langone Medical Center, New York, NY 10016, USA
| | - Alexander Shekhtman
- Department of Chemistry, State University of New York at Albany, Albany, NY 12222, USA.
| |
Collapse
|
121
|
Abstract
Inflammatory activation of microglia is a hallmark of several disorders of the central nervous system. In addition to protecting the brain against inflammatory insults, microglia are neuroprotective and play a significant role in maintaining neuronal connectivity, but the prolongation of an inflammatory status may limit the beneficial functions of these immune cells. The finding that estrogen receptors are present in monocyte-derived cells and that estrogens prevent and control the inflammatory response raise the question of the role that this sex steroid plays in the manifestation and progression of pathologies that have a clear sex difference in prevalence, such as multiple sclerosis, Parkinson's disease, and Alzheimer's disease. The present review aims to provide a critical review of the current literature on the actions of estrogen in microglia and on the involvement of estrogen receptors in the manifestation of selected neurological disorders. This current understanding highlights a research area that should be expanded to identify appropriate replacement therapies to slow the progression of such diseases.
Collapse
Affiliation(s)
- Alessandro Villa
- Center of Excellence on Neurodegenerative Diseases and Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy
| | - Elisabetta Vegeto
- Center of Excellence on Neurodegenerative Diseases and Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy
| | - Angelo Poletti
- Center of Excellence on Neurodegenerative Diseases and Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy
| | - Adriana Maggi
- Center of Excellence on Neurodegenerative Diseases and Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy
| |
Collapse
|
122
|
Watts BA, George T, Badalamenti A, Good DW. High-mobility group box 1 inhibits HCO3- absorption in the medullary thick ascending limb through RAGE-Rho-ROCK-mediated inhibition of basolateral Na+/H+ exchange. Am J Physiol Renal Physiol 2016; 311:F600-13. [PMID: 27358052 DOI: 10.1152/ajprenal.00185.2016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 06/27/2016] [Indexed: 02/07/2023] Open
Abstract
High-mobility group box 1 (HMGB1) is a nuclear protein released extracellularly in response to infection or injury, where it activates immune responses and contributes to the pathogenesis of kidney dysfunction in sepsis and sterile inflammatory disorders. Recently, we demonstrated that HMGB1 inhibits HCO3 (-) absorption in perfused rat medullary thick ascending limbs (MTAL) through a basolateral receptor for advanced glycation end products (RAGE)-dependent pathway that is additive to Toll-like receptor 4 (TLR4)-ERK-mediated inhibition by LPS (Good DW, George T, Watts BA III. Am J Physiol Renal Physiol 309: F720-F730, 2015). Here, we examined signaling and transport mechanisms that mediate inhibition by HMGB1. Inhibition of HCO3 (-) absorption by HMGB1 was eliminated by the Rho-associated kinase (ROCK) inhibitor Y27632 and by a specific inhibitor of Rho, the major upstream activator of ROCK. HMGB1 increased RhoA and ROCK1 activity. HMGB1-induced ROCK1 activation was eliminated by the RAGE antagonist FPS-ZM1 and by inhibition of Rho. The Rho and ROCK inhibitors had no effect on inhibition of HCO3 (-) absorption by bath LPS. Inhibition of HCO3 (-) absorption by HMGB1 was eliminated by bath amiloride, 0 Na(+) bath, and the F-actin stabilizer jasplakinolide, three conditions that selectively prevent inhibition of MTAL HCO3 (-) absorption mediated through NHE1. HMGB1 decreased basolateral Na(+)/H(+) exchange activity through activation of ROCK. We conclude that HMGB1 inhibits HCO3 (-) absorption in the MTAL through a RAGE-RhoA-ROCK1 signaling pathway coupled to inhibition of NHE1. The HMGB1-RAGE-RhoA-ROCK1 pathway thus represents a potential target to attenuate MTAL dysfunction during sepsis and other inflammatory disorders. HMGB1 and LPS inhibit HCO3 (-) absorption through different receptor signaling and transport mechanisms, which enables these pathogenic mediators to act directly and independently to impair MTAL function.
Collapse
Affiliation(s)
- Bruns A Watts
- Department of Internal Medicine, The University of Texas Medical Branch, Galveston, Texas; and
| | - Thampi George
- Department of Internal Medicine, The University of Texas Medical Branch, Galveston, Texas; and
| | - Andrew Badalamenti
- Department of Internal Medicine, The University of Texas Medical Branch, Galveston, Texas; and
| | - David W Good
- Department of Internal Medicine, The University of Texas Medical Branch, Galveston, Texas; and Department of Neuroscience and Cell Biology, The University of Texas Medical Branch, Galveston, Texas
| |
Collapse
|
123
|
Kato J, Agalave NM, Svensson CI. Pattern recognition receptors in chronic pain: Mechanisms and therapeutic implications. Eur J Pharmacol 2016; 788:261-273. [PMID: 27343378 DOI: 10.1016/j.ejphar.2016.06.039] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Revised: 06/22/2016] [Accepted: 06/22/2016] [Indexed: 12/13/2022]
Abstract
For the individual, it is vital to promptly detect and recognize a danger that threatens the integrity of the body. Pattern recognition receptors (PRRs) are several classes of protein families originally classified as receptors detecting exogenous pathogens. PRRs are also capable of recognizing molecules released from damaged tissues (damage-associated molecular pattern molecules; DAMPs) and thereby contribute to danger recognition. Importantly, it is now evident that PRRs, such as toll-like receptors (TLRs) and receptors for advanced glycation end products (RAGE), are not only expressed in peripheral immune cells but also present in neurons and glial cells in the nervous system. These PRR-expressing cells work in concert, enabling highly sensitive danger recognition. However, this sensitiveness can act as a double-edged sword. Accumulated evidence has led to the hypothesis that aberrant activation of PRRs may play a crucial role in the pathogenesis of pathological pain. Indeed, numerous studies employing gene deletion or pharmacological inhibition of PRRs successfully reversed or prevented pathological pain in experimental animal models. Furthermore, a number of preclinical studies have shown the therapeutic potential of targeting PRRs for chronic pain. Here, we review the current knowledge regarding the role of PRRs in chronic pain and discuss the promise and challenges of targeting PRRs as a novel therapeutic approach for chronic pain.
Collapse
Affiliation(s)
- Jungo Kato
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
| | - Nilesh M Agalave
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Camilla I Svensson
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
124
|
Juranek JK, Daffu GK, Geddis MS, Li H, Rosario R, Kaplan BJ, Kelly L, Schmidt AM. Soluble RAGE Treatment Delays Progression of Amyotrophic Lateral Sclerosis in SOD1 Mice. Front Cell Neurosci 2016; 10:117. [PMID: 27242430 PMCID: PMC4860390 DOI: 10.3389/fncel.2016.00117] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 04/22/2016] [Indexed: 12/12/2022] Open
Abstract
The etiology of amyotrophic lateral sclerosis (ALS), a fatal motor neuron disorder characterized by progressive muscle weakness and spasticity, remains largely unknown. Approximately 5-10% of cases are familial, and of those, 15-20% are associated with mutations in the gene encoding Cu/Zn superoxide dismutase (SOD1). Mutations of the SOD1 gene interrupt cellular homeostasis and contribute to cellular toxicity evoked by the presence of altered SOD1, along with other toxic species, such as advanced glycation end products (AGEs). AGEs trigger activation of their chief cell surface receptor, RAGE (receptor for advanced glycation end products), and induce RAGE-dependent cellular stress and inflammation in neurons, thereby affecting their function and leading to apoptosis. Here, we show for the first time that the expression of RAGE is higher in the SOD1 transgenic mouse model of ALS vs. wild-type mouse spinal cord. We tested whether pharmacological blockade of RAGE may delay the onset and progression of disease in this mouse model. Our findings reveal that treatment of SOD1 transgenic mice with soluble RAGE (sRAGE), a natural competitor of RAGE that sequesters RAGE ligands and blocks their interaction with cell surface RAGE, significantly delays the progression of ALS and prolongs life span compared to vehicle treatment. We demonstrate that in sRAGE-treated SOD1 transgenic animals at the final stage of the disease, a significantly higher number of neurons and lower number of astrocytes is detectable in the spinal cord. We conclude that RAGE antagonism may provide a novel therapeutic strategy for ALS intervention.
