1
|
Haider AA, Rex TS, Wareham LK. cGMP Signaling in the Neurovascular Unit-Implications for Retinal Ganglion Cell Survival in Glaucoma. Biomolecules 2022; 12:1671. [PMID: 36421684 PMCID: PMC9687235 DOI: 10.3390/biom12111671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/07/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
Glaucoma is a progressive age-related disease of the visual system and the leading cause of irreversible blindness worldwide. Currently, intraocular pressure (IOP) is the only modifiable risk factor for the disease, but even as IOP is lowered, the pathology of the disease often progresses. Hence, effective clinical targets for the treatment of glaucoma remain elusive. Glaucoma shares comorbidities with a multitude of vascular diseases, and evidence in humans and animal models demonstrates an association between vascular dysfunction of the retina and glaucoma pathology. Integral to the survival of retinal ganglion cells (RGCs) is functional neurovascular coupling (NVC), providing RGCs with metabolic support in response to neuronal activity. NVC is mediated by cells of the neurovascular unit (NVU), which include vascular cells, glial cells, and neurons. Nitric oxide-cyclic guanosine monophosphate (NO-cGMP) signaling is a prime mediator of NVC between endothelial cells and neurons, but emerging evidence suggests that cGMP signaling is also important in the physiology of other cells of the NVU. NO-cGMP signaling has been implicated in glaucomatous neurodegeneration in humans and mice. In this review, we explore the role of cGMP signaling in the different cell types of the NVU and investigate the potential links between cGMP signaling, breakdown of neurovascular function, and glaucoma pathology.
Collapse
Affiliation(s)
| | | | - Lauren K. Wareham
- Vanderbilt Eye Institute, Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, Nashville, TN 37212, USA
| |
Collapse
|
2
|
Jehle A, Garaschuk O. The Interplay between cGMP and Calcium Signaling in Alzheimer's Disease. Int J Mol Sci 2022; 23:7048. [PMID: 35806059 PMCID: PMC9266933 DOI: 10.3390/ijms23137048] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/31/2022] [Accepted: 06/22/2022] [Indexed: 02/04/2023] Open
Abstract
Cyclic guanosine monophosphate (cGMP) is a ubiquitous second messenger and a key molecule in many important signaling cascades in the body and brain, including phototransduction, olfaction, vasodilation, and functional hyperemia. Additionally, cGMP is involved in long-term potentiation (LTP), a cellular correlate of learning and memory, and recent studies have identified the cGMP-increasing drug Sildenafil as a potential risk modifier in Alzheimer's disease (AD). AD development is accompanied by a net increase in the expression of nitric oxide (NO) synthases but a decreased activity of soluble guanylate cyclases, so the exact sign and extent of AD-mediated imbalance remain unclear. Moreover, human patients and mouse models of the disease present with entangled deregulation of both cGMP and Ca2+ signaling, e.g., causing changes in cGMP-mediated Ca2+ release from the intracellular stores as well as Ca2+-mediated cGMP production. Still, the mechanisms governing such interplay are poorly understood. Here, we review the recent data on mechanisms underlying the brain cGMP signaling and its interconnection with Ca2+ signaling. We also discuss the recent evidence stressing the importance of such interplay for normal brain function as well as in Alzheimer's disease.
Collapse
Affiliation(s)
| | - Olga Garaschuk
- Department of Neurophysiology, Institute of Physiology, Eberhard Karls University of Tübingen, 72074 Tübingen, Germany;
| |
Collapse
|
3
|
Modulation of Inflammatory Cytokine Production in Human Monocytes by cGMP and IRAK3. Int J Mol Sci 2022; 23:ijms23052552. [PMID: 35269704 PMCID: PMC8909980 DOI: 10.3390/ijms23052552] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 02/23/2022] [Accepted: 02/24/2022] [Indexed: 12/13/2022] Open
Abstract
Interleukin-1 receptor-associated kinase-3 (IRAK3) is a critical checkpoint molecule of inflammatory responses in the innate immune system. The pseudokinase domain of IRAK3 contains a guanylate cyclase (GC) centre that generates small amounts of cyclic guanosine monophosphate (cGMP) associated with IRAK3 functions in inflammation. However, the mechanisms of IRAK3 actions are poorly understood. The effects of low cGMP levels on inflammation are unknown, therefore a dose–response effect of cGMP on inflammatory markers was assessed in THP-1 monocytes challenged with lipopolysaccharide (LPS). Sub-nanomolar concentrations of membrane permeable 8-Br-cGMP reduced LPS-induced NFκB activity, IL-6 and TNF-α cytokine levels. Pharmacologically upregulating cellular cGMP levels using a nitric oxide donor reduced cytokine secretion. Downregulating cellular cGMP using a soluble GC inhibitor increased cytokine levels. Knocking down IRAK3 in THP-1 cells revealed that unlike the wild type cells, 8-Br-cGMP did not suppress inflammatory responses. Complementation of IRAK3 knockdown cells with wild type IRAK3 suppressed cytokine production while complementation with an IRAK3 mutant at GC centre only partially restored this function. Together these findings indicate low levels of cGMP form a critical component in suppressing cytokine production and in mediating IRAK3 action, and this may be via a cGMP enriched nanodomain formed by IRAK3 itself.
Collapse
|
4
|
Ju Hwang C, Choi DY, Park MH, Hong JT. NF-κB as a Key Mediator of Brain Inflammation in Alzheimer's Disease. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2020; 18:3-10. [PMID: 28782486 DOI: 10.2174/1871527316666170807130011] [Citation(s) in RCA: 118] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 04/24/2017] [Accepted: 04/24/2017] [Indexed: 12/22/2022]
Abstract
Alzheimer's disease is the most common form of dementia. It is characterized by betaamyloid peptide fibrils which are extracellular deposition of a specific protein, accompanied by extensive neuroinflammation. Various studies show the presence of a number of inflammation markers in the AD brain: elevated inflammatory cytokines and chemokines, and an accumulation of activated microglia in the damaged regions. NF-κB is a family of redox sensitive transcriptional factors, and it is known that NF-κB has binding sites in the promoter region of the genes involved in amyloidogenesis and inflammation. Long-term use of non-steroidal anti-inflammatory drugs prevents progression of AD and delays its onset, suggesting that there is a close correlation between NF-κB and AD pathogenesis. This study aims to (1) assess the association between NF-κB activity and AD through discussion of a variety of experimental and clinical studies on AD and (2) review treatment strategies designed to treat or prevent AD with NF-κB inhibitors.
