1
|
Hilgers RHP, Das KC. Redox Regulation of K + Channel: Role of Thioredoxin. Antioxid Redox Signal 2024. [PMID: 39099341 DOI: 10.1089/ars.2023.0416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/06/2024]
Abstract
Significance: Potassium channels regulate the influx and efflux of K+ ions in various cell types that generate and propagate action potential associated with excitation, contraction, and relaxation of various cell types. Although redox active cysteines are critically important for channel activity, the redox regulation of K+ channels by thioredoxin (Trx) has not been systematically reviewed. Recent Advances: Redox regulation of K+ channel is now increasingly recognized as drug targets in the pathological condition of several cardiovascular disease processes. The role of Trx in regulation of these channels and its implication in pathological conditions have not been adequately reviewed. This review specifically focuses on the redox-regulatory role of Trx on K+ channel structure and function in physiological and pathophysiological conditions. Critical Issues: Ion channels, including K+ channel, have been implicated in the functioning of cardiomyocyte excitation-contraction coupling, vascular hyperpolarization, cellular proliferation, and neuronal stimulation in physiological and pathophysiological conditions. Although oxidation-reduction of ion channels is critically important in their function, the role of Trx, redox regulatory protein in regulation of these channels, and its implication in pathological conditions need to be studied to gain further insight into channel function. Future Directions: Future studies need to map all redox regulatory pathways in channel structure and function using novel mouse models and redox proteomic and signal transduction studies, which modulate various currents and altered excitability of relevant cells implicated in a pathological condition. We are yet at infancy of studies related to redox control of various K+ channels and structured and focused studies with novel animal models. Antioxid. Redox Signal. 00, 00-00.
Collapse
Affiliation(s)
- Rob H P Hilgers
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| | - Kumuda C Das
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| |
Collapse
|
2
|
Patel J, Dawson VL, Dawson TM. Blocking the Self-Destruct Program of Dopamine Neurons through Macrophage Migration Inhibitory Factor Nuclease Inhibition. Mov Disord 2024; 39:644-650. [PMID: 38396375 PMCID: PMC11160583 DOI: 10.1002/mds.29748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 01/10/2024] [Accepted: 02/05/2024] [Indexed: 02/25/2024] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative condition that pathognomonically involves the death of dopaminergic neurons in the substantia nigra pars compacta, resulting in a myriad of motor and non-motor symptoms. Given the insurmountable burden of this disease on the population and healthcare system, significant efforts have been put forth toward generating disease modifying therapies. This class of treatments characteristically alters disease course, as opposed to current strategies that focus on managing symptoms. Previous literature has implicated the cell death pathway known as parthanatos in PD progression. Inhibition of this pathway by targeting poly (ADP)-ribose polymerase 1 (PARP1) prevents neurodegeneration in a model of idiopathic PD. However, PARP1 has a vast repertoire of functions within the body, increasing the probability of side effects with the long-term treatment likely necessary for clinically significant neuroprotection. Recent work culminated in the development of a novel agent targeting the macrophage migration inhibitory factor (MIF) nuclease domain, also named parthanatos-associated apoptosis-inducing factor nuclease (PAAN). This nuclease activity specifically executes the terminal step in parthanatos. Parthanatos-associated apoptosis-inducing factor nuclease inhibitor-1 was neuroprotective in multiple preclinical mouse models of PD. This piece will focus on contextualizing this discovery, emphasizing its significance, and discussing its potential implications for parthanatos-directed treatment. © 2024 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Jaimin Patel
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Valina L. Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ted M. Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
3
|
Jia X, Xi J, Tian B, Zhang Y, Wang Z, Wang F, Li Z, Long J, Wang J, Fan GH, Li Q. The Tautomerase Activity of Tumor Exosomal MIF Promotes Pancreatic Cancer Progression by Modulating MDSC Differentiation. Cancer Immunol Res 2024; 12:72-90. [PMID: 37956411 DOI: 10.1158/2326-6066.cir-23-0205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 07/28/2023] [Accepted: 11/09/2023] [Indexed: 11/15/2023]
Abstract
Pancreatic cancer is a deadly disease that is largely resistant to immunotherapy, in part because of the accumulation of immunosuppressive cells in the tumor microenvironment (TME). Much evidence suggests that tumor-derived exosomes (TDE) contribute to the immunosuppressive activity mediated by myeloid-derived suppressor cells (MDSC) within the pancreatic cancer TME. However, the underlying mechanisms remain elusive. Herein, we report that macrophage migration inhibitory factor (MIF) in TDEs has a key role in inducing MDSC formation in pancreatic cancer. We identified MIF in both human and murine pancreatic cancer-derived exosomes. Upon specific shRNA-mediated knockdown of MIF, the ability of pancreatic cancer-derived exosomes to promote MDSC differentiation was abrogated. This phenotype was rescued by reexpression of the wild-type form of MIF rather than a tautomerase-null mutant or a thiol-protein oxidoreductase-null mutant, indicating that both MIF enzyme activity sites play a role in exosome-induced MDSC formation in pancreatic cancer. RNA sequencing data indicated that MIF tautomerase regulated the expression of genes required for MDSC differentiation, recruitment, and activation. We therefore developed a MIF tautomerase inhibitor, IPG1576. The inhibitor effectively inhibited exosome-induced MDSC differentiation in vitro and reduced tumor growth in an orthotopic pancreatic cancer model, which was associated with decreased numbers of MDSCs and increased infiltration of CD8+ T cells in the TME. Collectively, our findings highlight a pivotal role for MIF in exosome-induced MDSC differentiation in pancreatic cancer and underscore the potential of MIF tautomerase inhibitors to reverse the immunosuppressive pancreatic cancer microenvironment, thereby augmenting anticancer immune responses.
Collapse
Affiliation(s)
- Xuebing Jia
- Cancer Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Pancreatic Disease, Institute of Pancreatic Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianbei Xi
- Department of Medicinal Chemistry, Immunophage Biotech Co., Ltd., Shanghai, China
| | - Binle Tian
- Cancer Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Pancreatic Disease, Institute of Pancreatic Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuanyuan Zhang
- Department of Oncology, Immunophage Biotech Co., Ltd., Shanghai, China
| | - Zhilong Wang
- Department of Oncology, Immunophage Biotech Co., Ltd., Shanghai, China
| | - Fan Wang
- Cancer Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zheng Li
- Department of Autoimmune Disease, Immunophage Biotech Co., Ltd., Shanghai, China
| | - Jiang Long
- Department of Pancreatic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - JianFei Wang
- Excecutive Office, Immunophage Biotech Co., Ltd., Shanghai, China
- Shanghai Laboratory Animal Research Center, Shanghai, China
| | - Guo-Huang Fan
- Excecutive Office, Immunophage Biotech Co., Ltd., Shanghai, China
| | - Qi Li
- Cancer Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Pancreatic Disease, Institute of Pancreatic Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
4
|
Xuan W, Xie W, Li F, Huang D, Zhu Z, Lin Y, Lu B, Yu W, Li Y, Li P. Dualistic roles and mechanistic insights of macrophage migration inhibitory factor in brain injury and neurodegenerative diseases. J Cereb Blood Flow Metab 2023; 43:341-356. [PMID: 36369735 PMCID: PMC9941868 DOI: 10.1177/0271678x221138412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 09/21/2022] [Accepted: 10/03/2022] [Indexed: 11/15/2022]
Abstract
Macrophage migration inhibitory factor (MIF) is involved in various immune-mediated pathologies and regulates both innate and adaptive immune reactions, thus being related to several acute and chronic inflammatory diseases such as rheumatoid arthritis, septic shock, and atherosclerosis. Its role in acute and chronic brain pathologies, such as stroke and neurodegenerative diseases, has attracted increasing attention in recent years. In response to stimuli like hypoxia, inflammation or infection, different cell types can rapidly release MIF, including immune cells, endothelial cells, and neuron cells. Notably, clinical data from past decades also suggested a possible link between serum MIF levels and the severity of stroke and the evolving of neurodegenerative diseases. In this review, we summarize the major and recent findings focusing on the mechanisms of MIF modulating functions in brain injury and neurodegenerative diseases, which may provide important therapeutic targets meriting further investigation.
Collapse
Affiliation(s)
- Wei Xuan
- Department of Anesthesiology, Clinical Research Center, Renji
Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai,
China
| | - Wanqing Xie
- Department of Anesthesiology, Clinical Research Center, Renji
Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai,
China
| | - Fengshi Li
- Department of Neurosurgery, Renji Hospital, Shanghai Jiao Tong
University School of Medicine, Shanghai, China
| | - Dan Huang
- Department of Anesthesiology, Clinical Research Center, Renji
Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai,
China
| | - Ziyu Zhu
- Department of Anesthesiology, Clinical Research Center, Renji
Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai,
China
| | - Yuxuan Lin
- Department of Anesthesiology, Clinical Research Center, Renji
Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai,
China
| | - Binwei Lu
- Department of Anesthesiology, Clinical Research Center, Renji
Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai,
China
| | - Weifeng Yu
- Department of Anesthesiology, Clinical Research Center, Renji
Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai,
China
| | - Yan Li
- Department of Anesthesiology, Clinical Research Center, Renji
Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai,
China
| | - Peiying Li
- Department of Anesthesiology, Clinical Research Center, Renji
Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai,
China
| |
Collapse
|
5
|
Ischemic stroke protected by ISO-1 inhibition of apoptosis via mitochondrial pathway. Sci Rep 2023; 13:2788. [PMID: 36797398 PMCID: PMC9935850 DOI: 10.1038/s41598-023-29907-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 02/13/2023] [Indexed: 02/18/2023] Open
Abstract
Macrophage migration inhibitory factor (MIF) is an immune mediator associated with inflammation, which is upregulated after ischemia in brain tissue. ISO-1 is a potent inhibitor of MIF tautomerase and can protect neurons by reducing the permeability of blood brain barrier (BBB). In this study, we investigated the role of ISO-1 in cerebral ischemia/reperfusion injury by establishing a model of middle cerebral artery occlusion/reperfusion in rats. Rats were randomly divided into four groups: the sham operation group, the ISO-1group, the cerebral I/R group, and the ISO-1 + I/R group. We assessed the degree of neurological deficit in each group and measured the volume of cerebral infarction. We detected the expression of MIF in the core necrotic area and penumbra. We detected the expression of apoptosis-related proteins, apoptosis-inducing factor (AIF), endonuclease G (EndoG) and cytochrome c oxidase-IV (COX-IV) in the ischemic penumbra region. The results showed that MIF was expressed in the ischemic penumbra, while the injection of ISO-1 was able to alleviate neurological damage and reduce the infarction volume. In the cerebral ischemic penumbra region, ISO-1 could reduce the expression of Bax and Caspase3 and inhibit the displacement of AIF and EndoG to the nucleus simultaneously. Besides, ISO-1 also exhibited the ability to reduce apoptosis. In summary, ISO-1 may inhibit neuronal apoptosis through the endogenous mitochondrial pathway and reduce the injury of brain I/R after ischemic stroke.
Collapse
|
6
|
Svikle Z, Peterfelde B, Sjakste N, Baumane K, Verkauskiene R, Jeng CJ, Sokolovska J. Ubiquitin-proteasome system in diabetic retinopathy. PeerJ 2022; 10:e13715. [PMID: 35873915 PMCID: PMC9306563 DOI: 10.7717/peerj.13715] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 06/21/2022] [Indexed: 01/22/2023] Open
Abstract
Diabetic retinopathy (DR) is the most common complication of diabetes, being the most prevalent reason for blindness among the working-age population in the developed world. Despite constant improvement of understanding of the pathogenesis of DR, identification of novel biomarkers of DR is needed for improvement of patient risk stratification and development of novel prevention and therapeutic approaches. The ubiquitin-proteasome system (UPS) is the primary protein quality control system responsible for recognizing and degrading of damaged proteins. This review aims to summarize literature data on modifications of UPS in diabetes and DR. First, we briefly review the structure and functions of UPS in physiological conditions. We then describe how UPS is involved in the development and progression of diabetes and touch upon the association of UPS genetic factors with diabetes and its complications. Further, we focused on the effect of diabetes-induced hyperglycemia, oxidative stress and hypoxia on UPS functioning, with examples of studies on DR. In other sections, we discussed the association of several other mechanisms of DR (endoplasmic reticulum stress, neurodegeneration etc) with UPS modifications. Finally, UPS-affecting drugs and remedies are reviewed. This review highlights UPS as a promising target for the development of therapies for DR prevention and treatment and identifies gaps in existing knowledge and possible future study directions.