Collapse
Affiliation(s)
- Judyta K Juranek
- Division of Endocrinology, Department of Medicine, New York University Langone Medical CenterNew York, NY, USA; Department of Surgery, Columbia University Medical CenterNew York, NY, USA
| | - Gurdip K Daffu
- Division of Endocrinology, Department of Medicine, New York University Langone Medical Center New York, NY, USA
| | - Matthew S Geddis
- Department of Surgery, Columbia University Medical CenterNew York, NY, USA; Department of Science, Borough of Manhattan Community College-City University of New YorkNew York, NY, USA
| | - Huilin Li
- Division of Biostatistics, Department of Population Health, New York University Langone Medical Center New York, NY, USA
| | - Rosa Rosario
- Division of Endocrinology, Department of Medicine, New York University Langone Medical CenterNew York, NY, USA; Department of Surgery, Columbia University Medical CenterNew York, NY, USA
| | - Benjamin J Kaplan
- Department of Surgery, Columbia University Medical Center New York, NY, USA
| | - Lauren Kelly
- Department of Surgery, Columbia University Medical Center New York, NY, USA
| | - Ann Marie Schmidt
- Division of Endocrinology, Department of Medicine, New York University Langone Medical CenterNew York, NY, USA; Department of Surgery, Columbia University Medical CenterNew York, NY, USA
| |
Collapse
|
125
|
Liu S, Zhang Y, Zhao Y, Cui H, Cao C, Guo J. Effects of hypothermia on S100B and glial fibrillary acidic protein in asphyxia rats after cardiopulmonary resuscitation. Cell Biochem Biophys 2016; 71:401-6. [PMID: 25209741 DOI: 10.1007/s12013-014-0212-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
The aim of the study was to investigate the effects of hypothermia on S100B and glial fibrillary acidic protein (GFAP) in serum and hippocampus CA1 area in asphyxiated rats after cardiopulmonary resuscitation (CPR). A total of 100 SD rats were designated into four groups: group A, sham operation group; group B, rats received conventional resuscitation; group C, rats received conventional resuscitation and hypothermia at cardiac arrest; group D, rats received conventional resuscitation and hypothermia at 30 min after restoration of spontaneous circulation (ROSC). Rats were then killed by cardiac arrest at 2 and 4 h after ROSC; brain tissue was taken to observe dynamic changes of S100B and GFAP in serum and hippocampus CA1 area. Following ROSC, S100B levels increased from 2 to 4 h in group B, C, and D. In addition, S100B in serum and hippocampus CA1 area was all significantly increased at different time points compared with group A (P < 0.05). Following ROSC, serum S100B level at 2 h in group C was significantly decreased compared with group B, but the difference was not statistically significant (P > 0.05). Moreover, S100B in serum at 4 h after ROSC was significantly decreased (P < 0.05), S100B in cortex was significantly decreased (P < 0.05). The expression of GFAP was also examined. GFAP level in hippocampus CA1 area was significantly decreased in group B, C, and D at 4 h after ROSC compared with group A (P < 0.05). S100B and GFAP were expressed in rat serum and hippocampus CA2 area at early stage after ROSC, which can be used as sensitive markers for brain injury diagnosis and prognosis prediction. Hypothermia is also shown to reduce brain injury after CPR.
Collapse
Affiliation(s)
- Sha Liu
- School of Traditional Chinese Medicine, Chongqing Medical University, Jieqing Road, Huxi University City, Chongqing, People's Republic of China
| | | | | | | | | | | |
Collapse
|
126
|
Manigrasso MB, Pan J, Rai V, Zhang J, Reverdatto S, Quadri N, DeVita RJ, Ramasamy R, Shekhtman A, Schmidt AM. Small Molecule Inhibition of Ligand-Stimulated RAGE-DIAPH1 Signal Transduction. Sci Rep 2016; 6:22450. [PMID: 26936329 PMCID: PMC4776135 DOI: 10.1038/srep22450] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 02/15/2016] [Indexed: 12/21/2022] Open
Abstract
The receptor for advanced glycation endproducts (RAGE) binds diverse ligands linked to chronic inflammation and disease. NMR spectroscopy and x-ray crystallization studies of the extracellular domains of RAGE indicate that RAGE ligands bind by distinct charge- and hydrophobicity-dependent mechanisms. The cytoplasmic tail (ct) of RAGE is essential for RAGE ligand-mediated signal transduction and consequent modulation of gene expression and cellular properties. RAGE signaling requires interaction of ctRAGE with the intracellular effector, mammalian diaphanous 1 or DIAPH1. We screened a library of 58,000 small molecules and identified 13 small molecule competitive inhibitors of ctRAGE interaction with DIAPH1. These compounds, which exhibit in vitro and in vivo inhibition of RAGE-dependent molecular processes, present attractive molecular scaffolds for the development of therapeutics against RAGE-mediated diseases, such as those linked to diabetic complications, Alzheimer’s disease, and chronic inflammation, and provide support for the feasibility of inhibition of protein-protein interaction (PPI).
Collapse
Affiliation(s)
- Michaele B Manigrasso
- Diabetes Research Program, Department of Medicine, New York University Langone Medical Center, 550 First Avenue, New York, 10016 New York, USA
| | - Jinhong Pan
- Department of Chemistry, University at Albany, State University of New York, 1400 Washington Avenue, Albany, 12222 New York, USA
| | - Vivek Rai
- Diabetes Research Program, Department of Medicine, New York University Langone Medical Center, 550 First Avenue, New York, 10016 New York, USA
| | - Jinghua Zhang
- Diabetes Research Program, Department of Medicine, New York University Langone Medical Center, 550 First Avenue, New York, 10016 New York, USA
| | - Sergey Reverdatto
- Department of Chemistry, University at Albany, State University of New York, 1400 Washington Avenue, Albany, 12222 New York, USA
| | - Nosirudeen Quadri
- Diabetes Research Program, Department of Medicine, New York University Langone Medical Center, 550 First Avenue, New York, 10016 New York, USA
| | - Robert J DeVita
- RJD Medicinal Chemistry and Drug Discovery Consulting LLC, 332 W. Dudley Avenue, Westfield, New Jersey 07090, USA
| | - Ravichandran Ramasamy
- Diabetes Research Program, Department of Medicine, New York University Langone Medical Center, 550 First Avenue, New York, 10016 New York, USA
| | - Alexander Shekhtman
- Department of Chemistry, University at Albany, State University of New York, 1400 Washington Avenue, Albany, 12222 New York, USA
| | - Ann Marie Schmidt
- Diabetes Research Program, Department of Medicine, New York University Langone Medical Center, 550 First Avenue, New York, 10016 New York, USA
| |
Collapse
|
127
|
Leis AA, Ross MA, Verheijde JL, Leis JF. Immunoablation and Stem Cell Transplantation in Amyotrophic Lateral Sclerosis: The Ultimate Test for the Autoimmune Pathogenesis Hypothesis. Front Neurol 2016; 7:12. [PMID: 26903945 PMCID: PMC4749695 DOI: 10.3389/fneur.2016.00012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2015] [Accepted: 01/22/2016] [Indexed: 12/12/2022] Open
Affiliation(s)
- A Arturo Leis
- Department of Neurology, Mayo Clinic Arizona, Scottsdale, AZ, USA; Center for Neuroscience and Neurological Recovery, Methodist Rehabilitation Center, Jackson, MS, USA
| | - Mark A Ross
- Department of Neurology, Mayo Clinic Arizona , Scottsdale, AZ , USA
| | - Joseph L Verheijde
- Department of Physical Medicine and Rehabilitation, Mayo Clinic Arizona , Scottsdale, AZ , USA
| | - Jose F Leis
- Division of Hematology Oncology, Mayo Clinic Arizona , Phoenix, AZ , USA
| |
Collapse
|
128
|
Wu H, Brown EV, Acharya NK, Appelt DM, Marks A, Nagele RG, Venkataraman V. Age-dependent increase of blood-brain barrier permeability and neuron-binding autoantibodies in S100B knockout mice. Brain Res 2016; 1637:154-167. [PMID: 26907191 DOI: 10.1016/j.brainres.2016.02.026] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 01/26/2016] [Accepted: 02/14/2016] [Indexed: 11/29/2022]
Abstract
S100B is a calcium-sensor protein that impacts multiple signal transduction pathways. It is widely considered to be an important biomarker for several neuronal diseases as well as blood-brain barrier (BBB) breakdown. In this report, we demonstrate a BBB deficiency in mice that lack S100B through detection of leaked Immunoglobulin G (IgG) in the brain parenchyma. IgG leaks and IgG-binding to selected neurons were observed in S100B knockout (S100BKO) mice at 6 months of age but not at 3 months. By 9 months, IgG leaks persisted and the density of IgG-bound neurons increased significantly. These results reveal a chronic increase in BBB permeability upon aging in S100BKO mice for the first time. Moreover, coincident with the increase in IgG-bound neurons, autoantibodies targeting brain proteins were detected in the serum via western blots. These events were concurrent with compromise of neurons, increase of activated microglia and lack of astrocytic activation as evidenced by decreased expression of microtubule-associated protein type 2 (MAP2), elevated number of CD68 positive cells and unaltered expression of glial fibrillary acidic protein (GFAP) respectively. Results suggest a key role for S100B in maintaining BBB functional integrity and, further, propose the S100BKO mouse as a valuable model system to explore the link between chronic functional compromise of the BBB, generation of brain-reactive autoantibodies and neuronal dysfunctions.