Collapse
Affiliation(s)
- Chul Ju Hwang
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro 194-31, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, 361-951, Korea
| | - Dong-Young Choi
- College of Pharmacy, Yeungnam University, 280 Daehak-ro, Gyeongsan, Gyeongbuk 38541, Korea
| | - Mi Hee Park
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro 194-31, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, 361-951, Korea
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro 194-31, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, 361-951, Korea
| |
Collapse
|
5
|
Maksoud MJE, Tellios V, An D, Xiang Y, Lu W. Nitric oxide upregulates microglia phagocytosis and increases transient receptor potential vanilloid type 2 channel expression on the plasma membrane. Glia 2019; 67:2294-2311. [DOI: 10.1002/glia.23685] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 07/02/2019] [Accepted: 07/06/2019] [Indexed: 12/21/2022]
Affiliation(s)
- Matthew J. E. Maksoud
- Graduate Program of Neuroscience The University of Western Ontario London Canada
- Department of Molecular Medicine Robarts Research Institute, The University of Western Ontario London Canada
| | - Vasiliki Tellios
- Graduate Program of Neuroscience The University of Western Ontario London Canada
- Department of Molecular Medicine Robarts Research Institute, The University of Western Ontario London Canada
| | - Dong An
- Department of Molecular Medicine Robarts Research Institute, The University of Western Ontario London Canada
| | - Yun‐Yan Xiang
- Department of Molecular Medicine Robarts Research Institute, The University of Western Ontario London Canada
| | - Wei‐Yang Lu
- Graduate Program of Neuroscience The University of Western Ontario London Canada
- Department of Molecular Medicine Robarts Research Institute, The University of Western Ontario London Canada
- Department of Physiology and Pharmacology The University of Western Ontario London Canada
| |
Collapse
|
6
|
Mahinrad S, Bulk M, van der Velpen I, Mahfouz A, van Roon-Mom W, Fedarko N, Yasar S, Sabayan B, van Heemst D, van der Weerd L. Natriuretic Peptides in Post-mortem Brain Tissue and Cerebrospinal Fluid of Non-demented Humans and Alzheimer's Disease Patients. Front Neurosci 2018; 12:864. [PMID: 30534047 PMCID: PMC6275179 DOI: 10.3389/fnins.2018.00864] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 11/05/2018] [Indexed: 12/13/2022] Open
Abstract
Animal studies suggest the involvement of natriuretic peptides (NP) in several brain functions that are known to be disturbed during Alzheimer's disease (AD). However, it remains unclear whether such findings extend to humans. In this study, we aimed to: (1) map the gene expression and localization of NP and their receptors (NPR) in human post-mortem brain tissue; (2) compare the relative amounts of NP and NPR between the brain tissue of AD patients and non-demented controls, and (3) compare the relative amounts of NP between the cerebrospinal fluid (CSF) of AD patients and non-demented controls. Using the publicly available Allen Human Brain Atlas dataset, we mapped the gene expression of NP and NPR in healthy humans. Using immunohistochemistry, we visualized the localization of NP and NPR in the frontal cortex of AD patients (n = 10, mean age 85.8 ± 6.2 years) and non-demented controls (mean age = 80.2 ± 9.1 years). Using Western blotting and ELISA, we quantified the relative amounts of NP and NPR in the brain tissue and CSF of these AD patients and non-demented controls. Our results showed that NP and NPR genes were ubiquitously expressed throughout the brain in healthy humans. NP and NPR were present in various cellular structures including in neurons, astrocyte-like structures, and cerebral vessels in both AD patients and non-demented controls. Furthermore, we found higher amounts of NPR type-A in the brain of AD patients (p = 0.045) and lower amounts of NP type-B in the CSF of AD patients (p = 0.029). In conclusion, this study shows the abundance of NP and NPR in the brain of humans suggesting involvement of NP in various brain functions. In addition, our findings suggest alterations of NP levels in the brain of AD patients. The role of NP in the development and progression of AD remains to be elucidated.
Collapse
Affiliation(s)
- Simin Mahinrad
- Department of Internal Medicine, Section Gerontology and Geriatrics, Leiden University Medical Center, Leiden, Netherlands
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Marjolein Bulk
- Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands
- Percuros BV, Leiden, Netherlands
| | - Isabelle van der Velpen
- Department of Internal Medicine, Section Gerontology and Geriatrics, Leiden University Medical Center, Leiden, Netherlands
| | - Ahmed Mahfouz
- Leiden Computational Biology Center, Leiden University Medical Center, Leiden, Netherlands
- Delft Bioinformatics Lab, Delft University of Technology, Delft, Netherlands
| | - Willeke van Roon-Mom
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands
| | - Neal Fedarko
- Clinical Research Core Laboratory, Johns Hopkins Institute for Clinical and Translational Research, Baltimore, MD, United States
| | - Sevil Yasar
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Behnam Sabayan
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Diana van Heemst
- Department of Internal Medicine, Section Gerontology and Geriatrics, Leiden University Medical Center, Leiden, Netherlands
| | - Louise van der Weerd
- Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
7
|
Gu C, Hu Q, Wu J, Mu C, Ren H, Liu CF, Wang G. P7C3 Inhibits LPS-Induced Microglial Activation to Protect Dopaminergic Neurons Against Inflammatory Factor-Induced Cell Death in vitro and in vivo. Front Cell Neurosci 2018; 12:400. [PMID: 30455635 PMCID: PMC6230654 DOI: 10.3389/fncel.2018.00400] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 10/17/2018] [Indexed: 12/17/2022] Open
Abstract
Parkinson’s disease (PD) is the second most common neurodegenerative disorder. Although its pathogenesis remains unclear, growing evidencce suggests that microglia-mediated neuroinflammation contributes greatly to the progression of PD. P7C3, an aminopropyl carbazole, possesses significant neuroprotective effects in several neurodegenerative disease animal models, including PD. In this study, we designed to investigate the effects of P7C3 on neuroinflammation. We showed that P7C3 specially suppressed the expression of lipopolysaccharide (LPS)-induced pro-inflammatory factors but not influenced the anti-inflammatory factors in microglia. The inhibition of the nuclear factor κB (NF-κB) signaling pathway was involved in the mechanisms of the anti-inflammatory effects by P7C3. LPS-induced activation of IκB kinase (IKK), degradation of the inhibitory κB alpha (IκBα) and nuclear translocation of NF-κB can be attenuated by the pretreatment of P7C3 in microglia. Furthermore, in LPS-treated microglia, P7C3-pretreatment decreased the toxicity of conditioned media to MES23.5 cells (a dopaminergic (DA) cell line). Most importantly, the anti-inflammatory effects of P7C3 were observed in LPS-stimulated mouse model. In general, our study demonstrates that P7C3 inhibits LPS-induced microglial activation through repressing the NF-κB pathway both in vivo and in vitro, providing a theoretical basis for P7C3 in anti-inflammation.
Collapse
Affiliation(s)
- Chao Gu
- Laboratory of Molecular Neuropathology, Jiangsu Key laboratory of Translational Research and Therapy for Neuropsychiatric Disorders & Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Qingsong Hu
- Laboratory of Molecular Neuropathology, Jiangsu Key laboratory of Translational Research and Therapy for Neuropsychiatric Disorders & Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Jiayuan Wu
- The Key Laboratory, The Second Affiliated Hospital of Jiaxing University, Hangzhou, China
| | - Chenchen Mu
- Laboratory of Molecular Neuropathology, Jiangsu Key laboratory of Translational Research and Therapy for Neuropsychiatric Disorders & Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Haigang Ren
- Laboratory of Molecular Neuropathology, Jiangsu Key laboratory of Translational Research and Therapy for Neuropsychiatric Disorders & Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Chun-Feng Liu
- Institute of Neuroscience, Soochow University, Suzhou, China.,Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Guanghui Wang
- Laboratory of Molecular Neuropathology, Jiangsu Key laboratory of Translational Research and Therapy for Neuropsychiatric Disorders & Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| |
Collapse
|
8
|
Li X, Peng H, Wu J, Xu Y. Brain Natriuretic Peptide-Regulated Expression of Inflammatory Cytokines in Lipopolysaccharide (LPS)-Activated Macrophages via NF-κB and Mitogen Activated Protein Kinase (MAPK) Pathways. Med Sci Monit 2018; 24:3119-3126. [PMID: 29754152 PMCID: PMC5973487 DOI: 10.12659/msm.905580] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background This study aimed to investigate the effects of recombinant human brain natriuretic peptide (rhBNP) on IL-6, TNF-α, and IL-10 secretion in LPS-activated RAW 264.7 cells and human peripheral blood mononuclear cells (PBMCs) in vitro and to explore the related signaling pathways of the regulation mechanisms of BNP in systemic inflammatory response syndrome (SIRS). Material/Methods MTT assay was used to evaluate the effects of rhBNP on cell viabilities. Lipopolysaccharide (LPS) was used to induce inflammation response. The whole study was divided into 8 groups: Control, low, middle, and high concentrations of rhBNP, LPS, LPS with low, middle, and high concentrations of rhBNP. Levels of IL-6, TNF-α, and IL-10 were evaluated using the Cytometric Bead Array Kit and RT-PCR assay. Western blotting was used to test the effects of rhBNP on inflammation-related NF-κB and MAPK pathways. Results Except for the concentrations ≥1.6 ng/mL, all concentrations of rhBNP showed little effect on cell viabilities of RAW264.7 cells and PBMCs after 24 h and 48 h, suggesting a weak cytotoxicity to cells. Expression of IL-6 and TNF-α significantly increased and expression of IL-10 significantly decreased at protein and mRNA levels after LPS treatment, and these effects were strongly inhibited in a dose-dependent manner by pretreatment of rhBNP. Similarly, the LPS-induced increase of NF-κB and MAPK pathway phosphorylation levels were also significantly inhibited by rhBNP. Conclusions rhBNP can regulate expression of IL-6, TNF-α, and IL-10 in LPS-activated RAW 264.7 cells and PBMCs through inhibiting NF-κB and MAPK pathways. These results may reveal potential causes of the increase of BNP in SIRS and may provide an experimental basis for treatment of SIRS.
