1
|
Xuan L, Hu Z, Jiang Z, Zhang C, Sun X, Ming W, Liu H, Qiao R, Shen L, Liu S, Wang G, Wen L, Luan Z, Yin J. Pregnane X receptor (PXR) deficiency protects against spinal cord injury by activating NRF2/HO-1 pathway. CNS Neurosci Ther 2023; 29:3460-3478. [PMID: 37269088 PMCID: PMC10580351 DOI: 10.1111/cns.14279] [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: 10/28/2022] [Revised: 05/12/2023] [Accepted: 05/14/2023] [Indexed: 06/04/2023] Open
Abstract
INTRODUCTION As a devastating neurological disease, spinal cord injury (SCI) results in severe tissue loss and neurological dysfunction. Pregnane X receptor (PXR) is a ligand-activated nuclear receptor with a major regulatory role in xenobiotic and endobiotic metabolism and recently has been implicated in the central nervous system. In the present study, we aimed to investigate the role and mechanism of PXR in SCI. METHODS The clip-compressive SCI model was performed in male wild-type C57BL/6 (PXR+/+ ) and PXR-knockout (PXR-/- ) mice. The N2a H2 O2 -induced injury model mimicked the pathological process of SCI in vitro. Pregnenolone 16α-carbonitrile (PCN), a mouse-specific PXR agonist, was used to activate PXR in vivo and in vitro. The siRNA was applied to knock down the PXR expression in vitro. Transcriptome sequencing analysis was performed to discover the relevant mechanism, and the NRF2 inhibitor ML385 was used to validate the involvement of PXR in influencing the NRF2/HO-1 pathway in the SCI process. RESULTS The expression of PXR decreased after SCI and reached a minimum on the third day. In vivo, PXR knockout significantly improved the motor function of mice after SCI, meanwhile, inhibited apoptosis, inflammation, and oxidative stress induced by SCI. On the contrary, activation of PXR by PCN negatively influenced the recovery of SCI. Mechanistically, transcriptome sequencing analysis revealed that PXR activation downregulated the mRNA level of heme oxygenase-1 (HO-1) after SCI. We further verified that PXR deficiency activated the NRF2/HO-1 pathway and PXR activation inhibited this pathway in vitro. CONCLUSION PXR is involved in the recovery of motor function after SCI by regulating NRF2/HO-1 pathway.
Collapse
Affiliation(s)
- Li‐Na Xuan
- Department of Neurosurgerythe Second Affiliated Hospital of Dalian Medical UniversityDalianChina
- Epileptic Center of Liaoningthe Second Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Zhen‐Xin Hu
- Department of OrthopedicsThe First Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Zhen‐Fu Jiang
- Department of Neurosurgerythe Second Affiliated Hospital of Dalian Medical UniversityDalianChina
- Epileptic Center of Liaoningthe Second Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Cong Zhang
- Advanced Institute for Medical SciencesDalian Medical UniversityDalianChina
| | - Xiao‐Wan Sun
- Advanced Institute for Medical SciencesDalian Medical UniversityDalianChina
| | - Wen‐Hua Ming
- Advanced Institute for Medical SciencesDalian Medical UniversityDalianChina
| | - Hui‐Tao Liu
- Department of OrthopedicsTaizhou Hospital of Zhejiang ProvinceLinhaiChina
| | - Rong‐Fang Qiao
- Advanced Institute for Medical SciencesDalian Medical UniversityDalianChina
| | - Lin‐Jie Shen
- Department of GastroenterologyNingbo First HospitalNingboChina
| | - Shao‐Bo Liu
- Department of Neurosurgerythe Second Affiliated Hospital of Dalian Medical UniversityDalianChina
- Epileptic Center of Liaoningthe Second Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Guan‐Yu Wang
- Department of Neurosurgerythe Second Affiliated Hospital of Dalian Medical UniversityDalianChina
- Epileptic Center of Liaoningthe Second Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Lin Wen
- Advanced Institute for Medical SciencesDalian Medical UniversityDalianChina
| | - Zhi‐Lin Luan
- Advanced Institute for Medical SciencesDalian Medical UniversityDalianChina
- Dalian Key Laboratory for Nuclear Receptors in Major Metabolic DiseasesDalianChina