Collapse
Affiliation(s)
- Hao Wu
- Graduate School of Biomedical Sciences, Rowan University, Stratford, NJ 08084, USA
| | - Eric V Brown
- Graduate School of Biomedical Sciences, Rowan University, Stratford, NJ 08084, USA
| | - Nimish K Acharya
- Biomarker Discovery Center, New Jersey Institute for Successful Aging, Rowan University School of Osteopathic Medicine, Stratford, NJ 08084, USA; Department of Geriatrics and Gerontology, Rowan University School of Osteopathic Medicine, Stratford, NJ 08084, USA
| | - Denah M Appelt
- Philadelphia College of Osteopathic Medicine, Philadelphia, PA 19131, USA
| | - Alexander Marks
- Donnelly Centre, University of Toronto, Toronto, ON, Canada M6J 3X5
| | - Robert G Nagele
- Biomarker Discovery Center, New Jersey Institute for Successful Aging, Rowan University School of Osteopathic Medicine, Stratford, NJ 08084, USA; Department of Geriatrics and Gerontology, Rowan University School of Osteopathic Medicine, Stratford, NJ 08084, USA
| | - Venkat Venkataraman
- Graduate School of Biomedical Sciences, Rowan University, Stratford, NJ 08084, USA; Department of Cell Biology, Rowan School of Osteopathic Medicine, Stratford, NJ 08084, USA.
| |
Collapse
|
129
|
Zhang J, Zhao GQ, Qu J, Che CY, Lin J, Jiang N, Zhao H, Wang XJ. Expression of S100B during the innate immune of corneal epithelium against fungi invasion. Int J Ophthalmol 2016; 9:191-7. [PMID: 26949634 DOI: 10.18240/ijo.2016.02.02] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 09/09/2015] [Indexed: 01/01/2023] Open
Abstract
AIM To explore the expression of S100B in corneal epithelial cells under Aspergillus stimulation both in vivo and in vitro. METHODS Immortalized human corneal epithelial cells (HCECs) were exposed to inactive Aspergillus fumigatus (A. fumigatus) conidia at 0, 4, 8, 12, 16, and 24h respectively. The corneas of Wistar rats were exposed to active A. fumigatus at 0, 12, 24, 48h and the normal rat corneas were used for normal control. The mRNA level of S100B was evaluated by real time quantitative reverse transcription-polymerase chain reaction (qRT-PCR). S100B protein expression in cornea epithelium was detected by immunohistochemical/immunocytochemical staining (IHC/ICC). RESULTS Histopathology revealed a significant inflammatory cell infiltration in fungal keratitis human and rat cornea. Corneal epithelial cells didn't express or rarely express S100B at baseline. A. fumigatus significantly induced S100B mRNA expression in cultured corneal epithelial cells in a time depended manner in vitro, the mRNA began to rise significantly at 8h in vitro (P<0.05) and continue to rise as time prolonged (P<0.01). In vivo, S100B mRNA level was low in the normal corneas. However, it was increased in keratitis corneas from 12h after infection (P<0.05) and reached to a peak at 24h (P<0.001). Immunochemistry revealed an obvious staining in fungal keratitis corneas as well as immortalized HCECs compared to the normal ones respectively, indicating an increased expression of S100B protein. CONCLUSION S100B exists in corneal epithelial cells and is over-expressed under A. fumigatus stimulation. S100B may play an important role in the innate immune response of the corneal epithelium during A. fumigatus infection.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Gui-Qiu Zhao
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Jing Qu
- Department of Administrative Management, the Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Cheng-Ye Che
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Jing Lin
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Nan Jiang
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Han Zhao
- Department of Pathology, the Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Xue-Jun Wang
- Department of Pharmacy, the Second People's Hospital of Linyi, Linyi 276499, Shandong Province, China
| |
Collapse
|
130
|
Bangert A, Andrassy M, Müller AM, Bockstahler M, Fischer A, Volz CH, Leib C, Göser S, Korkmaz-Icöz S, Zittrich S, Jungmann A, Lasitschka F, Pfitzer G, Müller OJ, Katus HA, Kaya Z. Critical role of RAGE and HMGB1 in inflammatory heart disease. Proc Natl Acad Sci U S A 2016; 113:E155-64. [PMID: 26715748 PMCID: PMC4720305 DOI: 10.1073/pnas.1522288113] [Citation(s) in RCA: 130] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Autoimmune response to cardiac troponin I (TnI) induces inflammation and fibrosis in the myocardium. High-mobility group box 1 (HMGB1) is a multifunctional protein that exerts proinflammatory activity by mainly binding to receptor for advanced glycation end products (RAGE). The involvement of the HMGB1-RAGE axis in the pathogenesis of inflammatory cardiomyopathy is yet not fully understood. Using the well-established model of TnI-induced experimental autoimmune myocarditis (EAM), we demonstrated that both local and systemic HMGB1 protein expression was elevated in wild-type (wt) mice after TnI immunization. Additionally, pharmacological inhibition of HMGB1 using glycyrrhizin or anti-HMGB1 antibody reduced inflammation in hearts of TnI-immunized wt mice. Furthermore, RAGE knockout (RAGE-ko) mice immunized with TnI showed no structural or physiological signs of cardiac impairment. Moreover, cardiac overexpression of HMGB1 using adeno-associated virus (AAV) vectors induced inflammation in the hearts of both wt and RAGE-ko mice. Finally, patients with myocarditis displayed increased local and systemic HMGB1 and soluble RAGE (sRAGE) expression. Together, our study highlights that HMGB1 and its main receptor, RAGE, appear to be crucial factors in the pathogenesis of TnI-induced EAM, because inhibition of HMGB1 and ablation of RAGE suppressed inflammation in the heart. Moreover, the proinflammatory effect of HMGB1 is not necessarily dependent on RAGE only. Other receptors of HMGB1 such as Toll-like receptors (TLRs) may also be involved in disease pathogenesis. These findings could be confirmed by the clinical relevance of HMGB1 and sRAGE. Therefore, blockage of one of these molecules might represent a novel therapeutic strategy in the treatment of autoimmune myocarditis and inflammatory cardiomyopathy.
Collapse
Affiliation(s)
- Anna Bangert
- Department of Internal Medicine III, University of Heidelberg, 69120 Heidelberg, Germany
| | - Martin Andrassy
- Department of Internal Medicine III, University of Heidelberg, 69120 Heidelberg, Germany
| | - Anna-Maria Müller
- Department of Internal Medicine III, University of Heidelberg, 69120 Heidelberg, Germany
| | - Mariella Bockstahler
- Department of Internal Medicine III, University of Heidelberg, 69120 Heidelberg, Germany
| | - Andrea Fischer
- Department of Internal Medicine III, University of Heidelberg, 69120 Heidelberg, Germany
| | - Christian H Volz
- Department of Internal Medicine III, University of Heidelberg, 69120 Heidelberg, Germany
| | - Christoph Leib
- Department of Internal Medicine III, University of Heidelberg, 69120 Heidelberg, Germany
| | - Stefan Göser
- Department of Internal Medicine III, University of Heidelberg, 69120 Heidelberg, Germany
| | - Sevil Korkmaz-Icöz
- Department of Cardiac Surgery, University of Heidelberg, 69120 Heidelberg, Germany
| | - Stefan Zittrich
- Institute of Vegetative Physiology, University of Cologne, 50931 Cologne, Germany
| | - Andreas Jungmann
- Department of Internal Medicine III, University of Heidelberg, 69120 Heidelberg, Germany
| | - Felix Lasitschka
- Institute of Pathology, University of Heidelberg, 69120 Heidelberg, Germany
| | - Gabriele Pfitzer
- Institute of Vegetative Physiology, University of Cologne, 50931 Cologne, Germany
| | - Oliver J Müller
- Department of Internal Medicine III, University of Heidelberg, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, 69120 Heidelberg, Germany
| | - Hugo A Katus
- Department of Internal Medicine III, University of Heidelberg, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, 69120 Heidelberg, Germany
| | - Ziya Kaya
- Department of Internal Medicine III, University of Heidelberg, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, 69120 Heidelberg, Germany
| |
Collapse
|
131
|
Changyaleket B, Xu H, Vetri F, Valyi-Nagy T, Paisansathan C, Chong ZZ, Pelligrino DA, Testai FD. Intracerebroventricular application of S100B selectively impairs pial arteriolar dilating function in rats. Brain Res 2016; 1634:171-178. [PMID: 26773687 DOI: 10.1016/j.brainres.2015.12.061] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 12/22/2015] [Accepted: 12/29/2015] [Indexed: 10/22/2022]
Abstract
S100B is an astrocyte-derived protein that can act through the receptor for advanced glycation endproducts (RAGE) to mediate either "trophic" or "toxic" responses. Its levels increase in many neurological conditions with associated microvascular dysregulation, such as subarachnoid hemorrhage (SAH) and traumatic brain injury. The role of S100B in the pathogenesis of microvasculopathy has not been addressed. This study was designed to examine whether S100B alters pial arteriolar vasodilating function. Rats were randomized to receive (1) artificial cerebrospinal fluid (aCSF), (2) exogenous S100B, and (3) exogenous S100B+the decoy soluble RAGE (sRAGE). S100B was infused intracerebroventricularly (icv) using an osmotic pump and its levels in the CSF were adjusted to achieve a concentration similar to what we observed in SAH. After 48 h of continuous icv infusion, a cranial window/intravital microscopy was applied to animals for evaluation of pial arteriolar dilating responses to sciatic nerve stimulation (SNS), hypercapnia, and topical suffusion of vasodilators including acetylcholine (ACh), s-nitroso-N-acetyl penicillamine (SNAP), or adenosine (ADO). Pial arteriolar dilating responses were calculated as the percentage change of arteriolar diameter in relation to baseline. The continuous S100B infusion for 48 h was associated with reduced responses to the neuronal-dependent vasodilator SNS (p<0.05) and the endothelial-dependent vasodilator ACh (p<0.05), compared to controls. The inhibitory effects of S100B were prevented by sRAGE. On the other hand, S100B did not alter the responses elicited by vascular smooth muscle cell-dependent vasodilators, namely hypercapnia, SNAP, or ADO. These findings indicate that S100B regulates neuronal and endothelial dependent cerebral arteriolar dilation and suggest that this phenomenon is mediated through RAGE-associated pathways.