Collapse
Affiliation(s)
- Zane Svikle
- Faculty of Medicine, University of Latvia, Riga, Latvia
| | - Beate Peterfelde
- Faculty of Medicine, University of Latvia, Riga, Latvia,Ophthalmology Department, Riga East University Hospital, Riga, Latvia
| | | | - Kristine Baumane
- Faculty of Medicine, University of Latvia, Riga, Latvia,Ophthalmology Department, Riga East University Hospital, Riga, Latvia
| | - Rasa Verkauskiene
- Institute of Endocrinology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Chi-Juei Jeng
- Ophthalmology Department, Taipei Medical University Shuang Ho Hospital, Ministry of Health and Welfare, Taipei, The Republic of China (Taiwan),College of Medicine, Graduate Institute of Clinical Medicine, National Taiwan University, Taipei, Taiwan
| | | |
Collapse
|
7
|
Macrophage migration inhibitory factor in Nodding syndrome. PLoS Negl Trop Dis 2021; 15:e0009821. [PMID: 34662363 PMCID: PMC8553141 DOI: 10.1371/journal.pntd.0009821] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 10/28/2021] [Accepted: 09/18/2021] [Indexed: 11/19/2022] Open
Abstract
Nodding syndrome (NS) is a catastrophic and enigmatic childhood epilepsy, accompanied by multiple neurological impairments and neuroinflammation. Of all the infectious, environmental and psychological factors associated with NS, the major culprit is Onchocerca Volvulus (Ov)-a parasitic worm transmitted to human by blackflies. NS seems to be an 'Autoimmune Epilepsy' in light of the recent findings of deleterious autoimmune antibodies to Glutamate receptors and to Leiomodin-I in NS patients. Moreover, we recently found immunogenetic fingerprints in HLA peptide-binding grooves associate with protection or susceptibility to NS. Macrophage migration inhibitory factor (MIF) is an immune-regulatory cytokine playing a central role in modulating innate and adaptive immunity. MIF is also involved in various pathologies: infectious, autoimmune and neurodegenerative diseases, epilepsy and others. Herein, two functional polymorphisms in the MIF gene, a -794 CATT5-8 microsatellite repeat and a -173 G/C single-nucleotide polymorphism, were assessed in 49 NS patients and 51 healthy controls from South Sudan. We also measured MIF plasma levels in established NS patients and healthy controls. We discovered that the frequency of the high-expression MIF -173C containing genotype was significantly lower in NS patients compared to healthy controls. Interestingly however, MIF plasma levels were significantly elevated in NS patients than in healthy controls. We further demonstrated that the HLA protective and susceptibility associations are dominant over the MIF association with NS. Our findings suggest that MIF might have a dual role in NS. Genetically controlled high-expression MIF genotype is associated with disease protection. However, elevated MIF in the plasma may contribute to the detrimental autoimmunity, neuroinflammation and epilepsy.
Collapse
|
8
|
Macrophage Migration Inhibitory Factor Alters Functional Properties of CA1 Hippocampal Neurons in Mouse Brain Slices. Int J Mol Sci 2019; 21:ijms21010276. [PMID: 31906137 PMCID: PMC6981710 DOI: 10.3390/ijms21010276] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 12/23/2019] [Accepted: 12/24/2019] [Indexed: 02/06/2023] Open
Abstract
Neuroinflammation is implicated in a host of neurological insults, such as traumatic brain injury (TBI), ischemic stroke, Alzheimer's disease, Parkinson's disease, and epilepsy. The immune response to central nervous system (CNS) injury involves sequelae including the release of numerous cytokines and chemokines. Macrophage migration inhibitory factor (MIF), is one such cytokine that is elevated following CNS injury, and is associated with the prognosis of TBI, and ischemic stroke. MIF has been identified in astrocytes and neurons, and some of the trophic actions of MIF have been related to its direct and indirect actions on astrocytes. However, the potential modulation of CNS neuronal function by MIF has not yet been explored. This study tests the hypothesis that MIF can directly influence hippocampal neuronal function. MIF was microinjected into the hippocampus and the genetically encoded calcium indicator, GCaMP6f, was used to measure Ca2+ events in acute adult mouse brain hippocampal slices. Results demonstrated that a single injection of 200 ng MIF into the hippocampus significantly increased baseline calcium signals in CA1 pyramidal neuron somata, and altered calcium responses to N-methyl-d-aspartate (NMDA) + D-serine in pyramidal cell apical dendrites located in the stratum radiatum. These data are the first to show direct effects of MIF on hippocampal neurons and on NMDA receptor function. Considering that MIF is elevated after brain insults such as TBI, the data suggest that, in addition to the previously described role of MIF in astrocyte reactivity, elevated MIF can have significant effects on neuronal function in the hippocampus.
Collapse
|
9
|
The emerging role of red blood cells in cytokine signalling and modulating immune cells. Blood Rev 2019; 41:100644. [PMID: 31812320 DOI: 10.1016/j.blre.2019.100644] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 11/13/2019] [Accepted: 11/22/2019] [Indexed: 02/07/2023]
Abstract
For many years red blood cells have been described as inert bystanders rather than participants in intercellular signalling, immune function, and inflammatory processes. However, studies are now reporting that red blood cells from healthy individuals regulate immune cell activity and maturation, and red blood cells from disease cohorts are dysfunctional. These cells have now been shown to bind more than 50 cytokines and have been described as a sink for these molecules, and the loss of this activity has been correlated with disease progression. In this review, we summarise what is currently understood about the role of red blood cells in cytokine signalling and in modulating the activity of immune cells. We also discuss the implications of these findings for transfusion medicine and in furthering our understanding of anaemia of chronic inflammation. By bringing these disparate units of work together, we aim to shine a light on an area that requires significantly more investigation.
Collapse
|
10
|
Yang T, Rodriguez V, Malphurs WL, Schmidt JT, Ahmari N, Sumners C, Martyniuk CJ, Zubcevic J. Butyrate regulates inflammatory cytokine expression without affecting oxidative respiration in primary astrocytes from spontaneously hypertensive rats. Physiol Rep 2019; 6:e13732. [PMID: 30039527 PMCID: PMC6056753 DOI: 10.14814/phy2.13732] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 05/07/2018] [Accepted: 05/09/2018] [Indexed: 01/16/2023] Open
Abstract
Neurons and glia exhibit metabolic imbalances in hypertensive animal models, and loss of metabolic homeostasis can lead to neuroinflammation and oxidative stress. The objective of this study was to determine the effects of the microbial metabolite butyrate on mitochondrial bioenergetics and inflammatory markers in mixed brainstem and hypothalamic primary cultures of astrocytes between normotensive (Sprague-Dawley, S-D) and spontaneously hypertensive (SHR) rats. Bioenergetics of mitochondria in astrocytes from normotensive S-D rats were modified with butyrate, but this was not the case in astrocytes derived from SHR, suggesting aberrant mitochondrial function. Transcripts related to oxidative stress, butyrate transporters, butyrate metabolism, and neuroinflammation were quantified in astrocyte cultures treated with butyrate at 0, 200, 600, and 1000 μmol/L. Butyrate decreased catalase and monocarboxylate transporter 1 mRNA in astrocytes of S-D rats but not in the SHR. Moreover, while butyrate did not directly regulate the expression of 3-hydroxybutyrate dehydrogenase 1 and 2 in astrocytes of either strain, the expression levels for these transcripts in untreated cultures were lower in the SHR compared to S-D. We observed higher levels of specific inflammatory cytokines in astrocytes of SHR, and treatment with butyrate decreased expression of Ccl2 and Tlr4 in SHR astrocytes only. Conversely, butyrate treatment increased expression of tumor necrosis factor in astrocytes from SHR but not from the S-D rats. This study improves our understanding of the role of microbial metabolites in regulating astrocyte function, and provides support that butyrate differentially regulates both the bioenergetics and transcripts related to neuroinflammation in astrocytes from SHR versus S-D rats.
Collapse
Affiliation(s)
- Tao Yang
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida
| | - Vermali Rodriguez
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, Florida
| | - Wendi L Malphurs
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida
| | - Jordan T Schmidt
- Department of Physiological Sciences and Center for Environmental and Human Toxicology, University of Florida Genetics Institute, Interdisciplinary Program in Biomedical Sciences Neuroscience, College of Veterinary Medicine, University of Florida, Gainesville, Florida
| | - Niousha Ahmari
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida
| | - Colin Sumners
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, Florida
| | - Christopher J Martyniuk
- Department of Physiological Sciences and Center for Environmental and Human Toxicology, University of Florida Genetics Institute, Interdisciplinary Program in Biomedical Sciences Neuroscience, College of Veterinary Medicine, University of Florida, Gainesville, Florida
| | - Jasenka Zubcevic
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida
| |
Collapse
|
11
|
Vincent FB, Lin E, Sahhar J, Ngian GS, Kandane-Rathnayake R, Mende R, Hoi AY, Morand EF, Lang T, Harris J. Analysis of serum macrophage migration inhibitory factor and D-dopachrome tautomerase in systemic sclerosis. Clin Transl Immunology 2018; 7:e1042. [PMID: 30546906 PMCID: PMC6283235 DOI: 10.1002/cti2.1042] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 11/21/2018] [Accepted: 11/21/2018] [Indexed: 12/25/2022] Open
Abstract
Objectives Macrophage migration inhibitory factor (MIF) and D‐dopachrome tautomerase (DDT), members of the same cytokine superfamily, are linked to the pathogenesis of a number of inflammatory diseases. The aim of this study was to investigate their clinical relevance in systemic sclerosis (SSc). Methods Serum MIF and DDT were quantified in 105 SSc patients by ELISA and levels compared to healthy controls (HC) (47) and patients with systemic lupus erythematosus (SLE) (184). Clinical parameters included organ involvement, serum laboratory markers and results of pulmonary function tests, and overall disease activity assessed using the European Scleroderma Trials and Research group (EUSTAR) activity index. Results There was no significant difference in serum DDT concentrations between patients with SSc and HC. However, serum MIF was significantly increased in SSc compared to both HC and SLE cohorts. Serum MIF was increased in SSc patients with low forced vital capacity (FVC) and was also associated with the use of angiotensin II receptor blockers and beta blockers in SSc, confirmed after adjusting for the presence of systemic hypertension and low FVC. Serum DDT was significantly higher in SSc patients with low FEV1 and negatively correlated with EUSTAR score, particularly in patients with limited disease. Conclusion Although not significantly linked to specific clinical parameters, serum MIF was significantly higher in SSc patients than in HC and SLE patients, suggesting a fundamental role for MIF in SSc. DDT, while closely related to MIF, did not show a similar expression profile, suggesting functional differences between these molecules.
Collapse
Affiliation(s)
- Fabien B Vincent
- Centre for Inflammatory Diseases School of Clinical Sciences at Monash Health Monash University Clayton VIC Australia
| | - Emily Lin
- Centre for Inflammatory Diseases School of Clinical Sciences at Monash Health Monash University Clayton VIC Australia
| | - Joanne Sahhar
- Centre for Inflammatory Diseases School of Clinical Sciences at Monash Health Monash University Clayton VIC Australia.,Department of Rheumatology Monash Health Clayton VIC Australia
| | - Gene-Siew Ngian
- Centre for Inflammatory Diseases School of Clinical Sciences at Monash Health Monash University Clayton VIC Australia.,Department of Rheumatology Monash Health Clayton VIC Australia
| | - Rangi Kandane-Rathnayake
- Centre for Inflammatory Diseases School of Clinical Sciences at Monash Health Monash University Clayton VIC Australia
| | - Rachel Mende
- Centre for Inflammatory Diseases School of Clinical Sciences at Monash Health Monash University Clayton VIC Australia
| | - Alberta Y Hoi
- Centre for Inflammatory Diseases School of Clinical Sciences at Monash Health Monash University Clayton VIC Australia
| | - Eric F Morand
- Centre for Inflammatory Diseases School of Clinical Sciences at Monash Health Monash University Clayton VIC Australia.,Department of Rheumatology Monash Health Clayton VIC Australia
| | - Tali Lang
- Centre for Inflammatory Diseases School of Clinical Sciences at Monash Health Monash University Clayton VIC Australia.,Present address: The Szalmuk Family Department of Medical Oncology Cabrini Institute Malvern VIC 3144 Australia
| | - James Harris
- Centre for Inflammatory Diseases School of Clinical Sciences at Monash Health Monash University Clayton VIC Australia
| |
Collapse
|
12
|
Haspula D, Clark MA. Neuroinflammation and sympathetic overactivity: Mechanisms and implications in hypertension. Auton Neurosci 2018; 210:10-17. [DOI: 10.1016/j.autneu.2018.01.002] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 01/02/2018] [Accepted: 01/08/2018] [Indexed: 02/07/2023]
|
13
|
Barbosa RM, Speretta GF, Dias DPM, Ruchaya PJ, Li H, Menani JV, Sumners C, Colombari E, Colombari DSA. Increased Expression of Macrophage Migration Inhibitory Factor in the Nucleus of the Solitary Tract Attenuates Renovascular Hypertension in Rats. Am J Hypertens 2017; 30:435-443. [PMID: 28158469 PMCID: PMC5861587 DOI: 10.1093/ajh/hpx001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 10/20/2016] [Accepted: 01/02/2017] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Macrophage migration inhibitory factor (MIF) is an intracellular inhibitory regulator of the actions of angiotensin II in the central nervous system. Renovascular hypertensive 2-kidney, 1-clip (2K1C) rats have an increased activity of the renin-angiotensin system and a decrease in baroreflex function compared to normotensive (NT) rats. In the present study, we tested the effects of MIF overexpression within the nucleus of the solitary tract (NTS), a key brainstem region for cardiovascular regulation, on the development of hypertension, on baroreflex function, and on water and food intake in 2K1C rats. METHODS Holtzman NT rats received a silver clip around the left renal artery to induce 2K1C hypertension. Three weeks later, rats were microinjected in the NTS with AAV2-CBA-MIF, to increase the expression of MIF, or with the control vector AAV2-CBA-enhanced green fluorescent protein. Mean arterial pressure (MAP) and heart rate were recorded by telemetry. Baroreflex function was tested, and water and food intake were also measured. RESULTS Increasing MIF expression in the NTS of 2K1C rats attenuated the development of hypertension, reversed the impairment of baroreflex function, and reduced the increase in water intake. In contrast to 2K1C rats, similar increases in MIF expression in the NTS of NT rats produced no changes in baseline MAP, baroreflex function, or water intake. CONCLUSIONS These results indicate that an increased expression of MIF within the NTS attenuates the development of hypertension and restores the baroreflex function in 2K1C rats.