| | - Jian Yin
- Department of Neurosurgerythe Second Affiliated Hospital of Dalian Medical UniversityDalianChina
- Epileptic Center of Liaoningthe Second Affiliated Hospital of Dalian Medical UniversityDalianChina
| |
Collapse
|
2
|
Tauroursodeoxycholic Acid Reduces Neuroinflammation but Does Not Support Long Term Functional Recovery of Rats with Spinal Cord Injury. Biomedicines 2022; 10:biomedicines10071501. [PMID: 35884805 PMCID: PMC9313003 DOI: 10.3390/biomedicines10071501] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 06/18/2022] [Accepted: 06/23/2022] [Indexed: 11/17/2022] Open
Abstract
The bile acid tauroursodeoxycholic acid (TUDCA) reduces cell death under oxidative stress and inflammation. Implants of bone marrow-derived stromal cells (bmSC) are currently under investigation in clinical trials of spinal cord injury (SCI). Since cell death of injected bmSC limits the efficacy of this treatment, the cytoprotective effect of TUDCA may enhance its benefit. We therefore studied the therapeutic effect of TUDCA and its use as a combinatorial treatment with human bmSC in a rat model of SCI. A spinal cord contusion injury was induced at thoracic level T9. Treatment consisted of i.p. injections of TUDCA alone or in combination with one injection of human bmSC into the cisterna magna. The recovery of motor functions was assessed during a surveillance period of six weeks. Biochemical and histological analysis of spinal cord tissue confirmed the anti-inflammatory activity of TUDCA. Treatment improved the recovery of autonomic bladder control and had a positive effect on motor functions in the subacute phase, however, benefits were only transient, such that no significant differences between vehicle and TUDCA-treated animals were observed 1–6 weeks after the lesion. Combinatorial treatment with TUDCA and bmSC failed to have an additional effect compared to treatment with bmSC only. Our data do not support the use of TUDCA as a treatment of SCI.
Collapse
|
3
|
Raas Q, van de Beek MC, Forss-Petter S, Dijkstra IM, Deschiffart A, Freshner BC, Stevenson TJ, Jaspers YR, Nagtzaam L, Wanders RJ, van Weeghel M, Engelen-Lee JY, Engelen M, Eichler F, Berger J, Bonkowsky JL, Kemp S. Metabolic rerouting via SCD1 induction impacts X-linked adrenoleukodystrophy. J Clin Invest 2021; 131:142500. [PMID: 33690217 DOI: 10.1172/jci142500] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 03/03/2021] [Indexed: 12/18/2022] Open
Abstract
X-linked adrenoleukodystrophy (ALD) is a progressive neurodegenerative disease caused by mutations in ABCD1, the peroxisomal very long-chain fatty acid (VLCFA) transporter. ABCD1 deficiency results in accumulation of saturated VLCFAs. A drug screen using a phenotypic motor assay in a zebrafish ALD model identified chloroquine as the top hit. Chloroquine increased expression of stearoyl-CoA desaturase-1 (scd1), the enzyme mediating fatty acid saturation status, suggesting that a shift toward monounsaturated fatty acids relieved toxicity. In human ALD fibroblasts, chloroquine also increased SCD1 levels and reduced saturated VLCFAs. Conversely, pharmacological inhibition of SCD1 expression led to an increase in saturated VLCFAs, and CRISPR knockout of scd1 in zebrafish mimicked the motor phenotype of ALD zebrafish. Importantly, saturated VLCFAs caused ER stress in ALD fibroblasts, whereas monounsaturated VLCFA did not. In parallel, we used liver X receptor (LXR) agonists to increase SCD1 expression, causing a shift from saturated toward monounsaturated VLCFA and normalizing phospholipid profiles. Finally, Abcd1-/y mice receiving LXR agonist in their diet had VLCFA reductions in ALD-relevant tissues. These results suggest that metabolic rerouting of saturated to monounsaturated VLCFAs may alleviate lipid toxicity, a strategy that may be beneficial in ALD and other peroxisomal diseases in which VLCFAs play a key role.