Collapse
Affiliation(s)
- Benjarat Changyaleket
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, IL, United States
| | - Haoliang Xu
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, IL, United States; Department of Pathology, University of Illinois at Chicago, Chicago, IL, United States.
| | - Francesco Vetri
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, IL, United States
| | - Tibor Valyi-Nagy
- Department of Pathology, University of Illinois at Chicago, Chicago, IL, United States
| | | | - Zhao Zhong Chong
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, IL, United States
| | - Dale A Pelligrino
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, IL, United States
| | - Fernando D Testai
- Department of Neurology and Rehabilitation, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
132
|
Juranek JK, Daffu GK, Wojtkiewicz J, Lacomis D, Kofler J, Schmidt AM. Receptor for Advanced Glycation End Products and its Inflammatory Ligands are Upregulated in Amyotrophic Lateral Sclerosis. Front Cell Neurosci 2015; 9:485. [PMID: 26733811 PMCID: PMC4686801 DOI: 10.3389/fncel.2015.00485] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 11/30/2015] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal motor neuron disorder of largely unknown pathogenesis. Recent studies suggest that enhanced oxidative stress and neuroinflammation contribute to the progression of the disease. Mounting evidence implicates the receptor for advanced glycation end-products (RAGE) as a significant contributor to the pathogenesis of certain neurodegenerative diseases and chronic conditions. It is hypothesized that detrimental actions of RAGE are triggered upon binding to its ligands, such as AGEs (advanced glycation end products), S100/calgranulin family members, and High Mobility Group Box-1 (HMGB1) proteins. Here, we examined the expression of RAGE and its ligands in human ALS spinal cord. Tissue samples from age-matched human control and ALS spinal cords were tested for the expression of RAGE, carboxymethyllysine (CML) AGE, S100B, and HMGB1, and intensity of the immunofluorescent and immunoblotting signals was assessed. We found that the expression of both RAGE and its ligands was significantly increased in the spinal cords of ALS patients versus age-matched control subjects. Our study is the first report describing co-expression of both RAGE and its ligands in human ALS spinal cords. These findings suggest that further probing of RAGE as a mechanism of neurodegeneration in human ALS is rational.
Collapse
Affiliation(s)
- Judyta K Juranek
- Department of Surgery, Columbia University Medical CenterNew York, NY, USA; Department of Medicine, New York University Langone Medical Center - New York University School of MedicineNew York, NY, USA
| | - Gurdip K Daffu
- Department of Medicine, New York University Langone Medical Center - New York University School of Medicine New York, NY, USA
| | - Joanna Wojtkiewicz
- Department of Pathophysiology, University of Warmia and Mazury Olsztyn, Poland
| | - David Lacomis
- Department of Neurology, University of Pittsburgh Pittsburgh, PA, USA
| | - Julia Kofler
- Department of Pathology, University of Pittsburgh Pittsburgh, PA, USA
| | - Ann Marie Schmidt
- Department of Surgery, Columbia University Medical CenterNew York, NY, USA; Department of Medicine, New York University Langone Medical Center - New York University School of MedicineNew York, NY, USA
| |
Collapse
|
133
|
Ramasamy R, Shekhtman A, Schmidt AM. The multiple faces of RAGE--opportunities for therapeutic intervention in aging and chronic disease. Expert Opin Ther Targets 2015; 20:431-46. [PMID: 26558318 DOI: 10.1517/14728222.2016.1111873] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION This review focuses on the multi-ligand receptor of the immunoglobulin superfamily--receptor for advanced glycation endproducts (RAGE). The accumulation of the multiple ligands of RAGE in cellular stress milieux links RAGE to the pathobiology of chronic disease and natural aging. AREAS COVERED In this review, we present a discussion on the ligands of RAGE and the implications of these ligand families in disease. We review the recent literature on the role of ligand-RAGE interaction in the consequences of natural aging; the macro- and microvascular complications of diabetes; obesity and insulin resistance; autoimmune disorders and chronic inflammation; and tumors and Alzheimer's disease. We discuss the mechanisms of RAGE signaling through its intracellular binding effector molecule--the formin DIAPH1. Physicochemical evidence of how the RAGE cytoplasmic domain binds to the FH1 (formin homology 1) domain of DIAPH1, and the consequences thereof, are also reviewed. EXPERT OPINION We discuss the modalities of RAGE antagonism currently in preclinical and clinical studies. Finally, we present the rationale behind potentially targeting the RAGE cytoplasmic domain-DIAPH1 interaction as a logical strategy for therapeutic intervention in the pathological settings of chronic diseases and aging wherein RAGE ligands accumulate and signal.
Collapse
Affiliation(s)
- Ravichandran Ramasamy
- a Diabetes Research Program, Division of Endocrinology, Department of Medicine , New York University Langone Medical Center , New York , NY 10016 , USA
| | - Alexander Shekhtman
- b Department of Chemistry , University at Albany, State University of New York , Albany , NY 12222 , USA
| | - Ann Marie Schmidt
- a Diabetes Research Program, Division of Endocrinology, Department of Medicine , New York University Langone Medical Center , New York , NY 10016 , USA
| |
Collapse
|
134
|
Huang J, Xie Y, Sun X, Zeh HJ, Kang R, Lotze MT, Tang D. DAMPs, ageing, and cancer: The 'DAMP Hypothesis'. Ageing Res Rev 2015; 24:3-16. [PMID: 25446804 DOI: 10.1016/j.arr.2014.10.004] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2014] [Revised: 09/24/2014] [Accepted: 10/22/2014] [Indexed: 12/25/2022]
Abstract
Ageing is a complex and multifactorial process characterized by the accumulation of many forms of damage at the molecular, cellular, and tissue level with advancing age. Ageing increases the risk of the onset of chronic inflammation-associated diseases such as cancer, diabetes, stroke, and neurodegenerative disease. In particular, ageing and cancer share some common origins and hallmarks such as genomic instability, epigenetic alteration, aberrant telomeres, inflammation and immune injury, reprogrammed metabolism, and degradation system impairment (including within the ubiquitin-proteasome system and the autophagic machinery). Recent advances indicate that damage-associated molecular pattern molecules (DAMPs) such as high mobility group box 1, histones, S100, and heat shock proteins play location-dependent roles inside and outside the cell. These provide interaction platforms at molecular levels linked to common hallmarks of ageing and cancer. They can act as inducers, sensors, and mediators of stress through individual plasma membrane receptors, intracellular recognition receptors (e.g., advanced glycosylation end product-specific receptors, AIM2-like receptors, RIG-I-like receptors, and NOD1-like receptors, and toll-like receptors), or following endocytic uptake. Thus, the DAMP Hypothesis is novel and complements other theories that explain the features of ageing. DAMPs represent ideal biomarkers of ageing and provide an attractive target for interventions in ageing and age-associated diseases.