Collapse
Affiliation(s)
- Rafaela Moreira Barbosa
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University, Araraquara, São Paulo, Brazil
| | - Guilherme F Speretta
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University, Araraquara, São Paulo, Brazil
| | - Daniel Penteado Martins Dias
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Prashant Jay Ruchaya
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University, Araraquara, São Paulo, Brazil
| | - Hongwei Li
- School of Biotechnology, Southern Medical University, Guangzhou, China
| | - José Vanderlei Menani
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University, Araraquara, São Paulo, Brazil
| | - Colin Sumners
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Eduardo Colombari
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University, Araraquara, São Paulo, Brazil
| | - Débora S A Colombari
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University, Araraquara, São Paulo, Brazil
| |
Collapse
|
14
|
Bloom J, Sun S, Al-Abed Y. MIF, a controversial cytokine: a review of structural features, challenges, and opportunities for drug development. Expert Opin Ther Targets 2016; 20:1463-1475. [PMID: 27762152 DOI: 10.1080/14728222.2016.1251582] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
INTRODUCTION Macrophage migration inhibitory factor (MIF) has emerged as a promising drug target in diseases including sepsis, rheumatoid arthritis, and cancer. MIF has multiple properties that favor development of specific, targeted therapies: it is expressed broadly among human cells, has noted roles in diverse inflammatory and oncological processes, and has intrinsic enzymatic activity amenable to high-throughput screening. Despite these advantages, anti-MIF therapy remains well behind other cytokine-targeted therapeutics, with no small molecules in the pipeline for clinical development and anti-MIF antibodies only recently beginning clinical trials. Areas covered: In this review we summarize current literature regarding MIF structure and function-including challenges and controversies that have arisen in studies of anti-MIF therapeutics-and propose a strategy for development of clinically relevant anti-MIF drugs. Expert opinion: We believe that the field of anti-MIF therapeutics would benefit from capitalizing on the protein's multiple assets while acknowledging their flaws. The tautomerase enzymatic site of MIF may not be active biologically, but can nonetheless offer a high-throughput method to highlight molecules of interest that can affect its other, frequently intertwined bioactivities. Future work should also focus on developing more robust assays for MIF bioactivity that can be used for second-pass screening and specificity studies.
Collapse
Affiliation(s)
- Joshua Bloom
- a Center for Molecular Innovation , The Feinstein Institute for Medical Research , Manhasset , NY , USA
| | - Shan Sun
- a Center for Molecular Innovation , The Feinstein Institute for Medical Research , Manhasset , NY , USA
| | - Yousef Al-Abed
- a Center for Molecular Innovation , The Feinstein Institute for Medical Research , Manhasset , NY , USA
| |
Collapse
|
15
|
Erdos B, Clifton RR, Liu M, Li H, McCowan ML, Sumners C, Scheuer DA. Novel mechanism within the paraventricular nucleus reduces both blood pressure and hypothalamic pituitary-adrenal axis responses to acute stress. Am J Physiol Heart Circ Physiol 2015; 309:H634-45. [PMID: 26071542 DOI: 10.1152/ajpheart.00207.2015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 06/05/2015] [Indexed: 02/07/2023]
Abstract
Macrophage migration inhibitory factor (MIF) counteracts pressor effects of angiotensin II (ANG II) in the paraventricular nucleus of the hypothalamus (PVN) in normotensive rats, but this mechanism is absent in spontaneously hypertensive rats (SHRs) due to a lack of MIF in PVN neurons. Since endogenous ANG II in the PVN modulates stress reactivity, we tested the hypothesis that replacement of MIF in PVN neurons would reduce baseline blood pressure and inhibit stress-induced increases in blood pressure and plasma corticosterone in adult male SHRs. Radiotelemetry transmitters were implanted to measure blood pressure, and then an adeno-associated viral vector expressing either enhanced green fluorescent protein (GFP) or MIF was injected bilaterally into the PVN. Cardiovascular responses to a 15-min water stress (1-cm deep, 25°C) and a 60-min restraint stress were evaluated 3-4 wk later. MIF treatment in the PVN attenuated average restraint-induced increases in blood pressure (37.4 ± 2.0 and 27.6 ± 3.5 mmHg in GFP and MIF groups, respectively, P < 0.05) and corticosterone (42 ± 2 and 36 ± 3 μg/dl in GFP and MIF groups, respectively, P < 0.05). MIF treatment in the PVN also reduced stress-induced elevations in the number of c-Fos-positive cells in the rostral ventrolateral medulla (71 ± 5 in GFP and 47 ± 5 in MIF SHRs, P < 0.01) and corticotropin-releasing factor mRNA expression in the PVN. However, MIF had no significant effects on the cardiovascular responses to water stress in SHRs or to either stress in Sprague-Dawley rats. Therefore, viral vector-mediated restoration of MIF in PVN neurons of SHRs attenuates blood pressure and hypothalamic pituitary adrenal axis responses to stress.
Collapse
Affiliation(s)
- Benedek Erdos
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida
| | - Rebekah R Clifton
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida
| | - Meng Liu
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida
| | - Hongwei Li
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida
| | - Michael L McCowan
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida
| | - Colin Sumners
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida
| | - Deborah A Scheuer
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida
| |
Collapse
|
16
|
Xu X, Hua Y, Nair S, Bucala R, Ren J. Macrophage migration inhibitory factor deletion exacerbates pressure overload-induced cardiac hypertrophy through mitigating autophagy. Hypertension 2013; 63:490-9. [PMID: 24366076 DOI: 10.1161/hypertensionaha.113.02219] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The proinflammatory cytokine macrophage migration inhibitory factor (MIF) has been shown to be cardioprotective under various pathological conditions. However, the underlying mechanisms still remain elusive. In this study, we revealed that MIF deficiency overtly exacerbated abdominal aorta constriction-induced cardiac hypertrophy and contractile anomalies. MIF deficiency interrupted myocardial autophagy in hypertrophied hearts. Rapamycin administration mitigated the exacerbated hypertrophic responses in MIF(-/-) mice. Using the phenylephrine-induced hypertrophy in vitro model in H9C2 myoblasts, we confirmed that MIF governed the activation of AMP-activated protein kinase-mammalian target of rapamycin-autophagy cascade. Confocal microscopic examination demonstrated that MIF depletion prevented phenylephrine-induced mitophagy in H9C2 myoblasts. Myocardial Parkin, an E3 ubiquitin ligase and a marker for mitophagy, was significantly upregulated after sustained pressure overload, the effect of which was prevented by MIF knockout. Furthermore, our data exhibited that levels of MIF, AMP-activated protein kinase activation, and autophagy were elevated concurrently in human failing hearts. These data indicate that endogenous MIF regulates the mammalian target of rapamycin signaling to activate autophagy to preserve cardiac geometry and protect against hypertrophic responses.
Collapse
Affiliation(s)
- Xihui Xu
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, 1000 E University Ave, Laramie, WY 82071.
| | | | | | | | | |
Collapse
|
17
|
Neuroimmune communication in hypertension and obesity: a new therapeutic angle? Pharmacol Ther 2013; 138:428-40. [PMID: 23458610 DOI: 10.1016/j.pharmthera.2013.02.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Accepted: 02/12/2013] [Indexed: 12/13/2022]
Abstract
Hypertension is an epidemic health concern and a major risk factor for the development of cardiovascular disease. Although there are available treatment strategies for hypertension, numerous hypertensive patients do not have their clinical symptoms under control and it is imperative that new avenues to treat or prevent high blood pressure in these patients are developed. It is well established that increases in sympathetic nervous system (SNS) outflow and enhanced renin-angiotensin system (RAS) activity are common features of hypertension and various pathological conditions that predispose individuals to hypertension. More recently, hypertension has also become recognized as an immune condition and accumulating evidence suggests that interactions between the RAS, SNS and immune systems play a role in blood pressure regulation. This review summarizes what is known about the interconnections between the RAS, SNS and immune systems in the neural regulation of blood pressure. Based on the reviewed studies, a model for RAS/neuroimmune interactions during hypertension is proposed and the therapeutic potential of targeting RAS/neuroimmune interactions in hypertensive patients is discussed. Special emphasis is placed on the applicability of the proposed model to obesity-related hypertension.
Collapse
|
18
|
Jiang N, Shi P, Desland F, Kitchen-Pareja MC, Sumners C. Interleukin-10 inhibits angiotensin II-induced decrease in neuronal potassium current. Am J Physiol Cell Physiol 2013; 304:C801-7. [PMID: 23426971 DOI: 10.1152/ajpcell.00398.2012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Previously we demonstrated that viral-mediated increased expression of the anti-inflammatory cytokine interleukin-10 within the paraventricular nucleus of the hypothalamus significantly reduces blood pressure in normal rats made hypertensive by infusion of angiotensin II. However, the exact cellular locus of this interleukin-10 action within the paraventricular nucleus is unknown. In the present study we tested whether interleukin-10 exerts direct effects at its receptors located on hypothalamic neurons to offset the neuronal excitatory actions of angiotensin II via its type 1 receptors. The results indicated the presence of immunoreactive interleukin-10 receptors on neurons in normal rat paraventricular nucleus and that receptors for this cytokine were also expressed in neurons cultured from the hypothalamus. Patch-clamp electrophysiological recordings from these cultures revealed that extracellular application of interleukin-10 alone did not exert any alterations in neuronal membrane delayed rectifier or transient potassium currents. However, angiotensin II elicited a significant decrease in delayed rectifier potassium current, an effect that was abolished by interleukin-10 application. Since decreases in delayed rectifier potassium current contribute to increased neuronal excitability, this result is consistent with a direct inhibitory action of interleukin-10 on angiotensin-induced excitation of hypothalamic neurons. As such, these data are the first indication of a neuronal locus of action of interleukin-10 to temper the actions of angiotensin II in the hypothalamus.