Collapse
Affiliation(s)
- Xiong Li
- Department of Burns, Third Xiangya Hospital, Central South University, Changsha, Hunan, China (mainland)
| | - Hao Peng
- Department of Burns, Third Xiangya Hospital, Central South University, Changsha, Hunan, China (mainland)
| | - Jiongxing Wu
- Department of Emergency Medicine, Xiangya Third Hospital, Central South University, Changsha, Hunan, China (mainland)
| | - Yangcheng Xu
- Department of Burns, Third Xiangya Hospital, Central South University, Changsha, Hunan, China (mainland)
| |
Collapse
|
9
|
Probucol inhibits LPS-induced microglia activation and ameliorates brain ischemic injury in normal and hyperlipidemic mice. Acta Pharmacol Sin 2016; 37:1031-44. [PMID: 27345627 DOI: 10.1038/aps.2016.51] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 03/21/2016] [Indexed: 12/20/2022] Open
Abstract
AIM Increasing evidence suggests that probucol, a lipid-lowering agent with anti-oxidant activities, may be useful for the treatment of ischemic stroke with hyperlipidemia via reduction in cholesterol and neuroinflammation. In this study we examined whether probucol could protect against brain ischemic injury via anti-neuroinflammatory action in normal and hyperlipidemic mice. METHODS Primary mouse microglia and murine BV2 microglia were exposed to lipopolysaccharide (LPS) for 3 h, and the release NO, PGE2, IL-1β and IL-6, as well as the changes in NF-κB, MAPK and AP-1 signaling pathways were assessed. ApoE KO mice were fed a high-fat diet containing 0.004%, 0.02%, 0.1% (wt/wt) probucol for 10 weeks, whereas normal C57BL/6J mice received probucol (3, 10, 30 mg·kg(-1)·d(-1), po) for 4 d. Then all the mice were subjected to focal cerebral ischemia through middle cerebral artery occlusion (MCAO). The neurological deficits were scored 24 h after the surgery, and then brains were removed for measuring the cerebral infarct size and the production of pro-inflammatory mediators. RESULTS In LPS-treated BV2 cells and primary microglial cells, pretreatment with probucol (1, 5, 10 μmol/L) dose-dependently inhibited the release of NO, PGE2, IL-1β and IL-6, which occurred at the transcription levels. Furthermore, the inhibitory actions of probucol were associated with the downregulation of the NF-κB, MAPK and AP-1 signaling pathways. In the normal mice with MCAO, pre-administration of probucol dose-dependently decreased the infarct volume and improved neurological function. These effects were accompanied by the decreased production of pro-inflammatory mediators (iNOS, COX-2, IL-1, IL-6). In ApoE KO mice fed a high-fat diet, pre-administration of 0.1% probucol significantly reduced the infarct volume, improved the neurological deficits following MCAO, and decreased the total- and LDL-cholesterol levels. CONCLUSION Probucol inhibits LPS-induced microglia activation and ameliorates cerebral ischemic injury in normal and hyperlipidemic mice via its anti-neuroinflammatory actions, suggesting that probucol has potential for the treatment of patients with or at risk for ischemic stroke and hyperlipidemia.
Collapse
|
10
|
Mahinrad S, de Craen AJM, Yasar S, van Heemst D, Sabayan B. Natriuretic peptides in the central nervous system: Novel targets for cognitive impairment. Neurosci Biobehav Rev 2016; 68:148-156. [PMID: 27229760 DOI: 10.1016/j.neubiorev.2016.05.022] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 04/22/2016] [Accepted: 05/22/2016] [Indexed: 02/06/2023]
Abstract
Natriuretic peptides (NPs) are traditionally known as cardiac hormones with diuretic, natriuretic and blood pressure lowering properties. Evidence indicates that NPs and their receptors are abundant in the central nervous system, suggesting their involvement in regulation of various brain functions. It has been shown that NPs are involved in the regulation of neurovascular and blood-brain barrier integrity, neuro-inflammation, neuroprotection, synaptic transmission and brain fluid homeostasis. In addition, NPs might contribute to the brain's inhibitory control over the hypothalamic-pituitary-adrenal axis. Studies have also shown that high systemic levels of NPs are associated with cognitive impairment independent of cardiovascular risk factors. In this review we discuss the potential roles of NPs in regulating structural and functional integrity of the brain. Based on the available neurobiological and clinical evidence, we propose that NPs might represent as potential novel diagnostic and therapeutic targets for cognitive impairment.
Collapse
Affiliation(s)
- Simin Mahinrad
- Department of Gerontology and Geriatrics, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, the Netherlands.
| | - Anton J M de Craen
- Department of Gerontology and Geriatrics, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, the Netherlands.
| | - Sevil Yasar
- Department of Medicine, Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, 5200 Eastern Avenue, MFL Center tower, Baltimore, MD 21224, United States.
| | - Diana van Heemst
- Department of Gerontology and Geriatrics, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, the Netherlands.
| | - Behnam Sabayan
- Department of Gerontology and Geriatrics, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, the Netherlands; Department of Radiology, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, the Netherlands.