Collapse
|
135
|
Glaser N, Lo W, Tancredi D, Orgain M, Puvenna V, Janigro D, O׳Donnell M. Levels of S100B in brain and blood of rats with diabetic ketoacidosis. Brain Res 2015; 1624:536-544. [DOI: 10.1016/j.brainres.2015.07.044] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 07/23/2015] [Accepted: 07/24/2015] [Indexed: 01/01/2023]
|
136
|
Aging causes morphological alterations in astrocytes and microglia in human substantia nigra pars compacta. Neurobiol Aging 2015; 36:3321-3333. [PMID: 26433682 DOI: 10.1016/j.neurobiolaging.2015.08.024] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 07/30/2015] [Accepted: 08/25/2015] [Indexed: 12/20/2022]
Abstract
Age being a risk factor for Parkinson's disease, assessment of age-related changes in the human substantia nigra may elucidate its pathogenesis. Increase in Marinesco bodies, α-synuclein, free radicals and so forth in the aging nigral neurons are clear indicators of neurodegeneration. Here, we report the glial responses in aging human nigra. The glial numbers were determined on Nissl-stained sections. The expression of glial fibrillary acidic protein, S100β, 2', 3'-cyclic nucleotide 3' phosphodiesterase, and Iba1 was assessed on cryosections of autopsied midbrains by immunohistochemistry and densitometry. The glial counts showed a biphasic increase, of which, the first prominent phase from fetal age to birth could be physiological gliogenesis whereas the second one after middle age may reflect mild age-related gliosis. Astrocytic morphology was altered, but glial fibrillary acidic protein expression increased only mildly. Presence of type-4 microglia suggests possibility of neuroinflammation. Mild reduction in 2', 3'-cyclic nucleotide 3' phosphodiesterase-labeled area denotes subtle demyelination. Stable age-related S100β expression indicates absence of calcium overload. Against the expected prominent gliosis, subtle age-related morphological alterations in human nigral glia attribute them a participatory role in aging.
Collapse
|
137
|
Niven J, Hoare J, McGowan D, Devarajan G, Itohara S, Gannagé M, Teismann P, Crane I. S100B Up-Regulates Macrophage Production of IL1β and CCL22 and Influences Severity of Retinal Inflammation. PLoS One 2015. [PMID: 26204512 PMCID: PMC4512682 DOI: 10.1371/journal.pone.0132688] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
S100B is a Ca2+ binding protein and is typically associated with brain and CNS disorders. However, the role of S100B in an inflammatory situation is not clear. The aim of the study was to determine whether S100B is likely to influence inflammation through its effect on macrophages. A murine macrophage cell line (RAW 264.7) and primary bone marrow derived macrophages were used for in vitro studies and a model of retinal inflammatory disease in which pathogenesis is highly dependent on macrophage infiltration, Experimental Autoimmune Uveoretinitis, for in vitro study. Experimental Autoimmune Uveoretinitis is a model for the human disease posterior endogenous uveoretinitis, a potentially blinding condition, with an autoimmune aetiology, that mainly affects the working age group. To date the involvement of S100B in autoimmune uveoretinitis has not been investigated. Real-time PCR array analysis on RAW 246.7 cells indicated up-regulation of gene expression for various cytokines/chemokines in response to S100B, IL-1β and CCL22 in particular and this was confirmed by real-time PCR. In addition flow cytometry and ELISA confirmed up-regulation of protein production in response to S100B for pro-IL-1β and CCL22 respectively. This was the case for both RAW 264.7 cells and bone marrow derived macrophages. Induction of EAU with retinal antigen in mice in which S100B had been deleted resulted in a significantly reduced level of disease compared to wild-type mice, as determined by topical endoscopic fundus imaging and histology grading. Macrophage infiltration was also significantly reduced in S100B deleted mice. Real-time PCR analysis indicated that this was associated with reduction in CCL22 and IL-1β in retinas from S100B knock-out mice. In conclusion S100B augments the inflammatory response in uveoretinitis and this is likely to be, at least in part, via a direct effect on macrophages.
Collapse
Affiliation(s)
- Jennifer Niven
- Division of Applied Medicine, University of Aberdeen Institute of Medical Sciences, Foresterhill, Aberdeen, Scotland, United Kingdom
- Division of Rheumatology and Department of Pathology and Immunology, School of Medicine, University of Geneva, Geneva, Switzerland
| | - Joseph Hoare
- Division of Applied Medicine, University of Aberdeen Institute of Medical Sciences, Foresterhill, Aberdeen, Scotland, United Kingdom
| | - Debbie McGowan
- Division of Applied Medicine, University of Aberdeen Institute of Medical Sciences, Foresterhill, Aberdeen, Scotland, United Kingdom
| | - Gayathri Devarajan
- Division of Applied Medicine, University of Aberdeen Institute of Medical Sciences, Foresterhill, Aberdeen, Scotland, United Kingdom
| | | | - Monique Gannagé
- Division of Rheumatology and Department of Pathology and Immunology, School of Medicine, University of Geneva, Geneva, Switzerland
| | - Peter Teismann
- Division of Applied Medicine, University of Aberdeen Institute of Medical Sciences, Foresterhill, Aberdeen, Scotland, United Kingdom
| | - Isabel Crane
- Division of Applied Medicine, University of Aberdeen Institute of Medical Sciences, Foresterhill, Aberdeen, Scotland, United Kingdom
- * E-mail:
| |
Collapse
|
138
|
Barateiro A, Afonso V, Santos G, Cerqueira JJ, Brites D, van Horssen J, Fernandes A. S100B as a Potential Biomarker and Therapeutic Target in Multiple Sclerosis. Mol Neurobiol 2015; 53:3976-3991. [DOI: 10.1007/s12035-015-9336-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 07/01/2015] [Indexed: 12/30/2022]
|
139
|
Litwinoff E, Hurtado Del Pozo C, Ramasamy R, Schmidt AM. Emerging Targets for Therapeutic Development in Diabetes and Its Complications: The RAGE Signaling Pathway. Clin Pharmacol Ther 2015; 98:135-44. [PMID: 25974754 DOI: 10.1002/cpt.148] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Revised: 05/08/2015] [Accepted: 05/08/2015] [Indexed: 12/16/2022]
Abstract
Types 1 and 2 diabetes are on the rise worldwide. Although the treatment of hyperglycemia has benefited from recent advances, aggressive efforts to maintain euglycemia may be fraught with risk, especially in older subjects or in subjects vulnerable to hypoglycemic unawareness. Hence, strategies to prevent and treat the complications of hyperglycemia are essential. In this review we summarize recent updates on the biology of the receptor for advanced glycation endproducts (RAGE) in the pathogenesis of both micro- and macrovascular complications of diabetes, insights from the study of mouse models of obesity and diabetic complications, and from associative studies in human subjects. The study of the mechanisms and consequences of the interaction of the RAGE cytoplasmic domain with the formin, mDia1, in RAGE signal transduction, will be discussed. Lastly, we review the "state-of-the-art" on RAGE-directed therapeutics. Tackling RAGE/mDia1 may identify a novel class of therapeutics preventing diabetes and its complications.
Collapse
Affiliation(s)
- Ems Litwinoff
- Diabetes Research Program, Division of Endocrinology, Department of Medicine, New York University School of Medicine, New York, New York, USA
| | - C Hurtado Del Pozo
- Diabetes Research Program, Division of Endocrinology, Department of Medicine, New York University School of Medicine, New York, New York, USA
| | - R Ramasamy
- Diabetes Research Program, Division of Endocrinology, Department of Medicine, New York University School of Medicine, New York, New York, USA
| | - A M Schmidt
- Diabetes Research Program, Division of Endocrinology, Department of Medicine, New York University School of Medicine, New York, New York, USA
| |
Collapse
|
140
|
Luo J, Liang M, Mitch WE, Danesh FR, Yu M, Cheng J. FSP-1 Impairs the Function of Endothelium Leading to Failure of Arteriovenous Grafts in Diabetic Mice. Endocrinology 2015; 156:2200-10. [PMID: 25774552 PMCID: PMC4430603 DOI: 10.1210/en.2014-1841] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
To understand how endothelial cell (EC) dysfunction contributes to the failure of arteriovenous graft (AVG), we investigated the role of fibroblast-specific protein 1 (FSP-1) in cultured ECs and a mouse AVG model. In vitro, we uncovered a new FSP-1-dependent pathway that activates rho-associated, coiled-coil-containing protein kinase 1 (ROCK1) in ECs, leading to phosphorylation of myosin light chain 2 resulting in EC dysfunction. In cultured ECs, high glucose stimulated FSP-1 expression and increased permeability of an EC monolayer. The increase in permeability by the high glucose concentration was mediated by FSP-1 expression. Treatment of cultured ECs with FSP-1 caused leakage of the endothelial barrier plus increased expression of adhesion molecules and decreased expression of junction molecules. These responses were initiated by binding of FSP-1 to receptor for advanced glycation end products, which resulted in ROCK1 activation. In vivo, diabetes increased infiltration of inflammatory cells into AVGs and stimulated neointima formation. Increased FSP-1 expression and ROCK1 activation were found in AVGs of diabetic mice. Blocking FSP-1 suppressed diabetes-induced ROCK1 activation in AVGs. In mice with FSP-1 knockout or with ROCK1 knockout, accumulation of inflammatory cells and neointima formation in AVG were attenuated despite diabetes. Thus, mechanisms of inhibiting FSP-1 in ECs could improve AVG function.