Collapse
Affiliation(s)
- Nan Jiang
- Department of Physiology and Functional Genomics, McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | | | | | | | | |
Collapse
|
19
|
Zhang Y, Gao Y, Speth RC, Jiang N, Mao Y, Sumners C, Li H. Adenoviral and adeno-associated viral vectors-mediated neuronal gene transfer to cardiovascular control regions of the rat brain. Int J Med Sci 2013; 10:607-16. [PMID: 23569423 PMCID: PMC3619099 DOI: 10.7150/ijms.5700] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Accepted: 03/11/2013] [Indexed: 02/03/2023] Open
Abstract
Viral vectors have been utilized extensively to introduce genetic material into the central nervous system. In order to investigate gene functions in cardiovascular control regions of rat brain, we applied WPRE (woodchuck hepatitis virus post-transcriptional regulatory element) enhanced-adenoviral (Ad) and adeno-assoicated virus (AAV) type 2 vectors to mediate neuronal gene delivery to the paraventricular nucleus of the hypothalamus, the nucleus tractus solitarius and the rostral ventrolateral medulla, three important cardiovascular control regions known to express renin-angiotensin system (RAS) genes. Ad or AAV2 harboring an enhanced green fluorescent protein (EGFP) reporter gene or the angiotensin type 2 receptor gene were microinjected into these brain regions in adult rats. Our results demonstrated that both AAV2 and Ad vectors elicited long-term neuronal transduction in these regions. Interestingly, we found that the WPRE caused expression of GFP driven by the synapsin1 promoter in pure glial cultures or co-cultures of neurons and glia derived from rat hypothalamus and brainstem. However, in rat paraventricular nucleus WPRE did not cause expression of GFP in glia. This demonstrates the potential use of these vectors in studies of physiological functions of certain genes in the cardiovascular control regions of the brain.
Collapse
Affiliation(s)
- Yanling Zhang
- School of Biotechnology, Southern Medical University, Guangzhou, China
| | | | | | | | | | | | | |
Collapse
|
20
|
Freiria-Oliveira AH, Blanch GT, Li H, Colombari E, Colombari DSA, Sumners C. Macrophage migration inhibitory factor in the nucleus of solitary tract decreases blood pressure in SHRs. Cardiovasc Res 2012; 97:153-60. [PMID: 22997157 DOI: 10.1093/cvr/cvs297] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
AIMS The macrophage migration inhibitory factor (MIF) is an intracellular inhibitor of the central nervous system actions of angiotensin II on blood pressure. Considering that angiotensin II actions at the nucleus of the solitary tract are important for the maintenance of hypertension in spontaneously hypertensive rats (SHRs), we tested if increased MIF expression in the nucleus of the solitary tract of SHR alters the baseline high blood pressure in these rats. METHODS AND RESULTS Eight-week-old SHRs or normotensive rats were microinjected with the vector AAV2-CBA-MIF into the nucleus of the solitary tract, resulting in MIF expression predominantly in neurons. Rats also underwent recordings of the mean arterial blood pressure (MAP) and heart rate (via telemetry devices implanted in the abdominal aorta), cardiac- and baroreflex function. Injections of AAV2-CBA-MIF into the nucleus of the solitary tract of SHRs produced significant decreases in the MAP, ranging from 10 to 20 mmHg, compared with age-matched SHRs that had received identical microinjections of the control vector AAV2-CBA-eGFP. This lowered MAP in SHRs was maintained through the end of the experiment at 31 days, and was associated with an improvement in baroreflex function to values observed in normotensive rats. In contrast to SHRs, similar increased MIF expression in the nucleus of the solitary tract of normotensive rats produced no changes in baseline MAP and baroreflex function. CONCLUSION These results indicate that an increased expression of MIF within the nucleus of the solitary tract neurons of SHRs lowers blood pressure and restores baroreflex function.
Collapse
Affiliation(s)
- André Henrique Freiria-Oliveira
- Department of Physiology and Functional Genomics and McKnight Brain Institute, College of Medicine, University of Florida, 1600 Southwest Archer Road, PO-BOX: 100274, Gainesville, FL 32610, USA
| | | | | | | | | | | |
Collapse
|
21
|
Honigman JS, DiGregorio KM, Dedkov EI, Leheste JR, Leng L, Bucala R, Torres G. Distribution maps of D-dopachrome tautomerase in the mouse brain. Neuroscience 2012; 226:382-7. [PMID: 23000624 DOI: 10.1016/j.neuroscience.2012.09.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2012] [Revised: 09/04/2012] [Accepted: 09/05/2012] [Indexed: 11/24/2022]
Abstract
D-Dopachrome tautomerase is an enzyme related by amino acid sequence and catalytic activity to macrophage migration inhibitory factor. Both of these small molecules are pro-inflammatory cytokines mediating broad innate immune responses. Although it is well established that the gene product of D-dopachrome tautomerase is widely expressed in liver and kidney cells, no study has mapped the distribution pattern of this tautomeric enzyme in the mammalian nervous system. Here, we address this void by characterizing the cellular localization of D-dopachrome tautomerase in the adult mouse brain. Two well-characterized polyclonal antibodies were used for Western blotting and immunohistochemical localization of the endogenous tautomeric enzyme. Our results show that D-dopachrome tautomerase is present throughout the brain parenchyma with a large fraction of heterogeneous interneurons harboring a stable and robust expression of the enzyme. These data point to a potential involvement of D-dopachrome tautomerase activity in the mature mouse brain, and suggest some functional and evolutionary relationship between innate immunity and tautomerization of D-dopachrome in mammalian species.
Collapse
Affiliation(s)
- J S Honigman
- Department of Neuroscience and Histology, New York College of Osteopathic Medicine of New York Institute of Technology, Old Westbury, NY 11568, USA
| | | | | | | | | | | | | |
Collapse
|
22
|
Brain miffed by macrophage migration inhibitory factor. Int J Cell Biol 2012; 2012:139573. [PMID: 22973314 PMCID: PMC3438795 DOI: 10.1155/2012/139573] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Revised: 07/06/2012] [Accepted: 07/12/2012] [Indexed: 12/31/2022] Open
Abstract
Macrophage migration inhibitory factor (MIF) is a cytokine which also exhibits enzymatic properties like oxidoreductase and tautomerase. MIF plays a pivotal role in innate and acquired immunity as well as in the neuroendocrine axis. Since it is involved in the pathogenesis of acute and chronic inflammation, neoangiogenesis, and cancer, MIF and its signaling components are considered suitable targets for therapeutic intervention in several fields of medicine. In neurodegenerative and neurooncological diseases, MIF is a highly relevant, but still a hardly investigated mediator. MIF operates via intracellular protein-protein interaction as well as in CD74/CXCR2/CXCR4 receptor-mediated pathways to regulate essential cellular systems such as redox balance, HIF-1, and p53-mediated senescence and apoptosis as well as multiple signaling pathways. Acting as an endogenous glucocorticoid antagonist, MIF thus represents a relevant resistance gene in brain tumor therapies. Alongside this dual action, a functional homolog-annotated D-dopachrome tautomerase/MIF-2 has been uncovered utilizing the same cell surface receptor signaling cascade as MIF. Here we review MIF actions with respect to redox regulation in apoptosis and in tumor growth as well as its extracellular function with a focus on its potential role in brain diseases. We consider the possibility of MIF targeting in neurodegenerative processes and brain tumors by novel MIF-neutralizing approaches.
Collapse
|
23
|
Renin-Angiotensin system hyperactivation can induce inflammation and retinal neural dysfunction. Int J Inflam 2012; 2012:581695. [PMID: 22536545 PMCID: PMC3321303 DOI: 10.1155/2012/581695] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2011] [Revised: 12/09/2011] [Accepted: 01/04/2012] [Indexed: 12/21/2022] Open
Abstract
The renin-angiotensin system (RAS) is a hormone system that has been classically known as a blood pressure regulator but is becoming well recognized as a proinflammatory mediator. In many diverse tissues, RAS pathway elements are also produced intrinsically, making it possible for tissues to respond more dynamically to systemic or local cues. While RAS is important for controlling normal inflammatory responses, hyperactivation of the pathway can cause neural dysfunction by inducing accelerated degradation of some neuronal proteins such as synaptophysin and by activating pathological glial responses. Chronic inflammation and oxidative stress are risk factors for high incidence vision-threatening diseases such as diabetic retinopathy (DR), age-related macular degeneration (AMD), and glaucoma. In fact, increasing evidence suggests that RAS inhibition may actually prevent progression of various ocular diseases including uveitis, DR, AMD, and glaucoma. Therefore, RAS inhibition may be a promising therapeutic approach to fine-tune inflammatory responses and to prevent or treat certain ocular and neurodegenerative diseases.
Collapse
|
24
|
Cardinale JP, Sriramula S, Mariappan N, Agarwal D, Francis J. Angiotensin II-induced hypertension is modulated by nuclear factor-κBin the paraventricular nucleus. Hypertension 2011; 59:113-21. [PMID: 22106405 DOI: 10.1161/hypertensionaha.111.182154] [Citation(s) in RCA: 133] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Hypertension is considered a low-grade inflammatory condition, and understanding the role of transcription factors in guiding this response is pertinent. A prominent transcription factor that governs inflammatory responses and has become a focal point in hypertensive research is nuclear factor-κB (NFκB). Within the hypothalamic paraventricular nucleus (PVN), a known brain cardioregulatory center, NFκB becomes potentially even more important in ultimately coordinating the systemic hypertensive response. To definitively demonstrate the role of NFκB in the neurogenic hypertensive response, we hypothesized that PVN NFκB blockade would attenuate angiotensin II-induced hypertension. Twelve-week-old male Sprague-Dawley rats were implanted with radiotelemetry probes for blood pressure measurement and allowed a 7-day recovery. After baseline blood pressure recordings, rats were administered either continuous NFκB decoy oligodeoxynucleotide infusion or microinjection of a serine mutated adenoviral inhibitory-κB vector, or their respective controls, bilaterally into the PVN to inhibit NFκB at two levels of its activation pathway. Simultaneously, rats were implanted subcutaneously with an angiotensin II or saline-filled 14-day osmotic minipump. After the 2-week treatments, rats were euthanized and brain tissues collected for PVN analysis. Bilaterally inhibited NFκB rats had a decrease in blood pressure, NFκB p65 subunit activity, proinflammatory cytokines, and reactive oxygen species, including the angiotensin II type 1 receptor, angiotensin-converting enzyme, tumor necrosis factor, and superoxide in angiotensin II-treated rats. Moreover, after NFκB blockade, key protective antihypertensive renin-angiotensin system components were upregulated. This demonstrates the important role that transcription factor NFκB plays within the PVN in modulating and perpetuating the hypertensive response via renin-angiotensin system modulation.
Collapse
Affiliation(s)
- Jeffrey P Cardinale
- Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | | | | | | | | |
Collapse
|
25
|
Sriramula S, Cardinale JP, Lazartigues E, Francis J. ACE2 overexpression in the paraventricular nucleus attenuates angiotensin II-induced hypertension. Cardiovasc Res 2011; 92:401-8. [PMID: 21952934 DOI: 10.1093/cvr/cvr242] [Citation(s) in RCA: 148] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
AIMS Angiotensin II (Ang II) has been shown to have both central and peripheral effects in mediating hypertension, for which the hypothalamic paraventricular nucleus (PVN) is an important brain cardio-regulatory centre. Angiotensin-converting enzyme 2 (ACE2) has been identified as a negative regulator of the pro-hypertensive actions of Ang II. Recent findings from our laboratory suggest that Ang II infusion decreases ACE2 expression in the PVN. In the present study, we hypothesized that ACE2 overexpression in the PVN will have beneficial effects in counteracting Ang II-induced hypertension. METHODS AND RESULTS Male Sprague-Dawley rats were used in this study. Bilateral microinjection of an adenovirus encoding hACE2 (Ad-ACE2) into the PVN was used to overexpress ACE2 within this region. Mean arterial pressure measured by radiotelemetry was significantly increased after 14 days in Ang II-infused (200 ng/kg/min) rats vs. saline-infused controls (162.9 ± 3.6 vs. 102.3 ± 1.5 mmHg). Bilateral PVN microinjection of Ad-ACE2 attenuated this Ang II-induced hypertension (130.2 ± 5.7 vs. 162.9 ± 3.6 mmHg). ACE2 overexpression also significantly decreased AT(1)R and ACE expression and increased AT(2)R and Mas expression in the PVN. Additionally, ACE2 overexpression in the PVN attenuated the Ang II-induced increase in the expression of the pro-inflammatory cytokines tumour necrosis factor-α, interleukin (IL)-1β and IL-6 in the PVN. CONCLUSION Our findings suggest that attenuation of pro-inflammatory cytokines in the PVN in combination with the shift of the renin-angiotensin system towards the anti-hypertensive axis (ACE2/Ang-(1-7)/Mas) may be responsible for the overall beneficial effects of ACE2 overexpression in the PVN on the Ang II-induced hypertensive response.