| |
Collapse
|
11
|
Baliga RS, Scotton CJ, Trinder SL, Chambers RC, MacAllister RJ, Hobbs AJ. Intrinsic defence capacity and therapeutic potential of natriuretic peptides in pulmonary hypertension associated with lung fibrosis. Br J Pharmacol 2015; 171:3463-75. [PMID: 24641440 PMCID: PMC4105933 DOI: 10.1111/bph.12694] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 03/10/2014] [Accepted: 03/11/2014] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND AND PURPOSE Idiopathic pulmonary fibrosis (IPF) is a progressive fibro-proliferative disorder refractory to current therapy commonly complicated by the development of pulmonary hypertension (PH); the associated morbidity and mortality are substantial. Natriuretic peptides possess vasodilator and anti-fibrotic actions, and pharmacological augmentation of their bioactivity ameliorates renal and myocardial fibrosis. Here, we investigated whether natriuretic peptides possess an intrinsic cytoprotective function preventing the development of pulmonary fibrosis and associated PH, and whether therapeutics targeting natriuretic peptide signalling demonstrate efficacy in this life-threatening disorder. EXPERIMENTAL APPROACH Pulmonary haemodynamics, right ventricular function and markers of lung fibrosis were determined in wild-type (WT) and natriuretic peptide receptor (NPR)-A knockout (KO) mice exposed to bleomycin (1 mg·kg−1). Human myofibroblast differentiation was studied in vitro. KEY RESULTS Exacerbated cardiac, vascular and fibrotic pathology was observed in NPR-A KO animals, compared with WT mice, exposed to bleomycin. Treatment with a drug combination that raised circulating natriuretic peptide levels (ecadotril) and potentiated natriuretic peptide-dependent signalling (sildenafil) reduced indices of disease progression, whether administered prophylactically or to animals with established lung disease. This positive pharmacodynamic effect was diminished in NPR-A KO mice. Atrial natriuretic peptide and sildenafil synergistically reduced TGFβ-induced human myofibroblast differentiation, a key driver of remodelling in IPF patients. CONCLUSIONS AND IMPLICATIONS These data highlight an endogenous host-defence capacity of natriuretic peptides in lung fibrosis and PH. A combination of ecadotril and sildenafil reversed the pulmonary haemodynamic aberrations and remodelling that characterize the disease, advocating therapeutic manipulation of natriuretic peptide bioactivity in patients with IPF.
Collapse
Affiliation(s)
- R S Baliga
- William Harvey Research Institute, Barts & The London School of Medicine, Queen Mary University of London, Charterhouse Square, London, UK
| | | | | | | | | | | |
Collapse
|
12
|
Zhao J, Ha Y, Liou GI, Gonsalvez GB, Smith SB, Bollinger KE. Sigma receptor ligand, (+)-pentazocine, suppresses inflammatory responses of retinal microglia. Invest Ophthalmol Vis Sci 2014; 55:3375-84. [PMID: 24812552 PMCID: PMC4042630 DOI: 10.1167/iovs.13-12823] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2013] [Accepted: 04/29/2014] [Indexed: 02/05/2023] Open
Abstract
PURPOSE To evaluate the effects of the σ 1 receptor (σR1) agonist, (+)-pentazocine, on lipopolysaccharide (LPS)-induced inflammatory changes in retinal microglia cells. METHODS Retinal microglia cells were isolated from Sprague-Dawley rat pups. Cells were treated with LPS with or without (+)-pentazocine and with or without the σR1 antagonist BD1063. Morphologic changes were assayed. Cell viability was assessed by using MTT assay. Supernatant levels of tumor necrosis factor α (TNF-α), interleukin 10, (IL-10), monocyte chemoattractant protein-1 (MCP-1), and nitric oxide (NO) were determined. Reactive oxygen species (ROS) formation was assayed, and levels of mitogen-activated protein kinases (MAPKs) were analyzed by using Western blot. RESULTS The σR1 protein was expressed in retinal microglia. Incubation with LPS and/or (+)-pentazocine did not alter cell viability or σR1 protein levels. Incubation with LPS for 24 hours induced a marked change in microglial morphology and a significant increase in secreted levels of TNF-α, IL-10, MCP-1, and NO. Pretreatment with (+)-pentazocine inhibited the LPS-induced morphologic changes. Release of TNF-α, IL-10, MCP-1, and NO was reduced with (+)-pentazocine. Intracellular ROS formation was suppressed with (+)-pentazocine. Phosphorylation of extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK) was reduced in the presence of (+)-pentazocine. The σR1 antagonist BD1063 blocked the (+)-pentazocine-mediated inhibition of LPS-induced morphologic changes. In addition, BD1063 treatment blocked (+)-pentazocine-mediated suppression of LPS-induced TNF-α, IL-10, MCP-1, NO, and intracellular ROS release. CONCLUSIONS Treatment with (+)-pentazocine suppressed inflammatory responses of retinal microglia and inhibited LPS-induced activation of ERK/JNK MAPK. In neurodegenerative disease, (+)-pentazocine may exert neuroprotective effects through manipulation of microglia.
Collapse
Affiliation(s)
- Jing Zhao
- James and Jean Culver Vision Discovery Institute, Georgia Regents University, Augusta, Georgia, United States
| | - Yonju Ha
- James and Jean Culver Vision Discovery Institute, Georgia Regents University, Augusta, Georgia, United States
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, United States
| | - Gregory I. Liou
- James and Jean Culver Vision Discovery Institute, Georgia Regents University, Augusta, Georgia, United States
- Department of Ophthalmology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, United States
| | - Graydon B. Gonsalvez
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, United States
| | - Sylvia B. Smith
- James and Jean Culver Vision Discovery Institute, Georgia Regents University, Augusta, Georgia, United States
| | - Kathryn E. Bollinger
- James and Jean Culver Vision Discovery Institute, Georgia Regents University, Augusta, Georgia, United States
| |
Collapse
|
13
|
Shi Q, Cao J, Fang L, Zhao H, Liu Z, Ran J, Zheng X, Li X, Zhou Y, Ge D, Zhang H, Wang L, Ran Y, Fu J. Geniposide suppresses LPS-induced nitric oxide, PGE2 and inflammatory cytokine by downregulating NF-κB, MAPK and AP-1 signaling pathways in macrophages. Int Immunopharmacol 2014; 20:298-306. [PMID: 24735815 DOI: 10.1016/j.intimp.2014.04.004] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2013] [Revised: 03/25/2014] [Accepted: 04/01/2014] [Indexed: 11/29/2022]
Abstract
Inflammatory responses are important to host immune reactions, but uncontrolled inflammatory mediators may aid in the pathogenesis of other inflammatory diseases. Geniposide, an iridoid glycoside found in the herb gardenia, is believed to have broad-spectrum anti-inflammatory effects in murine models but its mechanism of action is unclear. We investigated the action of this compound in murine macrophages stimulated by lipopolysaccharide (LPS), as the stimulation of macrophages by LPS is known to induce inflammatory reactions. We determined the effect of geniposide on LPS-induced production of the inflammatory mediators, nitric oxide (NO) and prostaglandin E2 (PGE2), the mRNA and protein expression of the NO and PGE2 synthases, inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2), respectively, and the mRNA and protein expression of the inflammatory cytokine, tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6). Furthermore, nuclear factor (NF)-κB, mitogen-activated protein kinase (MAPK) and activator protein (AP)-1 activity were assayed. To understand the action of geniposide on the NF-κB and MAPK pathways, we studied the effect of NF-κB and MAPK inhibitors on the LPS-induced production of NO, PGE2 and TNF-α. Our findings clearly showed that geniposide mainly exerts its anti-inflammatory effects by inhibiting the LPS-induced NF-κB, MAPK and AP-1 signaling pathways in macrophages, which subsequently reduces overexpression of the inducible enzymes iNOS and COX-2 and suppresses the expression and release of the inflammatory factors, TNF-α, IL-6, NO and PGE2. Thus, geniposide shows promise as a therapeutic agent in inflammatory diseases.