Collapse
Affiliation(s)
- Jinlong Luo
- Nephrology Division (J.L., M.L., W.E.M., M.Y., J.C.), Baylor College of Medicine, Houston, Texas 77030; Emergency Medicine (F.R.D.), University of Texas MD Anderson Cancer Center, Houston, Texas 77030; and Department of Emergency (J.L.), Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China 430074
| | | | | | | | | | | |
Collapse
|
141
|
McVicar CM, Ward M, Colhoun LM, Guduric-Fuchs J, Bierhaus A, Fleming T, Schlotterer A, Kolibabka M, Hammes HP, Chen M, Stitt AW. Role of the receptor for advanced glycation endproducts (RAGE) in retinal vasodegenerative pathology during diabetes in mice. Diabetologia 2015; 58:1129-37. [PMID: 25687235 PMCID: PMC4392170 DOI: 10.1007/s00125-015-3523-x] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 01/16/2015] [Indexed: 01/18/2023]
Abstract
AIMS/HYPOTHESIS The receptor for AGEs (RAGE) is linked to proinflammatory pathology in a range of tissues. The objective of this study was to assess the potential modulatory role of RAGE in diabetic retinopathy. METHODS Diabetes was induced in wild-type (WT) and Rage (-/-) mice (also known as Ager (-/-) mice) using streptozotocin while non-diabetic control mice received saline. For all groups, blood glucose, HbA1c and retinal levels of methylglyoxal (MG) were evaluated up to 24 weeks post diabetes induction. After mice were killed, retinal glia and microglial activation, vasopermeability, leucostasis and degenerative microvasculature changes were determined. RESULTS Retinal expression of RAGE in WT diabetic mice was increased after 12 weeks (p < 0.01) but not after 24 weeks. Rage (-/-) mice showed comparable diabetes but accumulated less MG and this corresponded to enhanced activity of the MG-detoxifying enzyme glyoxalase I in their retina when compared with WT mice. Diabetic Rage (-/-) mice showed significantly less vasopermeability, leucostasis and microglial activation (p < 0.05-0.001). Rage (-/-) mice were also protected against diabetes-related retinal acellular capillary formation (p < 0.001) but not against pericyte loss. CONCLUSIONS/INTERPRETATION Rage (-/-) in diabetic mice is protective against many retinopathic lesions, especially those related to innate immune responses. Inhibition of RAGE could be a therapeutic option to prevent diabetic retinopathy.
Collapse
Affiliation(s)
- Carmel M. McVicar
- Centre for Experimental Medicine, Queen’s University Belfast, Belfast, BT12 6BA Northern Ireland UK
| | - Micheal Ward
- Centre for Experimental Medicine, Queen’s University Belfast, Belfast, BT12 6BA Northern Ireland UK
| | - Liza M. Colhoun
- Centre for Experimental Medicine, Queen’s University Belfast, Belfast, BT12 6BA Northern Ireland UK
| | - Jasenka Guduric-Fuchs
- Centre for Experimental Medicine, Queen’s University Belfast, Belfast, BT12 6BA Northern Ireland UK
| | - Angelika Bierhaus
- Department of Medicine and Clinical Chemistry, University of Heidelberg, Heidelberg, Germany
| | - Thomas Fleming
- Department of Medicine and Clinical Chemistry, University of Heidelberg, Heidelberg, Germany
| | - Andreas Schlotterer
- Department of Medicine and Clinical Chemistry, University of Heidelberg, Heidelberg, Germany
| | - Matthias Kolibabka
- Department of Medicine and Clinical Chemistry, University of Heidelberg, Heidelberg, Germany
| | - Hans-Peter Hammes
- Department of Medicine and Clinical Chemistry, University of Heidelberg, Heidelberg, Germany
| | - Mei Chen
- Centre for Experimental Medicine, Queen’s University Belfast, Belfast, BT12 6BA Northern Ireland UK
| | - Alan W. Stitt
- Centre for Experimental Medicine, Queen’s University Belfast, Belfast, BT12 6BA Northern Ireland UK
| |
Collapse
|
142
|
Chuang CT, Guh JY, Lu CY, Wang YT, Chen HC, Chuang LY. Steap4 attenuates high glucose and S100B-induced effects in mesangial cells. J Cell Mol Med 2015; 19:1234-44. [PMID: 25817898 PMCID: PMC4459839 DOI: 10.1111/jcmm.12472] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 10/09/2014] [Indexed: 12/14/2022] Open
Abstract
Six-transmembrane epithelial antigen of prostate 4 (Steap4)-knockout mice develop hyperglycaemia and inflammation whereas Steap4 overexpression attenuates atherosclerosis in diabetic mice. Thus, we studied the roles of Steap4 in high glucose (HG, 27.5 mM) or S100B (1 μM, a ligand for the receptor for advanced glycation end-product or RAGE)-induced effects in mouse mesangial (MES13) cells. We found that HG-induced Steap4 protein expression was dependent on S100B. HG increased cell membrane, but not cytosolic, Steap4 protein expression. HG increased protein-protein interaction between Steap4 and S100B, which was confirmed by mass spectrometry of immunoprecipitated S100B. SP600125, LY294002 and AG490 attenuated S100B-induced Steap4 protein expression or gene transcriptional activity. A mutation in signal transducer and activator of transcription 3 (Stat3) site 2 of the Steap4 promoter constructs resulted in a marked decrease in HG or S100B-induced activation of Steap4 gene transcription. Overexpression of Steap4 attenuates HG or S100B-induced collagen IV, fibronectin and cyclooxygenase 2 protein expression. Overexpression of Steap4 attenuates HG or S100B-induced transforming growth factor-β (TGF-β). Moreover, overexpression of Steap4 attenuates S100B-induced signalling. Finally, overexpressing Steap4 attenuated renal expression of fibronectin, S100B, TGF-β, type IV collagen, p-Akt, p-extracellular signal regulated kinase 1/2 and p-Stat3 in streptozotocin-diabetic mice. Thus, overexpression of Steap4 attenuated HG or S100B-induced effects in MES13 cells and attenuated some of S100B-induced effects in diabetic mouse kidneys.
Collapse
Affiliation(s)
- Chao-Tang Chuang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jinn-Yuh Guh
- Department of Internal Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chi-Yu Lu
- Department of Biochemistry, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yeng-Tseng Wang
- Department of Biochemistry, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hung-Chun Chen
- Department of Internal Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Lea-Yea Chuang
- Department of Biochemistry, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Center for Lipid and Glycomedicine Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
143
|
Expression of inflammatory markers in a genetic rodent model of depression. Behav Brain Res 2015; 281:348-57. [DOI: 10.1016/j.bbr.2014.09.025] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 08/20/2014] [Accepted: 09/15/2014] [Indexed: 12/11/2022]
|
144
|
Chen B, Miller AL, Rebelatto M, Brewah Y, Rowe DC, Clarke L, Czapiga M, Rosenthal K, Imamichi T, Chen Y, Chang CS, Chowdhury PS, Naiman B, Wang Y, Yang D, Humbles AA, Herbst R, Sims GP. S100A9 induced inflammatory responses are mediated by distinct damage associated molecular patterns (DAMP) receptors in vitro and in vivo. PLoS One 2015; 10:e0115828. [PMID: 25706559 PMCID: PMC4338059 DOI: 10.1371/journal.pone.0115828] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 12/02/2014] [Indexed: 01/12/2023] Open
Abstract
Release of endogenous damage associated molecular patterns (DAMPs), including members of the S100 family, are associated with infection, cellular stress, tissue damage and cancer. The extracellular functions of this family of calcium binding proteins, particularly S100A8, S100A9 and S100A12, are being delineated. They appear to mediate their functions via receptor for advanced glycation endproducts (RAGE) or TLR4, but there remains considerable uncertainty over the relative physiological roles of these DAMPs and their pattern recognition receptors. In this study, we surveyed the capacity of S100 proteins to induce proinflammatory cytokines and cell migration, and the contribution RAGE and TLR4 to mediate these responses in vitro. Using adenoviral delivery of murine S100A9, we also examined the potential for S100A9 homodimers to trigger lung inflammation in vivo. S100A8, S100A9 and S100A12, but not the S100A8/A9 heterodimer, induced modest levels of TLR4-mediated cytokine production from human PBMC. In contrast, for most S100s including S100A9, RAGE blockade inhibited S100-mediated cell migration of THP1 cells and major leukocyte populations, whereas TLR4-blockade had no effect. Intranasal administration of murine S100A9 adenovirus induced a specific, time-dependent predominately macrophage infiltration that coincided with elevated S100A9 levels and proinflammatory cytokines in the BAL fluid. Inflammatory cytokines were markedly ablated in the TLR4-defective mice, but unexpectedly the loss of TLR4 signaling or RAGE-deficiency did not appreciably impact the S100A9-mediated lung pathology or the inflammatory cell infiltrate in the alveolar space. These data demonstrate that physiological levels of S100A9 homodimers can trigger an inflammatory response in vivo, and despite the capacity of RAGE and TLR4 blockade to inhibit responses in vitro, the response is predominately independent of both these receptors.