Collapse
Affiliation(s)
- Srinivas Sriramula
- Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, 1909 Skip Bertman Drive, Baton Rouge, LA 70803, USA
| | | | | | | |
Collapse
|
26
|
Inácio AR, Bucala R, Deierborg T. Lack of macrophage migration inhibitory factor in mice does not affect hallmarks of the inflammatory/immune response during the first week after stroke. J Neuroinflammation 2011; 8:75. [PMID: 21714902 PMCID: PMC3152909 DOI: 10.1186/1742-2094-8-75] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2011] [Accepted: 06/29/2011] [Indexed: 11/10/2022] Open
Abstract
Background Macrophage migration inhibitory factor (MIF) has been proposed to play a detrimental role in stroke. We recently showed that MIF promotes neuronal death and aggravates neurological deficits during the first week after experimental stroke, in mice. Since MIF regulates tissue inflammation, we studied the putative role of MIF in post-stroke inflammation. Methods We subjected C57BL/6 mice, Mif-/- (MIF-KO) or Mif+/+ (WT), to a transient occlusion of the right middle cerebral artery (tMCAo) or sham-surgery. We studied MIF expression, GFAP expression and the number of CD74-positive cells in the ischemic brain hemisphere 7 days after tMCAo using primarily immunohistochemistry. We determined IFN-γ, IL-2, IL-4, IL-5, IL-10, IL-12, KC/CXCL-1 and TNF-α protein levels in the brain (48 h after surgery) and serum (48 h and 7 days after surgery) by a multiplex immunoassay. Results We observed that MIF accumulates in neurons and astrocytes of the peri-infarct region, as well as in microglia/macrophages of the infarct core up to 7 days after stroke. Among the inflammatory mediators analyzed, we found a significant increase in cerebral IL-12 and KC levels after tMCAo, in comparison to sham-surgery. Importantly, the deletion of Mif did not significantly affect the levels of the cytokines evaluated, in the brain or serum. Moreover, the spleen weight 48 h and 7 days subsequent to tMCAo was similar in WT and MIF-KO mice. Finally, the extent of GFAP immunoreactivity and the number of MIF receptor (CD74)-positive cells within the ischemic brain hemisphere did not differ significantly between WT and MIF-KO mice subjected to tMCAo. Conclusions We conclude that MIF does not affect major components of the inflammatory/immune response during the first week after experimental stroke. Based on present and previous evidence, we propose that the deleterious MIF-mediated effects in stroke depend primarily on an intraneuronal and/or interneuronal action.
Collapse
Affiliation(s)
- Ana R Inácio
- Laboratory for Experimental Brain Research, Department of Clinical Sciences, Lund University, BMC A13, 22184 Lund, Sweden.
| | | | | |
Collapse
|
27
|
Zubcevic J, Waki H, Raizada MK, Paton JFR. Autonomic-immune-vascular interaction: an emerging concept for neurogenic hypertension. Hypertension 2011; 57:1026-33. [PMID: 21536990 DOI: 10.1161/hypertensionaha.111.169748] [Citation(s) in RCA: 143] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Jasenka Zubcevic
- Department of Physiology and Functional Genomics, McKnight Brain Institute, 1600 SW Archer Rd, Gainesville, FL 32610, USA
| | | | | | | |
Collapse
|
28
|
Koga K, Kenessey A, Powell SR, Sison CP, Miller EJ, Ojamaa K. Macrophage migration inhibitory factor provides cardioprotection during ischemia/reperfusion by reducing oxidative stress. Antioxid Redox Signal 2011; 14:1191-202. [PMID: 20831446 DOI: 10.1089/ars.2010.3163] [Citation(s) in RCA: 97] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Macrophage migration inhibitory factor (MIF) is a multifunctional protein that exhibits an intrinsic thiol protein oxidoreductase activity and proinflammatory activities. In the present study to examine intracellular MIF redox function, exposure of MIF-deficient cardiac fibroblasts to oxidizing conditions resulted in a 2.3-fold increase (p < 0.001) in intracellular ROS that could be significantly reduced by adenoviral-mediated reexpression of recombinant MIF. In an animal model of myocardial injury by ischemia/reperfusion (I/R), MIF-deficient hearts exhibited higher levels of oxidative stress than did wild-type hearts, as measured by significantly higher oxidized glutathione levels (decreased GSH/GSSG ratio), increased protein oxidation, reduced aconitase activity, and increased mitochondrial injury (increased cytochrome c release). The increased myocardial oxidative stress after I/R was reflected by larger infarct size (INF) in MIF-deficient hearts versus wild-type (WT) hearts (21 ± 6% vs. 8 ± 3% INF/LV; p < 0.05). In vivo hemodynamic measurements showed that left ventricular (LV) contractile function of MIF-deficient hearts subjected to 15-min ischemia failed to recover during reperfusion compared with WT hearts (LV developed pressure and ± dP/dt; p = 0.02). These data represent the first in vivo evidence in support of a cardioprotective role of MIF in the postischemic heart by reducing oxidative stress.
Collapse
Affiliation(s)
- Kiyokazu Koga
- Elmezzi Graduate School of Molecular Medicine, The Feinstein Institute for Medical Research, North Shore-LIJ Health System, Manhasset, New York, USA
| | | | | | | | | | | |
Collapse
|
29
|
Macrophage migration inhibitory factor promotes cell death and aggravates neurologic deficits after experimental stroke. J Cereb Blood Flow Metab 2011; 31:1093-106. [PMID: 21063426 PMCID: PMC3070968 DOI: 10.1038/jcbfm.2010.194] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Multiple mechanisms contribute to tissue demise and functional recovery after stroke. We studied the involvement of macrophage migration inhibitory factor (MIF) in cell death and development of neurologic deficits after experimental stroke. Macrophage migration inhibitory factor is upregulated in the brain after cerebral ischemia, and disruption of the Mif gene in mice leads to a smaller infarct volume and better sensory-motor function after transient middle cerebral artery occlusion (tMCAo). In mice subjected to tMCAo, we found that MIF accumulates in neurons of the peri-infarct region, particularly in cortical parvalbumin-positive interneurons. Likewise, in cultured cortical neurons exposed to oxygen and glucose deprivation, MIF levels increase, and inhibition of MIF by (S,R)-3-(4-hydroxyphenyl)-4,5-dihydro-5-isoxazole acetic acid methyl ester (ISO-1) protects against cell death. Deletion of MIF in Mif(-/-) mice does not affect interleukin-1β protein levels in the brain and serum after tMCAo. Furthermore, disruption of the Mif gene in mice does not affect CD68, but it is associated with higher galectin-3 immunoreactivity in the brain after tMCAo, suggesting that MIF affects the molecular/cellular composition of the macrophages/microglia response after experimental stroke. We conclude that MIF promotes neuronal death and aggravates neurologic deficits after experimental stroke, which implicates MIF in the pathogenesis of neuronal injury after stroke.
Collapse
|
30
|
Regulation of posttranscriptional modification as a possible therapeutic approach for retinal neuroprotection. J Ophthalmol 2010; 2011:506137. [PMID: 21076532 PMCID: PMC2975078 DOI: 10.1155/2011/506137] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2010] [Revised: 08/24/2010] [Accepted: 09/16/2010] [Indexed: 12/30/2022] Open
Abstract
Understanding pathogenesis at the molecular level is the first step toward developing new therapeutic approaches. Here, we review the molecular mechanisms of visual dysfunction in two common diseases, innate chorioretinal inflammation and diabetic retinopathy, and the role of the ubiquitin-proteasome system (UPS) in both processes. In innate chorioretinal inflammation, interleukin-6 family ligands induce STAT3 activation in photoreceptors, which causes UPS-mediated excessive degradation of the visual substance, rhodopsin. In diabetic retinopathy, angiotensin II type 1 receptor (AT1R) signaling activates ERK in the inner layers of the retina, causing UPS-mediated excessive degradation of the synaptic vesicle protein, synaptophysin. This latter effect may decrease synaptic activity, in turn adversely affecting neuronal survival. Both mechanisms involve increased UPS activity and the subsequent excessive degradation of a protein required for visual function. Finally, we review the therapeutic potential of regulating the UPS to protect tissue function, citing examples from clinical applications in other medical fields.
Collapse
|
31
|
Yao F, Modgil A, Zhang Q, Pingili A, Singh N, O'Rourke ST, Sun C. Pressor effect of apelin-13 in the rostral ventrolateral medulla: role of NAD(P)H oxidase-derived superoxide. J Pharmacol Exp Ther 2010; 336:372-80. [PMID: 21047952 DOI: 10.1124/jpet.110.174102] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Microinjection of apelin-13 into the rostral ventrolateral medulla (RVLM) in the brainstem increases blood pressure in rats. In the present study, we tested the hypotheses that apelin-13 directly stimulates neuronal activity in neurons cultured from the brainstem and that NAD(P)H oxidase-derived reactive oxygen species are involved in this action of apelin-13. Microinjection of apelin-13 into the RVLM resulted in increases in arterial pressure and in renal sympathetic nerve activity in Sprague-Dawley rats. The pressor effect of apelin-13 was attenuated by the specific NAD(P)H-oxidase inhibitor gp91ds-tat. In neurons cultured from the ventral brainstem, spontaneous action potentials were recorded using current-clamp recording. Superfusion of neurons with apelin-13 (100 nM) increased the neuronal firing rate from 0.79 ± 0.14 to 1.45 ± 0.26 Hz (n = 7, P < 0.01) in angiotensin II receptor-like 1-positive neurons, identified with single-cell reverse transcriptase-polymerase chain reaction. Neither the angiotensin II type 1 receptor antagonist losartan nor the angiotensin II type 2 receptor antagonist 1-[[4-(dimethylamino)-3-methylphenyl[methyl]-5-(diphenylacetyl)-4,5,6,7-tetrahydro-1H-imidazo[4,5-c]pyridine-6-carboxylic acid ditrifluoroacetate (PD123319) altered the positive chronotropic effect of apelin-13. Pretreatment of cells with either the reactive oxygen species scavenger superoxide dismutase [polyethylene glycol-superoxide dismutase (PEG-SOD), 25 U/ml] or with gp91ds-tat significantly attenuated the chronotropic action of apelin-13. PEG-SOD and gp91ds-tat alone had no effect on basal neuronal firing. In addition, apelin-13 significantly increased NAD(P)H oxidase activity and elevated intracellular superoxide levels in neuronal cultures. The superoxide generator xanthine-xanthine oxidase also increased neuronal activity in neurons, mimicking the neuronal response to apelin-13. These observations provide the first evidence that apelin-13 directly increases neuronal activity via stimulation of NAD(P)H oxidase-derived superoxide, a cellular signaling mechanism that may be involved in the pressor effect of apelin-13 in the RVLM.
Collapse
Affiliation(s)
- Fanrong Yao
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58108-6050, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Colombari E, Colombari DSA, Li H, Shi P, Dong Y, Jiang N, Raizada MK, Sumners C, Murphy D, Paton JFR. Macrophage migration inhibitory factor in the paraventricular nucleus plays a major role in the sympathoexcitatory response to salt. Hypertension 2010; 56:956-63. [PMID: 20937969 DOI: 10.1161/hypertensionaha.110.155101] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Central hyperosmotic stimulation (HS) evokes increases in sympathetic nerve activity mediated by activation of angiotensin type 1 receptors in the hypothalamic paraventricular nucleus (PVN). Macrophage inhibitory migration factor (MIF) is an intracellular inhibitory regulator of angiotensin type 1 receptor-mediated actions of angiotensin II within neurons of the PVN. MIF mediates its actions via its intrinsic thiol-protein oxidoreductase activity. We demonstrate that intracerebroventricular injection of hypertonic saline into Sprague-Dawley rats elicits a significant (≈112%) increase in MIF mRNA expression in the PVN. Next, we evaluated the effect of viral-mediated expression of either MIF or [C60S]-MIF (which lacks thiol-protein oxidoreductase activity) in the PVN on the sympathoexcitation evoked by HS. We used a decorticate, arterially perfused in situ preparation of male Wistar rats (60 to 80 g). HS was induced by raising perfusate osmolality from 290 to 380 milliosmoles for 40 seconds. Seven to 10 days before experiments, rats were injected bilaterally (500 nL per side) with 0.9% saline (control) or with adenoassociated virus to express MIF, [C60S]-MIF, or enhanced green fluorescent protein in the PVN. HS produced sympathoexcitation in both the 0.9% saline and enhanced green fluorescent protein groups (sympathetic nerve activity increase of +27±4% and +25±4%, respectively; P<0.05), an effect that was not observed in the MIF group (+4±5%). Conversely, the HS-induced increase in sympathetic nerve activity was potentiated in the [C60S]-MIF group (+45±6%; P<0.05). We propose that MIF acting within the PVN is a major counterregulator of HS-induced sympathoexcitation, an effect that depends on thiol-protein oxidoreductase activity.