Collapse
Affiliation(s)
- Qinghai Shi
- Clinical Laboratory Diagnostic Center, Urumqi General Hospital of Lanzhou Military Region, Urumqi 830000, Xinjiang, China.
| | - Jinjun Cao
- Clinical Laboratory Diagnostic Center, Urumqi General Hospital of Lanzhou Military Region, Urumqi 830000, Xinjiang, China
| | - Li Fang
- Second Section, Lintong Sanatorium of Lanzhou Military Region, Xi'an 710600, Shaanxi, China
| | - Hongyan Zhao
- Clinical Laboratory Diagnostic Center, Urumqi General Hospital of Lanzhou Military Region, Urumqi 830000, Xinjiang, China
| | - Zhengxiang Liu
- Clinical Laboratory Diagnostic Center, Urumqi General Hospital of Lanzhou Military Region, Urumqi 830000, Xinjiang, China
| | - Jihua Ran
- Clinical Laboratory Diagnostic Center, Urumqi General Hospital of Lanzhou Military Region, Urumqi 830000, Xinjiang, China
| | - Xinchuan Zheng
- Medical Research Center, Southwestern Hospital, Third Military Medical University, Chongqing 400038, China
| | - Xiaoling Li
- Clinical Laboratory Diagnostic Center, Urumqi General Hospital of Lanzhou Military Region, Urumqi 830000, Xinjiang, China
| | - Yu Zhou
- Clinical Laboratory Diagnostic Center, Urumqi General Hospital of Lanzhou Military Region, Urumqi 830000, Xinjiang, China
| | - Di Ge
- Clinical Laboratory Diagnostic Center, Urumqi General Hospital of Lanzhou Military Region, Urumqi 830000, Xinjiang, China
| | - Hongming Zhang
- Clinical Laboratory Diagnostic Center, Urumqi General Hospital of Lanzhou Military Region, Urumqi 830000, Xinjiang, China
| | - Li Wang
- Clinical Laboratory Diagnostic Center, Urumqi General Hospital of Lanzhou Military Region, Urumqi 830000, Xinjiang, China
| | - Ying Ran
- Clinical Laboratory Diagnostic Center, Urumqi General Hospital of Lanzhou Military Region, Urumqi 830000, Xinjiang, China
| | - Jianfeng Fu
- Clinical Laboratory Diagnostic Center, Urumqi General Hospital of Lanzhou Military Region, Urumqi 830000, Xinjiang, China.
| |
Collapse
|
14
|
Sun F, Zhou K, Wang SJ, Liang PF, Zhu MZ, Qiu JH. Expression patterns of atrial natriuretic peptide and its receptors within the cochlear spiral ganglion of the postnatal rat. Hear Res 2013; 309:103-12. [PMID: 24333928 DOI: 10.1016/j.heares.2013.11.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Revised: 10/31/2013] [Accepted: 11/28/2013] [Indexed: 10/25/2022]
Abstract
The spiral ganglion, which is primarily composed of spiral ganglion neurons and satellite glial cells, transmits auditory information from sensory hair cells to the central nervous system. Atrial natriuretic peptide (ANP), acting through specific receptors, is a regulatory peptide required for a variety of cardiac, neuronal and glial functions. Although previous studies have provided direct evidence for the presence of ANP and its functional receptors (NPR-A and NPR-C) in the inner ear, their presence within the cochlear spiral ganglion and their regulatory roles during auditory neurotransmission and development is not known. Here we investigated the expression patterns and levels of ANP and its receptors within the cochlear spiral ganglion of the postnatal rat using immunofluorescence and immunoelectron microscopy techniques, reverse transcription-polymerase chain reaction and Western blot analysis. We have demonstrated that ANP and its receptors colocalize in both subtypes of spiral ganglion neurons and in perineuronal satellite glial cells. Furthermore, we have analyzed differential expression levels associated with both mRNA and protein of ANP and its receptors within the rat spiral ganglion during postnatal development. Collectively, our research provides direct evidence for the presence and synthesis of ANP and its receptors in both neuronal and non-neuronal cells within the cochlear spiral ganglion, suggesting possible roles for ANP in modulating neuronal and glial functions, as well as neuron-satellite glial cell communication, within the spiral ganglion during auditory neurotransmission and development.
Collapse
Affiliation(s)
- Fei Sun
- Department of Otolaryngology/Head and Neck Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province 710032, China; Department of Physiology, School of Basic Medical Sciences, Fourth Military Medical University, Xi'an, Shaanxi Province 710032, China
| | - Ke Zhou
- Department of Otolaryngology/Head and Neck Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province 710032, China; Center of Clinical Laboratory Medicine of PLA, Department of Clinical Laboratory, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province 710032, China
| | - Shu-juan Wang
- Department of Otolaryngology/Head and Neck Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province 710032, China
| | - Peng-fei Liang
- Department of Otolaryngology/Head and Neck Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province 710032, China
| | - Miao-zhang Zhu
- Department of Physiology, School of Basic Medical Sciences, Fourth Military Medical University, Xi'an, Shaanxi Province 710032, China.
| | - Jian-hua Qiu
- Department of Otolaryngology/Head and Neck Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province 710032, China.
| |
Collapse
|
15
|
Fujita S, Shimojo N, Terasaki F, Otsuka K, Hosotani N, Kohda Y, Tanaka T, Nishioka T, Yoshida T, Hiroe M, Kitaura Y, Ishizaka N, Imanaka-Yoshida K. Atrial natriuretic peptide exerts protective action against angiotensin II-induced cardiac remodeling by attenuating inflammation via endothelin-1/endothelin receptor A cascade. Heart Vessels 2013; 28:646-57. [PMID: 23277455 DOI: 10.1007/s00380-012-0311-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Accepted: 11/30/2012] [Indexed: 01/17/2023]
Abstract
We aimed to investigate whether atrial natriuretic peptide (ANP) attenuates angiotensin II (Ang II)-induced myocardial remodeling and to clarify the possible molecular mechanisms involved. Thirty-five 8-week-old male Wistar-Kyoto rats were divided into control, Ang II, Ang II + ANP, and ANP groups. The Ang II and Ang II + ANP rats received 1 μg/kg/min Ang II for 14 days. The Ang II + ANP and ANP rats also received 0.1 μg/kg/min ANP intravenously. The Ang II and Ang II + ANP rats showed comparable blood pressure. Left ventricular fractional shortening and ejection fraction were lower in the Ang II rats than in controls; these indices were higher (P < 0.001) in the Ang II + ANP rats than in the Ang II rats. In the Ang II rats, the peak velocity of mitral early inflow and its ratio to atrial contraction-related peak flow velocity were lower, and the deceleration time of mitral early inflow was significantly prolonged; these changes were decreased by ANP. Percent fibrosis was higher (P < 0.001) and average myocyte diameters greater (P < 0.01) in the Ang II rats than in controls. ANP decreased both myocardial fibrosis (P < 0.01) and myocyte hypertrophy (P < 0.01). Macrophage infiltration, expression of mRNA levels of collagen types I and III, monocyte chemotactic protein-1, and a profibrotic/proinflammatory molecule, tenascin-C (TN-C) were increased in the Ang II rats; ANP significantly decreased these changes. In vitro, Ang II increased expression of TN-C and endothelin-1 (ET-1) in cardiac fibroblasts, which were reduced by ANP. ET-1 upregulated TN-C expression via endothelin type A receptor. These results suggest that ANP may protect the heart from Ang II-induced remodeling by attenuating inflammation, at least partly through endothelin 1/endothelin receptor A cascade.