Collapse
Affiliation(s)
- Bo Chen
- MedImmune LLC, One MedImmune Way, Gaithersburg, Maryland 20878, United States of America
| | - Allison L. Miller
- MedImmune LLC, One MedImmune Way, Gaithersburg, Maryland 20878, United States of America
| | - Marlon Rebelatto
- MedImmune LLC, One MedImmune Way, Gaithersburg, Maryland 20878, United States of America
| | - Yambasu Brewah
- MedImmune LLC, One MedImmune Way, Gaithersburg, Maryland 20878, United States of America
| | - Daniel C. Rowe
- MedImmune LLC, One MedImmune Way, Gaithersburg, Maryland 20878, United States of America
| | - Lori Clarke
- MedImmune LLC, One MedImmune Way, Gaithersburg, Maryland 20878, United States of America
| | - Meggan Czapiga
- MedImmune LLC, One MedImmune Way, Gaithersburg, Maryland 20878, United States of America
| | - Kim Rosenthal
- MedImmune LLC, One MedImmune Way, Gaithersburg, Maryland 20878, United States of America
| | - Tomozumi Imamichi
- MedImmune LLC, One MedImmune Way, Gaithersburg, Maryland 20878, United States of America
- Laboratory of Human Retrovirology, Applied and Developmental Directorate, Building 550 Room 126, Leidos Biomedical Research Inc, Frederick National Laboratory for Cancer Research, Frederick, Maryland 21702, United States of America
| | - Yan Chen
- MedImmune LLC, One MedImmune Way, Gaithersburg, Maryland 20878, United States of America
| | - Chew-Shun Chang
- MedImmune LLC, One MedImmune Way, Gaithersburg, Maryland 20878, United States of America
| | - Partha S. Chowdhury
- MedImmune LLC, One MedImmune Way, Gaithersburg, Maryland 20878, United States of America
| | - Brian Naiman
- MedImmune LLC, One MedImmune Way, Gaithersburg, Maryland 20878, United States of America
| | - Yue Wang
- MedImmune LLC, One MedImmune Way, Gaithersburg, Maryland 20878, United States of America
| | - De Yang
- Laboratory of Molecular Immunoregulation, National Cancer Institute, Frederick National Laboratory for Cancer Research, Frederick, Maryland 21702, United States of America
| | - Alison A. Humbles
- MedImmune LLC, One MedImmune Way, Gaithersburg, Maryland 20878, United States of America
| | - Ronald Herbst
- MedImmune LLC, One MedImmune Way, Gaithersburg, Maryland 20878, United States of America
| | - Gary P. Sims
- MedImmune LLC, One MedImmune Way, Gaithersburg, Maryland 20878, United States of America
- * E-mail:
| |
Collapse
|
145
|
Batkulwar KB, Bansode SB, Patil GV, Godbole RK, Kazi RS, Chinnathambi S, Shanmugam D, Kulkarni MJ. Investigation of phosphoproteome in RAGE signaling. Proteomics 2015; 15:245-259. [PMID: 25315903 DOI: 10.1002/pmic.201400169] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 08/14/2014] [Accepted: 10/06/2014] [Indexed: 01/03/2025]
Abstract
The receptor for advanced glycation end products (RAGE) is one of the most important proteins implicated in diabetes, cardiovascular diseases, neurodegenerative diseases, and cancer. It is a pattern recognition receptor by virtue of its ability to interact with multiple ligands, RAGE activates several signal transduction pathways through involvement of various kinases that phosphorylate their respective substrates. Only few substrates have been known to be phosphorylated in response to activation by RAGE (e.g., nuclear factor kappa B); however, it is possible that these kinases can phosphorylate multiple substrates depending upon their expression and localization, leading to altered cellular responses in different cell types and conditions. One such example is, glycogen synthase kinase 3 beta which is known to phosphorylate glycogen synthase, acts downstream to RAGE, and hyperphosphorylates microtubule-associated protein tau causing neuronal damage. Thus, it is important to understand the role of various RAGE-activated kinases and their substrates. Therefore, we have reviewed here the details of RAGE-activated kinases in response to different ligands and their respective phosphoproteome. Furthermore, we discuss the analysis of the data mined for known substrates of these kinases from the PhosphoSitePlus (http://www.phosphosite.org) database, and the role of some of the important substrates involved in cancer, diabetes, cardiovascular diseases, and neurodegenerative diseases. In summary, this review provides information on RAGE-activated kinases and their phosphoproteome, which will be helpful in understanding the possible role of RAGE and its ligands in progression of diseases.
Collapse
Affiliation(s)
- Kedar B Batkulwar
- Proteomics Facility, Division of Biochemical Sciences, CSIR-National Chemical Laboratory, Pune, India
| | | | | | | | | | | | | | | |
Collapse
|
146
|
Ye H, Wang L, Yang XK, Fan LP, Wang YG, Guo L. Serum S100B levels may be associated with cerebral infarction: a meta-analysis. J Neurol Sci 2014; 348:81-8. [PMID: 25434713 DOI: 10.1016/j.jns.2014.11.010] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 10/23/2014] [Accepted: 11/07/2014] [Indexed: 10/24/2022]
Abstract
OBJECTIVE The aim of this paper is to explore the potential association of serum human soluble protein-100B protein (S100B) levels with the diagnosis and prognosis of cerebral infarction (CI). METHODS Potential relevant studies were searched for in PubMed, Springerlink, Wiley, EBSCO, Ovid, Web of Science, Wanfang databases, China National Knowledge Infrastructure (CNKI) databases and VIP databases. Two investigators extracted data and assessed studies independently. Statistical analyses were carried out with the version 12.0 STATA statistical software. RESULTS A total of 10 case-control studies that assessed the correlation of S100B serum level with CI, including 1211 subjects (patients=773, healthy controls=438) were included. The results showed that S100B serum levels in CI victims were significantly higher compared with those of the control group. According to the subgroup analysis by ethnicity, S100B serum level in CI victims was statistically significant in Asians and the control group, but no statistical significance was found in Caucasians. An additional subgroup analysis was carried out based on sample size, revealing that the S100B serum levels in CI victims in small samples were of statistical significance; however, no statistical significance was discovered in large samples. CONCLUSIONS Elevator S100B serum levels might be negatively correlated with CI, suggesting that higher serum levels of S100B could lead to more serious condition and worse prognoses for CI patients. Therefore, S100B serum levels could be regarded as a biomarker for CI, and furthermore, S100B could aide in the diagnosis and prognosis of CI.
Collapse
Affiliation(s)
- Hua Ye
- Department of Neurology, the Third Affiliated Clinical Institute of Wenzhou Medical University, Wenzhou 325000, China
| | - Lu Wang
- Department of orthopedics, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang 325000, China
| | - Xiao-Kai Yang
- Department of Neurology, the Third Affiliated Clinical Institute of Wenzhou Medical University, Wenzhou 325000, China
| | - Lu-Ping Fan
- Department of Neurology, the Third Affiliated Clinical Institute of Wenzhou Medical University, Wenzhou 325000, China
| | - Yao-Guang Wang
- Department of Neurology, the Third Affiliated Clinical Institute of Wenzhou Medical University, Wenzhou 325000, China
| | - Lei Guo
- Department of ICU, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China.
| |
Collapse
|
147
|
Chen X, Zhang L, Zhang IY, Liang J, Wang H, Ouyang M, Wu S, da Fonseca ACC, Weng L, Yamamoto Y, Yamamoto H, Natarajan R, Badie B. RAGE expression in tumor-associated macrophages promotes angiogenesis in glioma. Cancer Res 2014; 74:7285-7297. [PMID: 25326491 DOI: 10.1158/0008-5472.can-14-1240] [Citation(s) in RCA: 117] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Interaction of RAGE (the receptor for advanced glycation endproducts) with its ligands can promote tumor progression, invasion, and angiogenesis. Although blocking RAGE signaling has been proposed as a potential anticancer strategy, functional contributions of RAGE expression in the tumor microenvironment (TME) have not been investigated in detail. Here, we evaluated the effect of genetic depletion of RAGE in TME on the growth of gliomas. In both invasive and noninvasive glioma models, animal survival was prolonged in RAGE knockout (Ager(-/-)) mice. However, the improvement in survival in Ager(-/-) mice was not due to changes in tumor growth rate but rather to a reduction in tumor-associated inflammation. Furthermore, RAGE ablation in the TME abrogated angiogenesis by downregulating the expression of proangiogenic factors, which prevented normal vessel formation, thereby generating a leaky vasculature. These alterations were most prominent in noninvasive gliomas, in which the expression of VEGF and proinflammatory cytokines were also lower in tumor-associated macrophages (TAM) in Ager(-/-) mice. Interestingly, reconstitution of Ager(-/-) TAM with wild-type microglia or macrophages normalized tumor vascularity. Our results establish that RAGE signaling in glioma-associated microglia and TAM drives angiogenesis, underscoring the complex role of RAGE and its ligands in gliomagenesis.