Collapse
Affiliation(s)
- Eduardo Colombari
- School of Physiology and Pharmacology, Bristol Heart Institute, University of Bristol, Bristol, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Inácio AR, Ruscher K, Wieloch T. Enriched environment downregulates macrophage migration inhibitory factor and increases parvalbumin in the brain following experimental stroke. Neurobiol Dis 2010; 41:270-8. [PMID: 20883785 DOI: 10.1016/j.nbd.2010.09.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2010] [Revised: 09/17/2010] [Accepted: 09/19/2010] [Indexed: 12/14/2022] Open
Abstract
Housing rodents in an enriched environment (EE) following experimental stroke enhances neurological recovery. Understanding the underlying neural cues may provide the basis for improving stroke rehabilitation. We studied the contribution of brain macrophage migration inhibitory factor (MIF) to functional recovery after permanent middle cerebral artery occlusion (pMCAo) in rats. In the cerebral cortex, MIF is predominantly found in neurons, particularly in parvalbumin interneurons. Following pMCAo, MIF increases around the infarct core, where it is located to neurons and astrocytes. Housing rats in an EE after pMCAo resulted in a decrease of MIF protein levels in peri-infarct areas, which was accompanied by an increase in parvalbumin immunoreactive interneurons. Our data suggest that MIF is part of a signaling network involved in brain plasticity, and elevated neuronal and/or astrocytic MIF levels repress the recovery of sensory-motor function after stroke. Downregulating MIF could constitute a new therapeutic approach to promote recovery after stroke.
Collapse
Affiliation(s)
- Ana R Inácio
- Laboratory for Experimental Brain Research, Department of Clinical Sciences, Lund University, BMC A13, 22184 Lund, Sweden.
| | | | | |
Collapse
|
34
|
Harrison RA, Sumners C. Redox regulation of macrophage migration inhibitory factor expression in rat neurons. Biochem Biophys Res Commun 2009; 390:171-5. [PMID: 19799867 DOI: 10.1016/j.bbrc.2009.09.112] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2009] [Accepted: 09/25/2009] [Indexed: 01/29/2023]
Abstract
Macrophage migration inhibitory factor (MIF) expression is induced by angiotensin II (Ang II) in normal rat neurons and serves a negative regulatory role by blunting the chronotropic actions of this peptide. The aim here was to determine whether hydrogen peroxide (H(2)O(2)), a reactive oxygen species (ROS) that is a key intracellular mediator of the neuronal actions of Ang II, is a trigger for MIF production in neurons. Thus, we tested the effects of H(2)O(2) on MIF expression in primary neuronal cultures from newborn normotensive (Wistar Kyoto [WKY] or Sprague-Dawley [SD]) rat brain, cells that respond to Ang II by increasing MIF levels. Treatment of WKY or SD rat neuronal cultures with a non-cytotoxic concentration of H(2)O(2) elicited a significant, time-dependent increase in MIF mRNA and protein levels. Glucose oxidase, which produces H(2)O(2) via oxidation of glucose in the cell-culture medium, elicited a similar increase in neuronal MIF mRNA levels. The stimulatory action of H(2)O(2) was not apparent in neuronal cultures from spontaneously hypertensive rats (SHR), cells that fail to express increased MIF in response to Ang II. Finally, preincubation of SD rat cultures with either polyethylene glycol-catalase or actinomycin D abolished the H(2)O(2)-induced increase in MIF, suggesting that this ROS is acting intracellularly to increase transcription of the MIF gene. These results suggest the presence of a redox regulatory mechanism for induction of MIF in normotensive rat neurons.
Collapse
Affiliation(s)
- Rachael A Harrison
- Department of Physiology and Functional Genomics & McKnight Brain Institute, University of Florida, Gainesville, FL 32610-0274, USA
| | | |
Collapse
|
35
|
Shi P, Raizada MK, Sumners C. Brain cytokines as neuromodulators in cardiovascular control. Clin Exp Pharmacol Physiol 2009; 37:e52-7. [PMID: 19566837 DOI: 10.1111/j.1440-1681.2009.05234.x] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
1. The role of cytokines in cardiovascular control, especially in neurogenic hypertension, has received considerable attention during the past few years. Brain cytokines have been shown to exert profound effects on neuronal activity. Recently, a number of studies have shown that administration of pro-inflammatory cytokines or anti-inflammatory cytokines into the central nervous system has a significant impact on sympathetic outflow, arterial pressure and cardiac remodelling in experimental models of hypertension and heart failure. 2. Our objective in this review is to present a succinct account of the effect of cytokines on neuronal activity and their role in cardiovascular disease. Furthermore, we propose a hypothesis for a neuromodulatory role of cytokines in the neural control of cardiovascular function.
Collapse
Affiliation(s)
- Peng Shi
- Department of Physiology and Functional Genomics, McKnight Brain Institute, University of Florida, Gainesville, Florida 32610-0274, USA
| | | | | |
Collapse
|
36
|
Zhang Q, Yao F, Raizada MK, O'Rourke ST, Sun C. Apelin gene transfer into the rostral ventrolateral medulla induces chronic blood pressure elevation in normotensive rats. Circ Res 2009; 104:1421-8. [PMID: 19443838 DOI: 10.1161/circresaha.108.192302] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The peripheral apelin system plays a significant role in cardiovascular homeostasis and in the pathophysiology of cardiovascular diseases. However, the central effect of this neurohormonal system in neural control of cardiovascular function remains poorly understood. Thus, this study was undertaken to evaluate the effect of apelin in the rostral ventrolateral medulla (RVLM) on blood pressure, cardiac function, and sympathetic nerve activity. Apelin mRNA and protein levels were detected with real-time RT-PCR and Western blots, respectively. Expression of apelin was significantly enhanced in the RVLM of spontaneously hypertensive rat (SHR) compared with normotensive Wistar-Kyoto (WKY) rats. To study the functional consequence of upregulated apelin expression, apelin was overexpressed by bilateral microinjection of the AAV2-apelin viral vector into the RVLM of WKY rats. Immunofluorescence staining and Western blots demonstrated that microinjection of AAV2-apelin into the RVLM resulted in a significant increase in apelin expression, which was associated with a chronic elevation in blood pressure and cardiac hypertrophy. In addition, direct microinjection of exogenous apelin-13 (200 pmol in 50 nL) into the RVLM caused a 20 mm Hg elevation in blood pressure and a 24% increase in sympathetic nerve activity. The present study is the first to show that apelin expression is enhanced in the RVLM of SHR versus WKY rats and that overexpression of this gene in the RVLM results in chronic blood pressure elevation and cardiac hypertrophy in normotensive rats. Thus, the apelin system in the RVLM may play a very important role in central blood pressure regulation and in the pathogenesis of hypertension.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Pharmaceutical Sciences, North Dakota State University, 1401 Albrecht Blvd, Fargo, ND 58105, USA
| | | | | | | | | |
Collapse
|
37
|
Herrick KA, Rossen LM, Nielsen SJ, Branum AM, Ogden CL. Macrophage migration inhibitory factor and autism spectrum disorders. Pediatrics 2008. [PMID: 26391940 DOI: 10.1542/peds] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
OBJECTIVE Autistic spectrum disorders are childhood neurodevelopmental disorders characterized by social and communicative impairment and repetitive and stereotypical behavior. Macrophage migration inhibitory factor (MIF) is an upstream regulator of innate immunity that promotes monocyte/macrophage-activation responses by increasing the expression of Toll-like receptors and inhibiting activation-induced apoptosis. On the basis of results of previous genetic linkage studies and reported altered innate immune response in autism spectrum disorder, we hypothesized that MIF could represent a candidate gene for autism spectrum disorder or its diagnostic components. METHODS Genetic association between autism spectrum disorder and MIF was investigated in 2 independent sets of families of probands with autism spectrum disorder, from the United States (527 participants from 152 families) and Holland (532 participants from 183 families). Probands and their siblings, when available, were evaluated with clinical instruments used for autism spectrum disorder diagnoses. Genotyping was performed for 2 polymorphisms in the promoter region of the MIF gene in both samples sequentially. In addition, MIF plasma analyses were conducted in a subset of Dutch patients from whom plasma was available. RESULTS There were genetic associations between known functional polymorphisms in the promoter for MIF and autism spectrum disorder-related behaviors. Also, probands with autism spectrum disorder exhibited higher circulating MIF levels than did their unaffected siblings, and plasma MIF concentrations correlated with the severity of multiple autism spectrum disorder symptoms. CONCLUSIONS These results identify MIF as a possible susceptibility gene for autism spectrum disorder. Additional research is warranted on the precise relationship between MIF and the behavioral components of autism spectrum disorder, the mechanism by which MIF contributes to autism spectrum disorder pathogenesis, and the clinical use of MIF genotyping.
Collapse
Affiliation(s)
| | - Lauren M Rossen
- Infant, Child, and Women's Health Statistics Branch, Office of Analysis and Epidemiology, and
| | | | - Amy M Branum
- Reproductive Statistics Branch, Division of Vital Statistics, National Center for Health Statistics, Centers for Disease Control and Prevention, Hyattsville, Maryland
| | | |
Collapse
|
38
|
Kurihara T, Ozawa Y, Nagai N, Shinoda K, Noda K, Imamura Y, Tsubota K, Okano H, Oike Y, Ishida S. Angiotensin II type 1 receptor signaling contributes to synaptophysin degradation and neuronal dysfunction in the diabetic retina. Diabetes 2008; 57:2191-8. [PMID: 18487452 PMCID: PMC2494692 DOI: 10.2337/db07-1281] [Citation(s) in RCA: 106] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Pathogenic mechanisms underlying diabetes-induced retinal dysfunction are not fully understood. The aim of the present study was to show the relationship of the renin-angiotensin system (RAS) with the synaptic vesicle protein synaptophysin and neuronal activity in the diabetic retina. RESEARCH DESIGN AND METHODS C57BL/6 mice with streptozotocin-induced diabetes were treated with the angiotensin II type 1 receptor (AT1R) blocker telimsartan or valsartan, and retinal function was analyzed by electroretinography. Retinal production of the RAS components and phosphorylation of ERK (extracellular-signal regulated kinase) were examined by immunoblotting. Retinal mRNA and protein levels of synaptophysin were measured by quantitative RT-PCR and immunoblot analyses, respectively. In vitro, synaptophysin levels were also evaluated using angiotensin II-stimulated PC12D neuronal cells cultured with or without the inhibition of ERK signaling or the ubiquitin-proteasome system (UPS). RESULTS Induction of diabetes led to a significant increase in retinal production of angiotensin II and AT1R together with ERK activation in the downstream of AT1R. AT1R blockade significantly reversed diabetes-induced electroretinography changes and reduction of synaptophysin protein, but not mRNA, levels in the diabetic retina. In agreement with the AT1R-mediated posttranscriptional downregulation of synaptophysin in vivo, in vitro application of angiotensin II to PC12D neuronal cells caused the UPS-mediated degradation of synaptophysin protein via AT1R, which proved to be induced by ERK activation. CONCLUSIONS These data indicate the first molecular evidence of the RAS-induced synaptophysin degradation and neuronal dysfunction in the diabetic retina, suggesting the possibility of the AT1R blockade as a novel neuroprotective treatment for diabetic retinopathy.
Collapse
Affiliation(s)
- Toshihide Kurihara
- Laboratory of Retinal Cell Biology, Keio University School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Grigorenko EL, Han SS, Yrigollen CM, Leng L, McDonald C, Mizue Y, Anderson GM, Mulder EJ, de Bildt A, Minderaa RB, Volkmar FR, Chang JT, Bucala R. Macrophage migration inhibitory factor and autism spectrum disorders. Pediatrics 2008; 122:e438-45. [PMID: 18676531 PMCID: PMC3816765 DOI: 10.1542/peds.2007-3604] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
OBJECTIVE Autistic spectrum disorders are childhood neurodevelopmental disorders characterized by social and communicative impairment and repetitive and stereotypical behavior. Macrophage migration inhibitory factor (MIF) is an upstream regulator of innate immunity that promotes monocyte/macrophage-activation responses by increasing the expression of Toll-like receptors and inhibiting activation-induced apoptosis. On the basis of results of previous genetic linkage studies and reported altered innate immune response in autism spectrum disorder, we hypothesized that MIF could represent a candidate gene for autism spectrum disorder or its diagnostic components. METHODS Genetic association between autism spectrum disorder and MIF was investigated in 2 independent sets of families of probands with autism spectrum disorder, from the United States (527 participants from 152 families) and Holland (532 participants from 183 families). Probands and their siblings, when available, were evaluated with clinical instruments used for autism spectrum disorder diagnoses. Genotyping was performed for 2 polymorphisms in the promoter region of the MIF gene in both samples sequentially. In addition, MIF plasma analyses were conducted in a subset of Dutch patients from whom plasma was available. RESULTS There were genetic associations between known functional polymorphisms in the promoter for MIF and autism spectrum disorder-related behaviors. Also, probands with autism spectrum disorder exhibited higher circulating MIF levels than did their unaffected siblings, and plasma MIF concentrations correlated with the severity of multiple autism spectrum disorder symptoms. CONCLUSIONS These results identify MIF as a possible susceptibility gene for autism spectrum disorder. Additional research is warranted on the precise relationship between MIF and the behavioral components of autism spectrum disorder, the mechanism by which MIF contributes to autism spectrum disorder pathogenesis, and the clinical use of MIF genotyping.