Collapse
Affiliation(s)
- Shuichi Fujita
- Department of Cardiology, Osaka Medical College, 2-7 Daigaku-machi, Takatsuki, Osaka, 569-8686, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Metallothioneins I/II are involved in the neuroprotective effect of sildenafil in focal brain injury. Neurochem Int 2012. [PMID: 23178801 DOI: 10.1016/j.neuint.2012.11.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We recently reported that administration of the non-selective cyclic GMP-phosphodiesterase (cGMP-PDE) inhibitor zaprinast to cortically cryoinjured rats results three days post-lesion in reduced neuronal cell death that was associated to decreased macrophage/microglial activation and oxidative stress and increased astrogliosis and angiogenesis. Similar effects have been observed in cryoinjured animals overexpressing metallothioneins I/II (MT-I/II), metal-binding cysteine-rich proteins that are up-regulated in response to injury. In this work we have examined the effect of administration of the selective PDE5 inhibitor sildenafil (10mg/kg, sc) 2h before and 24 and 48h after induction of cortical cryolesion in wild-type and MT-I/II-deficient mice. Our results show that in wild-type animals sildenafil induces similar changes in glial reactivity, angiogenesis and antioxidant and antiapoptotic effects in the cryolesioned cortex as those observed in rats with zaprinast, indicating that inhibition of PDE5 is responsible for the neuroprotective actions. However, these effects were not observed in mice deficient in MT-I/II. We further show that sildenafil significantly increases MT-I/II protein levels in homogenates of lesioned cortex and MT-I/II immunostaining in glial cells around the lesion. Taken together these results indicate that cGMP-mediated pathways regulate expression of MT-I/II and support the involvement of these proteins in the neuroprotective effects of sildenafil in focal brain lesion.
Collapse
|
17
|
Ding JH, Chang YS. Atrial natriuretic peptide: a possible mediator involved in dexamethasone's inhibition of cell proliferation in multiple myeloma. Med Hypotheses 2012; 79:207-9. [PMID: 22595807 DOI: 10.1016/j.mehy.2012.04.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Revised: 04/17/2012] [Accepted: 04/22/2012] [Indexed: 11/26/2022]
Abstract
Atrial natriuretic peptide (ANP) has been recognized for several decades for its role of regulating blood pressure. Recently, cumulating evidences show that ANP plays an anticancer role in various solid tumors via blocking the kinase cascade of Ras-MEK1/2-ERK1/2 with the result of inhibition of DNA synthesis. ANP, as well as its receptors (NPR-A and NPR-C) has been identified present in the embryonic stem cell and a wide range of cancer cells. Various lymphoid organs, such as lymph nodes, have been detected the presence of ANP. Multiple myeloma (MM), though the therapies have evolved significantly, is still an incurable disease as B lymphocyte cell neoplasm. Dexamethasone is the cornerstone in treatment of MM via inactivation of Ras-MEK1/2-ERK1/2 cascade reaction. Coincidently, dexamethasone can increase the expression of ANP markedly. Nevertheless, the role of ANP in MM is unclear. Based on these results above, we raise the hypothesis that ANP is involved in mediating dexamethasone's inhibition of proliferation in MM cells, which suggests that ANP may be a potential agent to treat MM.
Collapse
Affiliation(s)
- Jiang-Hua Ding
- Medical School of Nanchang University, Nanchang 330006, China.
| | | |
Collapse
|
18
|
Hong J, Kim BK, Lim H, Lee S, Lee SJ. Identification and characterization of triamcinolone acetonide, a microglial-activation inhibitor. Immunopharmacol Immunotoxicol 2012; 34:912-8. [PMID: 22551518 DOI: 10.3109/08923973.2012.671332] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Recent studies show that necrotic neuronal cells (NNC) activate microglia, thereby leading to neuronal cell death. This suggests that chemicals that inhibit microglia activation may be used as neuroprotective drugs. In this context, we screened a chemical library for inhibitors of microglia activation. Using a screening system based on a nitrite assay, we isolated two chemicals that inhibit nitric oxide (NO) release from activated microglia: triamcinolone acetonide (TA) and amcinonide. The half-maximal inhibitory concentrations (IC50) of TA and amcinonide for NO release inhibition were 1.78 nM and 3.38 nM, respectively. These chemicals also inhibited NNC-induced expression of the proinflammatory genes iNOS, TNF-α, and IL-1β in glial cells. A study based on a luciferase assay revealed that TA attenuated NNC-induced microglia activation by blocking the NF-κB signaling pathway. In addition, TA protected cortical neurons in coculture with microglia from LPS/IFN-γ-induced neuronal cell death. In conclusion, TA may inhibit microglia activation and may protect neuronal cells from death induced by microglial activation.
Collapse
Affiliation(s)
- Jinpyo Hong
- Department of Neuroscience, Dental Research Institute, and Brain Korea 21, School of Dentistry, Seoul National University, Seoul, Republic of Korea
| | | | | | | | | |
Collapse
|
19
|
Pifarre P, Prado J, Baltrons MA, Giralt M, Gabarro P, Feinstein DL, Hidalgo J, Garcia A. Sildenafil (Viagra) ameliorates clinical symptoms and neuropathology in a mouse model of multiple sclerosis. Acta Neuropathol 2011; 121:499-508. [PMID: 21234581 DOI: 10.1007/s00401-010-0795-6] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Revised: 12/21/2010] [Accepted: 12/30/2010] [Indexed: 12/14/2022]
Abstract
Cyclic GMP (cGMP)-mediated pathways regulate inflammatory responses in immune and CNS cells. Recently, cGMP phosphodiesterase inhibitors such as sildenafil, commonly used to treat sexual dysfunction in humans including multiple sclerosis (MS) patients, have been reported to be neuroprotective in animal models of stroke, Alzheimer's disease, and focal brain lesion. In this work, we have examined if sildenafil ameliorates myelin oligodendrocyte glycoprotein peptide (MOG₃₅₋₅₅)-induced experimental autoimmune encephalomyelitis (EAE), a mouse model of MS. We show for the first time that treatment with sildenafil after disease onset markedly reduces the clinical signs of EAE by preventing axonal loss and promoting remyelination. Furthermore, sildenafil decreases CD3+ leukocyte infiltration and microglial/macrophage activation in the spinal cord, while increasing forkhead box transcription factor 3-expressing T regulatory cells (Foxp3 Tregs). However, sildenafil treatment did not significantly affect MOG₃₅₋₅₅-stimulated proliferation or release of Th1/Th2 cytokines in splenocytes but decreased ICAM-1 in spinal cord infiltrated cells. The presence of reactive astrocytes forming scar-like structures around infiltrates was enhanced by sildenafil suggesting a possible mechanism for restriction of leukocyte spread into healthy parenchyma. These results highlight novel actions of sildenafil that may contribute to its beneficial effects in EAE and suggest that treatment with this widely used and well-tolerated drug may be a useful therapeutic intervention to ameliorate MS neuropathology.
Collapse
Affiliation(s)
- Paula Pifarre
- Institute of Biotechnology and Biomedicine, Universitat Autonoma de Barcelona, 08193 Bellaterra, Spain
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Byun JS, Lee JW, Kim SY, Kwon KJ, Sohn JH, Lee K, Oh D, Kim SS, Chun W, Lee HJ. Neuroprotective effects of stanniocalcin 2 following kainic acid-induced hippocampal degeneration in ICR mice. Peptides 2010; 31:2094-9. [PMID: 20713105 DOI: 10.1016/j.peptides.2010.08.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2010] [Revised: 08/04/2010] [Accepted: 08/08/2010] [Indexed: 02/06/2023]
Abstract
Stanniocalcin 2 (STC2), the paralog of STC1, has been shown to act as a novel target of the mammalian unfolded protein response. We investigated the potential neuroprotective actions of STC2 against kainic acid toxicity in the hippocampus of ICR mice. STC2-treated mice experienced less neuronal cell loss in the CA3 area of the hippocampus. Also, microglial activation and heme oxygenase 1 expression were attenuated in the hippocampus of STC2-treated mice. To confirm whether STC2 regulates microglial activation directly, nitric oxide levels were measured in BV2 cells cultured with and without 10nM STC2. STC2 decreased the level of nitric oxide induced by lipopolysaccharide (LPS) treatment significantly. Also, STC2 pretreatment significantly decreased TNF-α and IL-1β expression induced by LPS treatment. These observations demonstrated that STC2 exerts neuroprotective actions against excitotoxic insults through the inhibition of microglial activation.