Collapse
Affiliation(s)
- Xuebo Chen
- Department of General Surgery, China Japan Union Hospital of Jilin University, Changchun, Jilin Province, P.R.China
| | - Leying Zhang
- Division of Neurosurgery, City of Hope Beckman Research Institute
| | - Ian Y Zhang
- Division of Neurosurgery, City of Hope Beckman Research Institute
| | - Junling Liang
- Research Center of Siyuan Natural Pharmacy and Biotoxicology, College of Life Sciences, Zhejiang University, Hangzhou, P.R. China
| | - Huaqing Wang
- Department of Emergency Surgery, Qilu Hospital of Shandong University, Jinan, Shandong Province, P.R. China
| | - Mao Ouyang
- Department of Cardiology, Third Xiangya Hospital, Central South University, Changsha Hunan, P.R. China
| | - Shihua Wu
- Research Center of Siyuan Natural Pharmacy and Biotoxicology, College of Life Sciences, Zhejiang University, Hangzhou, P.R. China
| | - Anna Carolina Carvalho da Fonseca
- Laboratório de Morfogênese Celular, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil, Bolsista do CNPq
| | - Lihong Weng
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Beckman Research Institute
| | - Yasuhiko Yamamoto
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University, Japan
| | - Hiroshi Yamamoto
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University, Japan
| | - Rama Natarajan
- Division of Molecular Diabetes Research, City of Hope Beckman Research Institute
| | - Behnam Badie
- Division of Neurosurgery, City of Hope Beckman Research Institute.,Department of Cancer Immunotherapeutics & Tumor Immunology, City of Hope Beckman Research Institute
| |
Collapse
|
148
|
Yeh CH, Yang ML, Lee CY, Yang CP, Li YC, Chen CJ, Kuan YH. Wogonin attenuates endotoxin-induced prostaglandin E2 and nitric oxide production via Src-ERK1/2-NFκB pathway in BV-2 microglial cells. ENVIRONMENTAL TOXICOLOGY 2014; 29:1162-1170. [PMID: 23362215 DOI: 10.1002/tox.21847] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Revised: 12/25/2012] [Accepted: 12/27/2012] [Indexed: 06/01/2023]
Abstract
Microglia are the major component of intrinsic brain immune system in neuroinflammation. Although wogonin expresses anti-inflammatory function in microglia, little is known about the molecular mechanisms of the protective effect of wogonin against microglia activation. The aim of this study was to evaluate how wogonin exerts its anti-inflammatory function in BV2 microglial cells after LPS/INFγ administration. Wogonin not only inhibited LPS/ INFγ-induced PGE2 and NO production without affecting cell viability but also exhibited parallel inhibition on LPS/INFγ-induced expression of iNOS and COX-2 in the same concentration range. While LPS/INFγ-induced expression of P-p65 and P-IκB was inhibited by wogonin-only weak inhibition on P-p38 and P-JNK were observed, whereas it significantly attenuated the P-ERK1/2 and its upstream activators P-MEK1/2 and P-Src in a parallel concentration-dependent manner. These results indicated that the blockade of PGE2 and NO production by wogonin in LPS/INFγ-stimulated BV2 cells is attributed mainly to interference in the Src-MEK1/2-ERK1/2-NFκB-signaling pathway.
Collapse
Affiliation(s)
- Chung-Hsin Yeh
- Department of Neurology, Show Chwan Memorial Hospital, Changhua, Taiwan, Republic of China
| | | | | | | | | | | | | |
Collapse
|
149
|
Coque E, Raoul C, Bowerman M. ROCK inhibition as a therapy for spinal muscular atrophy: understanding the repercussions on multiple cellular targets. Front Neurosci 2014; 8:271. [PMID: 25221469 PMCID: PMC4148024 DOI: 10.3389/fnins.2014.00271] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 08/11/2014] [Indexed: 12/28/2022] Open
Abstract
Spinal muscular atrophy (SMA) is the most common genetic disease causing infant death, due to an extended loss of motoneurons. This neuromuscular disorder results from deletions and/or mutations within the Survival Motor Neuron 1 (SMN1) gene, leading to a pathological decreased expression of functional full-length SMN protein. Emerging studies suggest that the small GTPase RhoA and its major downstream effector Rho kinase (ROCK), which both play an instrumental role in cytoskeleton organization, contribute to the pathology of motoneuron diseases. Indeed, an enhanced activation of RhoA and ROCK has been reported in the spinal cord of an SMA mouse model. Moreover, the treatment of SMA mice with ROCK inhibitors leads to an increased lifespan as well as improved skeletal muscle and neuromuscular junction pathology, without preventing motoneuron degeneration. Although motoneurons are the primary target in SMA, an increasing number of reports show that other cell types inside and outside the central nervous system contribute to SMA pathogenesis. As administration of ROCK inhibitors to SMA mice was systemic, the improvement in survival and phenotype could therefore be attributed to specific effects on motoneurons and/or on other non-neuronal cell types. In the present review, we will present the various roles of the RhoA/ROCK pathway in several SMA cellular targets including neurons, myoblasts, glial cells, cardiomyocytes and pancreatic cells as well as discuss how ROCK inhibition may ameliorate their health and function. It is most likely a concerted influence of ROCK modulation on all these cell types that ultimately lead to the observed benefits of pharmacological ROCK inhibition in SMA mice.
Collapse
Affiliation(s)
- Emmanuelle Coque
- The Institute for Neurosciences of Montpellier, Saint Eloi Hospital, Institut National de la Santé et de la Recherche Médicale UMR1051 Montpellier, France ; Université de Montpellier 1 and 2 Montpellier, France
| | - Cédric Raoul
- The Institute for Neurosciences of Montpellier, Saint Eloi Hospital, Institut National de la Santé et de la Recherche Médicale UMR1051 Montpellier, France ; Université de Montpellier 1 and 2 Montpellier, France
| | - Mélissa Bowerman
- The Institute for Neurosciences of Montpellier, Saint Eloi Hospital, Institut National de la Santé et de la Recherche Médicale UMR1051 Montpellier, France ; Université de Montpellier 1 and 2 Montpellier, France
| |
Collapse
|
150
|
Hu X, Liou AKF, Leak RK, Xu M, An C, Suenaga J, Shi Y, Gao Y, Zheng P, Chen J. Neurobiology of microglial action in CNS injuries: receptor-mediated signaling mechanisms and functional roles. Prog Neurobiol 2014; 119-120:60-84. [PMID: 24923657 PMCID: PMC4121732 DOI: 10.1016/j.pneurobio.2014.06.002] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 05/31/2014] [Accepted: 06/03/2014] [Indexed: 12/28/2022]
Abstract
Microglia are the first line of immune defense against central nervous system (CNS) injuries and disorders. These highly plastic cells play dualistic roles in neuronal injury and recovery and are known for their ability to assume diverse phenotypes. A broad range of surface receptors are expressed on microglia and mediate microglial 'On' or 'Off' responses to signals from other host cells as well as invading microorganisms. The integrated actions of these receptors result in tightly regulated biological functions, including cell mobility, phagocytosis, the induction of acquired immunity, and trophic factor/inflammatory mediator release. Over the last few years, significant advances have been made toward deciphering the signaling mechanisms related to these receptors and their specific cellular functions. In this review, we describe the current state of knowledge of the surface receptors involved in microglial activation, with an emphasis on their engagement of distinct functional programs and their roles in CNS injuries. It will become evident from this review that microglial homeostasis is carefully maintained by multiple counterbalanced strategies, including, but not limited to, 'On' and 'Off' receptor signaling. Specific regulation of theses microglial receptors may be a promising therapeutic strategy against CNS injuries.
Collapse
Affiliation(s)
- Xiaoming Hu
- Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, Fudan University, Shanghai, China; Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15240, USA.
| | - Anthony K F Liou
- Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Rehana K Leak
- Division of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA 15282, USA
| | - Mingyue Xu
- State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, Fudan University, Shanghai, China
| | - Chengrui An
- State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, Fudan University, Shanghai, China
| | - Jun Suenaga
- Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Yejie Shi
- Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Yanqin Gao
- State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, Fudan University, Shanghai, China
| | - Ping Zheng
- State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, Fudan University, Shanghai, China
| | - Jun Chen
- Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, Fudan University, Shanghai, China; Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15240, USA.
| |
Collapse
|