Collapse
Affiliation(s)
- Elena L. Grigorenko
- Child Study Center, Yale University, 230 South Frontage Rd, New Haven, CT 06519, USA,Department of Psychology, Yale University, 2 Hillhouse Ave, New Haven, CT 06520, USA,Department of Epidemiology and Public Health, Yale University, 300 Cedar St, New Haven, CT 06520,Department of Psychology, Moscow State University, 11/5 Mokhovaia St, Moscow 125009, Russia,To whom correspondence should be addressed: Telephone: 203-737-1453, 203-737-2316, Fax: 203-785-7053, 203-785-3002, ,
| | - Summer S. Han
- Department of Psychology, Moscow State University, 11/5 Mokhovaia St, Moscow 125009, Russia
| | - Carolyn M. Yrigollen
- Child Study Center, Yale University, 230 South Frontage Rd, New Haven, CT 06519, USA
| | - Lin Leng
- Department of Epidemiology and Public Health, Yale University, 300 Cedar St, New Haven, CT 06520,Departments of Internal Medicine and Pathology, Yale University, 300 Cedar St, New Haven, CT 06520
| | - Courtney McDonald
- Department of Epidemiology and Public Health, Yale University, 300 Cedar St, New Haven, CT 06520,Departments of Internal Medicine and Pathology, Yale University, 300 Cedar St, New Haven, CT 06520
| | - Yuka Mizue
- Sapporo Immuno Diagnostic Laboratory, 12-20, Shinkawa 2-2, Kita-ku, Sapporo 001-0922, Japan
| | - George M. Anderson
- Child Study Center, Yale University, 230 South Frontage Rd, New Haven, CT 06519, USA
| | - Erik J. Mulder
- Accare/University Medical Center Groningen, University Center for Child and Adolescent Psychiatry, P.O. Box 660, 9700 AR Groningen, the Netherlands
| | - Annelies de Bildt
- Accare/University Medical Center Groningen, University Center for Child and Adolescent Psychiatry, P.O. Box 660, 9700 AR Groningen, the Netherlands
| | - Ruud B. Minderaa
- Accare/University Medical Center Groningen, University Center for Child and Adolescent Psychiatry, P.O. Box 660, 9700 AR Groningen, the Netherlands
| | - Fred R. Volkmar
- Child Study Center, Yale University, 230 South Frontage Rd, New Haven, CT 06519, USA,Department of Psychology, Yale University, 2 Hillhouse Ave, New Haven, CT 06520, USA
| | - Joseph T. Chang
- Department of Psychology, Moscow State University, 11/5 Mokhovaia St, Moscow 125009, Russia
| | - Richard Bucala
- Department of Epidemiology and Public Health, Yale University, 300 Cedar St, New Haven, CT 06520,Departments of Internal Medicine and Pathology, Yale University, 300 Cedar St, New Haven, CT 06520,To whom correspondence should be addressed: Telephone: 203-737-1453, 203-737-2316, Fax: 203-785-7053, 203-785-3002, ,
| |
Collapse
|
40
|
Pearce BD, Garvin SE, Grove J, Bonney EA, Dudley DJ, Schendel DE, Thorsen P. Serum macrophage migration inhibitory factor in the prediction of preterm delivery. Am J Obstet Gynecol 2008; 199:46.e1-6. [PMID: 18241824 DOI: 10.1016/j.ajog.2007.11.066] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2007] [Revised: 10/02/2007] [Accepted: 11/27/2007] [Indexed: 11/29/2022]
Abstract
OBJECTIVE Macrophage migration inhibitory factor is a soluble mediator that helps govern the interaction between cytokines and stress hormones (eg, cortisol). We determined whether maternal macrophage migration inhibitory factor levels predicted subsequent preterm delivery. STUDY DESIGN A nested case-control study measuring serum macrophage migration inhibitory factor concentration at 9-23 weeks' gestation in women who ultimately delivered preterm (n = 60) compared with control women who delivered at term (n = 122). We also examined the connection of macrophage migration inhibitory factor with self-reported psychosocial variables. RESULTS Macrophage migration inhibitory factor was elevated in the preterm delivery cases (P = .0004), and log macrophage migration inhibitory factor concentration showed a graded response relationship with likelihood of preterm delivery. High-macrophage migration inhibitory factor was also associated with maternal risk-taking behavior, which itself was a risk factor for preterm delivery. Macrophage migration inhibitory factor remained associated independently with preterm delivery after adjusting regression models for several other preterm delivery risk factors (odds ratio, 3.11, 95% confidence interval, 1.54-6.30). CONCLUSION High-serum macrophage migration inhibitory concentration in early to midpregnancy is linked with subsequent preterm delivery.
Collapse
Affiliation(s)
- Brad D Pearce
- Department of Psychology, Emory University, Atlanta, GA 30332, USA.
| | | | | | | | | | | | | |
Collapse
|
41
|
Li H, Gao Y, Qi Y, Katovich MJ, Jiang N, Braseth LN, Scheuer DA, Shi P, Sumners C. Macrophage migration inhibitory factor in hypothalamic paraventricular nucleus neurons decreases blood pressure in spontaneously hypertensive rats. FASEB J 2008; 22:3175-85. [PMID: 18535252 DOI: 10.1096/fj.08-108662] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Macrophage migration inhibitory factor (MIF) expression is increased by angiotensin II (Ang II) within paraventricular nucleus (PVN) neurons of normotensive rats and acts via its intrinsic thiol protein oxidoreductase (TPOR) to counterregulate the central nervous system-mediated pressor action of Ang II. Considering that the PVN-mediated actions of Ang II are enhanced in spontaneously hypertensive rats (SHRs) and contribute to the development of hypertension in these animals, we investigated this MIF regulatory mechanism in SHRs. Here, we have demonstrated that Ang II failed to increase MIF protein expression in the PVN of SHRs. Furthermore, although basal levels of MIF protein and mRNA were similar in the PVN of SHRs and normotensive rats, immunostaining revealed that MIF was either absent from or diminished in PVN neurons of SHRs. AAV2-mediated increases in MIF expression within PVN neurons of young (8 wk old) SHRs produced a chronic attenuation of hypertension and cardiac hypertrophy. However, similar AAV2-mediated transduction of [C60S]-MIF, which lacks TPOR activity, did not alter the development of hypertension or cardiac hypertrophy in SHRs. Collectively, these findings suggest that a lack of MIF expression within PVN neurons contributes to the development of hypertension and cardiac hypertrophy in SHRs.
Collapse
Affiliation(s)
- Hongwei Li
- Dept. of Physiology and Functional Genomics, College of Medicine, 1600 SW Archer Rd., University of Florida, Gainesville, FL 32610-0274, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Yao F, Sumners C, O'Rourke ST, Sun C. Angiotensin II increases GABAB receptor expression in nucleus tractus solitarii of rats. Am J Physiol Heart Circ Physiol 2008; 294:H2712-20. [PMID: 18424635 PMCID: PMC4422374 DOI: 10.1152/ajpheart.00729.2007] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2007] [Accepted: 04/15/2008] [Indexed: 11/22/2022]
Abstract
Increasing evidence indicates that both the angiotensin II (ANG II) and gamma-aminobutyric acid (GABA) systems play a very important role in the regulation of blood pressure (BP). However, there is little information concerning the interactions between these two systems in the nucleus tractus solitarii (NTS). In the present study, we examined the effects of ANG II on GABAA and GABAB receptor (GAR and GBR) expression in the NTS of Sprague-Dawley rats. The direct effect of ANG II on GBR expression was determined in neurons cultured from NTS. Treatment of neuronal cultures with ANG II (100 nM, 5 h) induced a twofold increase in GBR1 expression, as detected with real-time RT-PCR and Western blots, but had no effect on GBR2 or GAR expression. In electrophysiological experiments, perfusion of neuronal cultures with the GBR agonist baclofen decreased neuronal firing rate by 39% and 63% in neurons treated with either PBS (control) or ANG II, respectively, indicating that chronic ANG II treatment significantly enhanced the neuronal response to GBR activation. In contrast, ANG II had no significant effect on the inhibitory action of the GAR agonist muscimol. In whole animal studies, intracerebroventricular infusion of ANG II induced a sustained increase in mean BP and an elevation of GBR1 mRNA and protein levels in the NTS. These results indicate that ANG II stimulates GBR expression in NTS neurons, and this could contribute to the central nervous system actions of ANG II that result in dampening of baroreflexes and elevated BP in the central actions of ANG II.
Collapse
MESH Headings
- Action Potentials
- Angiotensin II/administration & dosage
- Angiotensin II/metabolism
- Animals
- Animals, Newborn
- Baclofen/pharmacology
- Baroreflex/drug effects
- Blood Pressure/drug effects
- Blotting, Western
- Cells, Cultured
- Disease Models, Animal
- GABA Agonists/pharmacology
- GABA-B Receptor Agonists
- Hypertension/chemically induced
- Hypertension/metabolism
- Hypertension/physiopathology
- Infusions, Parenteral
- Male
- Muscimol/pharmacology
- Neurons/drug effects
- Neurons/metabolism
- RNA, Messenger/metabolism
- Rats
- Rats, Sprague-Dawley
- Receptor, Angiotensin, Type 1/metabolism
- Receptor, Angiotensin, Type 2/metabolism
- Receptors, GABA-A/metabolism
- Receptors, GABA-B/genetics
- Receptors, GABA-B/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Solitary Nucleus/cytology
- Solitary Nucleus/drug effects
- Solitary Nucleus/metabolism
- Time Factors
- Up-Regulation
Collapse
Affiliation(s)
- Fanrong Yao
- Department of Pharmaceutical Sciences, College of Pharmacy, North Dakota State University, Fargo, North Dakota, USA
| | | | | | | |
Collapse
|
43
|
Matsuura T, Harrison RA, Westwell AD, Nakamura H, Martynyuk AE, Sumners C. Basal and angiotensin II-inhibited neuronal delayed-rectifier K+ current are regulated by thioredoxin. Am J Physiol Cell Physiol 2007; 293:C211-7. [PMID: 17360810 DOI: 10.1152/ajpcell.00615.2006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In previous studies, we determined that macrophage migration inhibitory factor (MIF), acting intracellularly via its intrinsic thiol-protein oxidoreductase (TPOR) activity, stimulates basal neuronal delayed-rectifier K+ current ( IKv) and inhibits basal and angiotensin (ANG) II-induced increases in neuronal activity. These findings are the basis for our hypothesis that MIF is a negative regulator of ANG II actions in neurons. MIF has recently been recategorized as a member of the thioredoxin (Trx) superfamily of small proteins. In the present study we have examined whether Trx influences basal and ANG II-modulated IKv in an effort to determine whether the Trx superfamily can exert a general regulatory influence over neuronal activity and the actions of ANG II. Intracellular application of Trx (0.8–80 nM) into rat hypothalamic/brain stem neurons in culture increased neuronal IKv, as measured by voltage-clamp recordings. This effect of Trx was abolished in the presence of the TPOR inhibitor PMX 464 (800 nM). Furthermore, the mutant protein recombinant human C32S/C35S-Trx, which lacks TPOR activity, failed to alter neuronal IKv. Trx applied at a concentration (0.08 nM) that does not alter basal IKv abolished the inhibition of neuronal IKv produced by ANG II (100 nM). Given our observation that ANG II increases Trx levels in neuronal cultures, it is possible that Trx (like MIF) has a negative regulatory role over basal and ANG II-stimulated neuronal activity via modulation of IKv. Moreover, these data suggest that TPOR may be a general mechanism for negatively regulating neuronal activity.