Collapse
Affiliation(s)
- Jong-Seon Byun
- Department of Pharmacology, School of Medicine, Kangwon National University, Chuncheon 200-701, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Prado J, Baltrons MA, Pifarré P, García A. Glial cells as sources and targets of natriuretic peptides. Neurochem Int 2010; 57:367-74. [PMID: 20302900 DOI: 10.1016/j.neuint.2010.03.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2010] [Accepted: 03/10/2010] [Indexed: 11/19/2022]
Abstract
Natriuretic peptides and their receptors are widely expressed in mammalian CNS and increasing evidence implicates them in the regulation of neural development, synaptic transmission and processing of information, and neuroprotection. Although the peptides have been mainly localized in neuronal populations they are also produced in glial cells. Astroglia and microglia also express functional natriuretic peptide receptors that can regulate important physiological responses. In this article we review evidence on the localization of natriuretic peptides and their receptors in astroglial and microglial cells and summarize data supporting the participation of this signalling system in neuron-glia and glia-brain blood vessel communication relevant to CNS function.
Collapse
Affiliation(s)
- Judith Prado
- Institut de Biotecnologia i de Biomedicina, Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Bellaterra (Cerdanyola del Vallès), Barcelona, Spain
| | | | | | | |
Collapse
|
22
|
Zhang F, Qian L, Flood PM, Shi JS, Hong JS, Gao HM. Inhibition of IkappaB kinase-beta protects dopamine neurons against lipopolysaccharide-induced neurotoxicity. J Pharmacol Exp Ther 2010; 333:822-33. [PMID: 20190013 DOI: 10.1124/jpet.110.165829] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Parkinson's disease (PD) is a progressive neurological disorder characterized by a selective loss of dopamine (DA) neurons in the substantia nigra (SN). Although current therapy can control symptoms of this disorder, there is no effective therapy available to halt its progression. Recently, neuroinflammation has been recognized as an important contributor to the pathogenesis of PD, and nuclear factor-kappaB (NF-kappaB) plays a key role in regulating neuroinflammation. Hence, the modulation of NF-kappaB pathway may have therapeutic potential for PD. Activation of NF-kappaB depends on the phosphorylation of its inhibitor, IkappaB, by the specific IkappaB kinase (IKK) subunit IKK-beta. Compound A (7-[2-(cyclopropylmethoxy)-6-hydroxyphenyl]-5-[(3S)-3-piperidinyl]-1, 4-dihydro-2H-pyrido[2,3-d][1,3]oxazin-2-one hydrochloride), a potent and selective inhibitor of IKK-beta, has recently been reported to provide cardioprotection through specific suppression of NF-kappaB signaling. The present study, for the first time, elucidates neuroprotective effects of compound A. Daily subcutaneous injection of compound A (1 mg/kg) for 7 days inhibited the activation of microglia induced by nigral stereotaxic injection of lipopolysaccharide (LPS) and significantly attenuated LPS-induced loss of DA neurons in the SN. In vitro mechanistic studies revealed that neuroprotective effects of compound A were mediated by 1) suppressing the activity of microglial NADPH oxidase and decreasing the production of reactive oxygen species, and 2) inhibiting NF-kappaB-mediated gene transcription of various proinflammatory mediators in microglia via IKK-beta suppression. These findings indicate that compound A afforded potent neuroprotection against LPS-induced neurodegeneration through selective inhibition of NF-kappaB activation and may be of potential benefit in the treatment of PD.
Collapse
Affiliation(s)
- Feng Zhang
- Neuropharmacology Section, Laboratory of Toxicology and Pharmacology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709, USA
| | | | | | | | | | | |
Collapse
|
23
|
Pifarré P, Prado J, Giralt M, Molinero A, Hidalgo J, Garcia A. Cyclic GMP phosphodiesterase inhibition alters the glial inflammatory response, reduces oxidative stress and cell death and increases angiogenesis following focal brain injury. J Neurochem 2009; 112:807-17. [PMID: 20002517 DOI: 10.1111/j.1471-4159.2009.06518.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Recent evidence obtained in cultured glial cells indicates that cGMP-mediated pathways regulate cytoskeleton dynamics, glial fibrillary acidic protein expression and motility in astrocytes, as well as inflammatory gene expression in microglia, suggesting a role in the regulation of the glial reactive phenotype. The aim of this work was to examine if cGMP regulates the glial inflammatory response in vivo following CNS damage caused by a focal cryolesion onto the cortex in rats. Results show that treatment with the cGMP phosphodiesterase inhibitor zaprinast (10 mg/kg i.p.) 2 h before and 24 and 48 h after the lesion results 3 days post-lesion in notably enhanced astrogliosis manifested by increased glial fibrillary acidic protein immunoreactivity and protein levels around the lesion. In contrast, zaprinast decreased the number of round/ameboid lectin-positive cells and the expression of the activated microglia/macrophage markers Iba-1 and CD11b indicating decreased recruitment and activation of these cells. This altered inflammatory response is accompanied by a decrease in protein oxidative stress, apoptotic cell death and neuronal degeneration. In addition, zaprinast enhanced angiogenesis in the lesioned cortex probably as a result of vascular endothelial growth factor expression in reactive astrocytes. These results suggest that regulation of the glial inflammatory response may contribute to the reported neuroprotective effects of cGMP-phosphodiesterase inhibitors in brain injury.
Collapse
Affiliation(s)
- Paula Pifarré
- Institute of Biotechnology and Biomedicine and Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | | | | | | | | | | |
Collapse
|
24
|
Natriuretic peptide/natriuretic peptide receptor-A (NPR-A) system has inhibitory effects in renal fibrosis in mice. ACTA ACUST UNITED AC 2009; 154:44-53. [PMID: 19223006 DOI: 10.1016/j.regpep.2009.02.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2008] [Revised: 12/11/2008] [Accepted: 02/03/2009] [Indexed: 11/21/2022]
Abstract
OBJECT This study was designed to examine whether natriuretic peptide/natriuretic peptide receptor-A (NPR-A) system attenuates renal fibrosis in a unilateral ureteral obstruction (UUO) model and also examined the mechanism involved. METHODS Three groups were studied: untreated UUO in wild-type mice; untreated UUO in NPR-A KO mice; and ANP treated (0.05 microg/kg/min) UUO in wild-type mice. We measured histological and immunohistochemical findings (alpha-SMA and F4/80), tissue cGMP levels, various mRNA expression levels by real-time PCR analysis, and transcription factor levels (AP-1 and NF-kappaB) in renal tissue. RESULTS Compared with wild-type UUO mice, NPRA-KO UUO mice had abnormal morphological findings (fibrous area: +26%, alpha-SMA expression: +30%) with lower tissue cGMP levels and increases in the mRNA expression levels of TGF-beta, collagen I, collagen III, PAI-1, renin and angiotensinogen, whereas there were no differences in F4/80 positive cells or the mRNA expression levels of ICAM-1, osteopontin, or MCP-1 between the two groups. In contrast, ANP pre-treatment significantly improved morphological changes with increase of tissue cGMP levels and reduction in the mRNA expression level of TGF-beta, collagen I, collagen III, PAI-1, ICAM-1, osteopontin, MCP-1, renin, and angiotensinogen. NPRA-KO UUO mice had higher AP-1 levels than wild-type UUO mice and ANP pre-treatment reduced AP-1 and NF-kappaB activity. CONCLUSION The endogenous natriuretic peptide/NPR-A system may inhibit renal fibrosis partly via inhibition of the angiotensin/AP-1/TGF-beta/collagen pathway and exogenous ANP pre-treatment may inhibit it partly via both the angiotensin/AP-1/TGF-beta/collagen and NF-kappaB/inflammatory pathways.
Collapse
|
25
|
NPRA-mediated suppression of AngII-induced ROS production contribute to the antiproliferative effects of B-type natriuretic peptide in VSMC. Mol Cell Biochem 2008; 324:165-72. [PMID: 19104909 DOI: 10.1007/s11010-008-9995-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2008] [Accepted: 12/11/2008] [Indexed: 12/31/2022]
Abstract
Excessive proliferation of vascular smooth cells (VSMCs) plays a critical role in the pathogenesis of diverse vascular disorders, and inhibition of VSMCs proliferation has been proved to be beneficial to these diseases. In this study, we investigated the antiproliferative effect of B-type natriuretic peptide (BNP), a natriuretic peptide with potent antioxidant capacity, on rat aortic VSMCs, and the possible mechanisms involved. The results indicate that BNP potently inhibited AngiotensinII (AngII)-induced VSMCs proliferation, as evaluated by [(3)H]-thymidine incorporation assay. Consistently, BNP significantly decreased AngII-induced intracellular reactive oxygen species (ROS) and NAD(P)H oxidase activity. 8-Br-cGMP, a cGMP analog, mimicked these effects. To confirm its mechanism, siRNA of natriuretic peptide receptor-A(NRPA) strategy technology was used to block cGMP production in VSMCs, and siNPRA attenuated the inhibitory effects of BNP in VSMCs. Taken together, these results indicate that BNP was capable of inhibiting VSMCs proliferation by NPRA/cGMP pathway, which might be associated with the suppression of ROS production. These results might be related, at least partly, to the anti-oxidant property of BNP.
Collapse
|
26
|
Cho IH, Hong J, Suh EC, Kim JH, Lee H, Lee JE, Lee S, Kim CH, Kim DW, Jo EK, Lee KE, Karin M, Lee SJ. Role of microglial IKKbeta in kainic acid-induced hippocampal neuronal cell death. ACTA ACUST UNITED AC 2008; 131:3019-33. [PMID: 18819987 DOI: 10.1093/brain/awn230] [Citation(s) in RCA: 136] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Microglial cells are activated during excitotoxin-induced neurodegeneration. However, the in vivo role of microglia activation in neurodegeneration has not yet been fully elucidated. To this end, we used Ikkbeta conditional knockout mice (LysM-Cre/Ikkbeta(F/F)) in which the Ikkbeta gene is specifically deleted in cells of myeloid lineage, including microglia, in the CNS. This deletion reduced IkappaB kinase (IKK) activity in cultured primary microglia by up to 40% compared with wild-type (Ikkbeta(F/F)), and lipopolysaccharide-induced proinflammatory gene expression was also compromised. Kainic acid (KA)-induced hippocampal neuronal cell death was reduced by 30% in LysM-Cre/Ikkbeta(F/F) mice compared with wild-type mice. Reduced neuronal cell death was accompanied by decreased KA-induced glial cell activation and subsequent expression of proinflammatory genes such as tumour necrosis factor (TNF)-alpha and interleukin (IL)-1beta. Similarly, neurons in organotypic hippocampal slice cultures (OHSCs) from LysM-Cre/Ikkbeta(F/F) mouse brain were less susceptible to KA-induced excitotoxicity compared with wild-type OHSCs, due in part to decreased TNF-alpha and IL-1beta expression. Based on these data, we concluded that IKK/nuclear factor-kappaB dependent microglia activation contributes to KA-induced hippocampal neuronal cell death in vivo through induction of inflammatory mediators.
Collapse
Affiliation(s)
- Ik-Hyun Cho
- Program in Molecular and Cellular Neuroscience, DRI, and Department of Oral Physiology, School of Dentistry, Seoul National University, Seoul, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Borán MS, Baltrons MA, García A. The ANP-cGMP-protein kinase G pathway induces a phagocytic phenotype but decreases inflammatory gene expression in microglial cells. Glia 2008; 56:394-411. [PMID: 18186083 DOI: 10.1002/glia.20618] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Reactive gliosis is a prominent feature of CNS injury that involves dramatic changes in glial cell morphology together with increased motility, phagocytic activity, and release of inflammatory mediators. We have recently demonstrated that stimulation of the cGMP-protein kinase G (PKG) pathway by NO or atrial natriuretic peptide (ANP) regulates cytoskeleton dynamics and motility in rat astrocytes in culture. In this work, we show that the cGMP-PKG pathway stimulated by ANP, but not by NO, regulates microglial cell morphology by inducing a dramatic reorganization in the actin cytoskeleton. Both ANP (0.01-1.0 microM) and the permeable cGMP analog, dibutyryl-cGMP (1-100 microM), promote a rapid (maximal at 30 min) and concentration-dependent increase in size, rounding, and lamellipodia and filopodia formation in rat brain cultured microglia. These morphological changes involve an augment and redistribution of F-actin and result in increased phagocytic activity. ANP-induced rearrangements in actin cytoskeleton and inert particle phagocytosis are prevented by the PKG inhibitor, Rp-8-Br-PET-cGMPS (0.5 microM), and involve inhibition of RhoA GTPase and activation of Rac1 and Cdc42. However, ANP does not induce NO synthase Type 2 (NOS-2) or tumor necrosis factor-alpha expression and is able to decrease lipopolysaccharide (LPS)-elicited induction of these inflammatory genes. The morphological changes and the decrease of LPS-induced NOS-2 expression produced by ANP in cultured microglia are also observed by immunostaining in organotypic cultures from rat hippocampus. These results suggest that stimulation of the ANP-cGMP-PKG pathway in microglia could play a beneficial role in the resolution of neuroinflammation by removing dead cells and decreasing levels of proinflammatory mediators.
Collapse
Affiliation(s)
- Mariela Susana Borán
- Institute of Biotechnology and Biomedicine, Universidad Autónoma de Barcelona, Bellaterra, Barcelona, Spain
| | | | | |
Collapse
|
28
|
Baltrons MA, Borán MS, Pifarré P, García A. Regulation and function of cyclic GMP-mediated pathways in glial cells. Neurochem Res 2008; 33:2427-35. [PMID: 18379874 DOI: 10.1007/s11064-008-9681-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2008] [Accepted: 03/14/2008] [Indexed: 01/21/2023]
Abstract
A large body of evidence supports a role for the NO-cGMP-protein kinase G pathway in the regulation of synaptic transmission and plasticity, brain development and neuroprotection. Circumstantial evidence implicates natriuretic peptide-stimulated cGMP formation in the same CNS functions. In addition to neurons, both cGMP-mediated pathways are functional in glial cells and an increasing number of reports indicate that they may control important aspects of glial cell physiology relevant to neuronal function. In this article we briefly review the regulation of cGMP formation in glial cells and summarize recent evidence indicating that cGMP-mediated pathways can play important roles in astroglial and microglial function in normal and diseased brain.
Collapse
Affiliation(s)
- María Antonia Baltrons
- Institute of Biotechnology and Biomedicine V. Villar Palasí and Departament of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| | | | | | | |
Collapse
|