Collapse
Affiliation(s)
- Tomokazu Matsuura
- Dept. of Physiology and Functional Genomics, College of Medicine, University of Florida, Box 100274, 1600 SW Archer Rd., Gainesville, FL 32610-0274, USA
| | | | | | | | | | | |
Collapse
|
44
|
Peterson JR, Sharma RV, Davisson RL. Reactive oxygen species in the neuropathogenesis of hypertension. Curr Hypertens Rep 2007; 8:232-41. [PMID: 17147922 DOI: 10.1007/s11906-006-0056-1] [Citation(s) in RCA: 112] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
New evidence that has emerged during the past several years clearly demonstrates that reactive oxygen species (ROS) in the brain play a crucial role in blood pressure regulation by serving as signaling molecules within neurons of cardiovascular control regions. In the forebrain, midbrain, and hindbrain, a key role for oxidant stress in the pathogenesis of angiotensin II-dependent and various other models of neurogenic hypertension has also been uncovered. As in the peripheral vasculature, NAD(P)H oxidase appears to be a major enzymatic source of brain ROS, and various homologues of the catalytic subunit of this enzyme appear to be differentially localized to cardiovascular-regulating nuclei in the brain. Recent studies have begun to elucidate the downstream effects of ROS in neurons, and it is now clear that ROS may interact with a number of well-described intracellular signaling pathways involved in neuronal activation. These exciting new discoveries have furthered our understanding of the pathogenesis of neurogenic hypertension and may ultimately lead to the development of new treatments. In this review, we discuss recent evidence in support of a role for brain ROS in the pathogenesis of hypertension and summarize current studies aimed at uncovering the complex mechanisms by which brain ROS regulate blood pressure in both health and cardiovascular disease.
Collapse
Affiliation(s)
- Jeffrey R Peterson
- Anatomy and Cell Biology, 1-251 Bowen Science Building, The Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA 52245, USA
| | | | | |
Collapse
|
45
|
Sun C, Li H, Gao Y, Matsuura T, Upchurch PA, Raizada MK, Sumners C. Lack of macrophage migration inhibitory factor regulation is linked to the increased chronotropic action of angiotensin II in SHR neurons. Hypertension 2007; 49:528-34. [PMID: 17261648 DOI: 10.1161/01.hyp.0000257877.11495.cb] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Macrophage migration inhibitory factor acts via its intrinsic thiol-protein oxidoreductase activity to negatively regulate the neuronal chronotropic actions of angiotensin II in normotensive rat neurons. Because the chronotropic action of angiotensin II is potentiated in spontaneously hypertensive rat neurons, we investigated whether this negative regulatory mechanism is absent in these rats. Angiotensin II (100 nM) elicited an approximately 89% increase in neuronal firing in Wistar-Kyoto rat hypothalamus and brain stem cultured neurons and an increase in intracellular macrophage migration inhibitory factor levels in the same cells. The chronotropic action of angiotensin II was significantly greater (approximately 212% increase) in spontaneously hypertensive rat neurons, but angiotensin II failed to alter macrophage migration inhibitory factor expression in these cells. Intracellular application of recombinant macrophage migration inhibitory factor (0.8 nM) or its specific neuronal overexpression via Ad5-SYN-MIF (1x10(7) infectious units) significantly attenuated the chronotropic action of angiotensin II in spontaneously hypertensive rat neurons, similar to results from Wistar-Kyoto rat neurons. In contrast, C60S-macrophage migration inhibitory factor (0.8 nM), which lacks thiol-protein oxidoreductase activity, failed to alter the chronotropic action of angiotensin II in neurons from either rat strain. Thus, whereas macrophage migration inhibitory factor has the potential to depress the chronotropic action of angiotensin II in spontaneously hypertensive rat neurons, it is unlikely that this regulatory mechanism occurs, because angiotensin II does not increase the expression of this protein. The lack of this regulatory mechanism may contribute to the increased chronotropic action of angiotensin II in spontaneously hypertensive rat neurons.
Collapse
Affiliation(s)
- Chengwen Sun
- Department of Physiology and Functional Genomics and McKnight Brain Institute, University of Florida, Gainesville, FL 32610-0274, USA
| | | | | | | | | | | | | |
Collapse
|
46
|
Li H, Gao Y, Freire CD, Raizada MK, Toney GM, Sumners C. Macrophage migration inhibitory factor in the PVN attenuates the central pressor and dipsogenic actions of angiotensin II. FASEB J 2006; 20:1748-50. [PMID: 16809436 DOI: 10.1096/fj.06-5836fje] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Macrophage migration inhibitory factor (MIF) acts intracellularly to counteract the angiotensin (ANG) II type 1 receptor (AT1-R)-mediated chronotropic effect of ANG II in hypothalamic neurons, an effect mediated by the thiol-protein oxidoreductase (TPOR) activity of the MIF molecule. Here we determined the in vivo actions of MIF in regulating the physiological actions of ANG II that are mediated via the paraventricular nucleus (PVN), an area that serves as a relay point in the central nervous system (CNS)-mediated effects of ANG II on cardiovascular functions and water intake. Intracerebroventricular (icv) injection of ANG II into normotensive rats selectively increased MIF protein levels in the PVN and produced significant pressor and drinking responses that were inhibited by PVN administration of the AT1-R antagonist losartan. Overexpression of MIF in PVN neurons via Ad-Syn-MIF gene transfer attenuated the pressor and drinking responses produced by icv-injected ANG II. Consistently, intracellular application of MIF or MIF-(50-65) (which harbors the TPOR activity of MIF) into PVN sympathetic regulatory neurons, blunted the electrophysiological actions of ANG II at these cells. These observations establish for the first time that MIF within the PVN, acting via TPOR, is an intracellular regulator of the central cardiovascular and dipsogenic effects of ANG II.
Collapse
Affiliation(s)
- Hongwei Li
- Department of Physiology and Functional Genomics and McKnight Brain Institute, University of Florida, Gainesville, Florida 32610-0274, USA
| | | | | | | | | | | |
Collapse
|
47
|
Matsuura T, Sun C, Leng L, Kapurniotu A, Bernhagen J, Bucala R, Martynyuk AE, Sumners C. Macrophage Migration Inhibitory Factor Increases Neuronal Delayed Rectifier K+ Current. J Neurophysiol 2006; 95:1042-8. [PMID: 16267117 DOI: 10.1152/jn.00499.2005] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Macrophage migration inhibitory factor (MIF) has widespread actions in the immune, endocrine, and nervous systems. Previously, we reported that increases in the intracellular levels of MIF depress the firing of hypothalamus/brain stem neurons in culture, including the chronotropic actions of angiotensin II. The objective of this study was to investigate the effects of MIF on delayed rectifier K+ current ( IKv), one of the component currents whose activity contributes to neuronal firing. Intracellular perfusion of MIF (80 nM) into Sprague–Dawley rat neuronal cultures caused a significant increase in IKv, as measured by patch-clamp recordings. This effect was apparent by 3 min, and was maximal after 20–30 min. IKv current density (pA/pF) increased from 31.58 ± 2.36 in controls to 41.88 ± 3.76 in MIF-treated neurons (mean ± SE; n = 9; P < 0.01). MIF that had been inactivated by boiling did not alter IKv, and MIF-neutralizing antibodies abolished the action of recombinant MIF (rMIF). The stimulatory effect of MIF on IKv current density was mimicked by intracellular application of either P1S-MIF (80 nM) or the peptide MIF-(50–65) (0.8–8 μM), both of which harbor the thiol-protein oxidoreductase (TPOR) activity of the MIF molecule. Conversely, neither C60S-MIF (80 nM) nor the MIF homologue D-dopachrome tautomerase (80 nM), both of which lack TPOR activity, altered IKv. Finally, the increase in IKv produced by rMIF was abolished by the superoxide scavenger Tiron (1 mM). These studies indicate that the neuronal action of MIF includes a stimulatory action on IKv that may be mediated by a TPOR/superoxide-scavenging mechanism.
Collapse
Affiliation(s)
- Tomokazu Matsuura
- Department of Physiology and Functional Genomics and McKnight Brain Institute, University of Florida, Gainesville, FL 32610-0274, USA
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Li HW, Gao YX, Raizada MK, Sumners C. Intronic enhancement of angiotensin II type 2 receptor transgene expression in vitro and in vivo. Biochem Biophys Res Commun 2005; 336:29-35. [PMID: 16122703 DOI: 10.1016/j.bbrc.2005.08.035] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2005] [Accepted: 08/05/2005] [Indexed: 11/24/2022]
Abstract
The angiotensin II type 2 receptor (AT2R) can influence a variety of intracellular signaling molecules and cellular functions. However, its physiological functions and the roles of introns in the regulation of its expression have not been well determined. We first demonstrated that both intron 1 and intron 2 of AT2R could significantly enhance AT2R overexpression. Thus, we have provided some new prerequisites for further studies on the mechanisms that control AT2R gene expression. Next, we established a highly efficient method of delivering this receptor in vitro and in vivo using an AT2R recombinant adenoviral vector containing two introns of the AT2R. The vector may be useful in helping to uncover AT2R physiological functions and also for gene therapy related to AT2R. Moreover, we determined the important role of introns in gene expression cassettes and the inconsistency of expression between the targeted gene and the reporter gene.
Collapse
Affiliation(s)
- Hong-wei Li
- Department of Physiology and Functional Genomics and McKnight Brain Institute, University of Florida, Gainesville, FL 32610-0274, USA.
| | | | | | | |
Collapse
|
49
|
Thiele M, Bernhagen J. Link between macrophage migration inhibitory factor and cellular redox regulation. Antioxid Redox Signal 2005; 7:1234-48. [PMID: 16115028 DOI: 10.1089/ars.2005.7.1234] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Macrophage migration inhibitory factor (MIF) is an evolutionary conserved 12.5-kDa protein mediator with multiple functions in innate and acquired immunity. Upon leaderless secretion, MIF acts as a typical inflammatory cytokine, but there is no structural homology between MIF and any of the known cytokine protein families. Also, MIF is unique among cytokines in that it exhibits certain endocrine properties and has enzymatic activity. The catalytic thiol-protein oxidoreductase (TPOR) activity of MIF is mediated by a Cys-Ala-Leu-Cys active site between residues 57 and 60 that can undergo reversible intramolecular disulfide formation. Such a redox motif is typically found in TPORs of the thioredoxin (Trx) family of proteins. MIF seems to act as a disulfide reductase, and structure-function analyses of the redox site indicate that this activity is not only observed in vitro, but plays a role in cellular redox homeostasis, apoptosis inhibition, MIF-mediated monocyte/macrophage activation, and possibly the modulation of the activity of MIF-binding proteins. In this Forum review, the biochemical and biological evidence for a role of the TPOR activity for various MIF functions is summarized and discussed. In particular, the marked functional homologies with Trx proteins, the MIF redox/MHC II link, and recent attempts to discern the intra- versus extracellular roles of the MIF TPOR activity are dealt with.
Collapse
Affiliation(s)
- Michael Thiele
- Department of Biochemistry and Molecular Cell Biology, Institute of Biochemistry, University Hospital RWTH Aachen, Aachen, Germany
| | | |
Collapse
|
50
|
Burger-Kentischer A, Finkelmeier D, Thiele M, Schmucker J, Geiger G, Tovar GEM, Bernhagen J. Binding of JAB1/CSN5 to MIF is mediated by the MPN domain but is independent of the JAMM motif. FEBS Lett 2005; 579:1693-701. [PMID: 15757663 DOI: 10.1016/j.febslet.2005.01.080] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2004] [Revised: 01/07/2005] [Accepted: 01/31/2005] [Indexed: 11/26/2022]
Abstract
Macrophage migration inhibitory factor (MIF) binds to c-Jun activation domain binding protein-1 (JAB1)/subunit 5 of COP9 signalosome (CSN5) and modulates cell signaling and the cell cycle through JAB1. The binding domain of JAB1 responsible for binding to MIF is unknown. We hypothesized that the conserved Mpr1p Pad1p N-terminal (MPN) domain of JAB1 may mediate binding to MIF. In fact, yeast two hybrid (YTH) and in vitro translation/coimmunoprecipitation (CoIP) analysis showed that a core MPN domain, which did not cover the functional JAB1/MPN/Mov34 metalloenzyme (JAMM) deneddylase sequence, binds to MIF comparable to full-length JAB1. YTH and pull-down analysis in conjunction with nanobead affinity matrix-assisted laser desorption ionization-time of flight (MALDI-TOF) mass spectrometry demonstrated that MIF(50-65) and MPN are sufficient to mediate MIF-JAB1 interaction, respectively. Finally, endogenous CoIP of MIF-CSN6 complexes from mammalian cells demonstrated that MPN is responsible for MIF-JAB1 binding in vivo, and, as CSN6 does not contain a functional JAMM motif, confirmed that the interaction does not require JAMM.
Collapse
Affiliation(s)
- Anke Burger-Kentischer
- Fraunhofer Institute for Interfacial Engineering and Biotechnology, Nobelstrasse 12, D-70569 Stuttgart, Germany
| | | | | | | | | | | | | |
Collapse
|