Collapse
Affiliation(s)
- Quentin Raas
- Department of Pediatrics, University of Utah, Brain and Spine Center, Primary Children's Hospital, Salt Lake City, Utah, USA
| | - Malu-Clair van de Beek
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam UMC, Amsterdam Gastroenterology & Metabolism, University of Amsterdam, Amsterdam, Netherlands
| | - Sonja Forss-Petter
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Inge Me Dijkstra
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam UMC, Amsterdam Gastroenterology & Metabolism, University of Amsterdam, Amsterdam, Netherlands
| | - Abigail Deschiffart
- Department of Pediatrics, University of Utah, Brain and Spine Center, Primary Children's Hospital, Salt Lake City, Utah, USA
| | - Briana C Freshner
- Department of Pediatrics, University of Utah, Brain and Spine Center, Primary Children's Hospital, Salt Lake City, Utah, USA
| | - Tamara J Stevenson
- Department of Pediatrics, University of Utah, Brain and Spine Center, Primary Children's Hospital, Salt Lake City, Utah, USA
| | - Yorrick Rj Jaspers
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam UMC, Amsterdam Gastroenterology & Metabolism, University of Amsterdam, Amsterdam, Netherlands
| | - Liselotte Nagtzaam
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam UMC, Amsterdam Gastroenterology & Metabolism, University of Amsterdam, Amsterdam, Netherlands
| | - Ronald Ja Wanders
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam UMC, Amsterdam Gastroenterology & Metabolism, University of Amsterdam, Amsterdam, Netherlands
| | - Michel van Weeghel
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam UMC, Amsterdam Gastroenterology & Metabolism, University of Amsterdam, Amsterdam, Netherlands
| | - Joo-Yeon Engelen-Lee
- Department of Neurology, Amsterdam UMC, Amsterdam Neuroscience, University of Amsterdam, Amsterdam, Netherlands
| | - Marc Engelen
- Department of Pediatric Neurology, Amsterdam UMC, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam Neuroscience, University of Amsterdam, Amsterdam, Netherlands
| | - Florian Eichler
- Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Johannes Berger
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Joshua L Bonkowsky
- Department of Pediatrics, University of Utah, Brain and Spine Center, Primary Children's Hospital, Salt Lake City, Utah, USA
| | - Stephan Kemp
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam UMC, Amsterdam Gastroenterology & Metabolism, University of Amsterdam, Amsterdam, Netherlands.,Department of Pediatric Neurology, Amsterdam UMC, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam Neuroscience, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
4
|
Protein Degradome of Spinal Cord Injury: Biomarkers and Potential Therapeutic Targets. Mol Neurobiol 2020; 57:2702-2726. [PMID: 32328876 DOI: 10.1007/s12035-020-01916-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 03/31/2020] [Indexed: 12/13/2022]
Abstract
Degradomics is a proteomics sub-discipline whose goal is to identify and characterize protease-substrate repertoires. With the aim of deciphering and characterizing key signature breakdown products, degradomics emerged to define encryptic biomarker neoproteins specific to certain disease processes. Remarkable improvements in structural and analytical experimental methodologies as evident in research investigating cellular behavior in neuroscience and cancer have allowed the identification of specific degradomes, increasing our knowledge about proteases and their regulators and substrates along with their implications in health and disease. A physiologic balance between protein synthesis and degradation is sought with the activation of proteolytic enzymes such as calpains, caspases, cathepsins, and matrix metalloproteinases. Proteolysis is essential for development, growth, and regeneration; however, inappropriate and uncontrolled activation of the proteolytic system renders the diseased tissue susceptible to further neurotoxic processes. In this article, we aim to review the protease-substrate repertoires as well as emerging therapeutic interventions in spinal cord injury at the degradomic level. Several protease substrates and their breakdown products, essential for the neuronal structural integrity and functional capacity, have been characterized in neurotrauma including cytoskeletal proteins, neuronal extracellular matrix glycoproteins, cell junction proteins, and ion channels. Therefore, targeting exaggerated protease activity provides a potentially effective therapeutic approach in the management of protease-mediated neurotoxicity in reducing the extent of damage secondary to spinal cord injury.
Collapse
|
5
|
Choudhary M, Ismail EN, Yao PL, Tayyari F, Radu RA, Nusinowitz S, Boulton ME, Apte RS, Ruberti JW, Handa JT, Tontonoz P, Malek G. LXRs regulate features of age-related macular degeneration and may be a potential therapeutic target. JCI Insight 2020; 5:131928. [PMID: 31829999 DOI: 10.1172/jci.insight.131928] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 12/05/2019] [Indexed: 12/11/2022] Open
Abstract
Effective treatments and animal models for the most prevalent neurodegenerative form of blindness in elderly people, called age-related macular degeneration (AMD), are lacking. Genome-wide association studies have identified lipid metabolism and inflammation as AMD-associated pathogenic pathways. Given liver X receptors (LXRs), encoded by the nuclear receptor subfamily 1 group H members 2 and 3 (NR1H3 and NR1H2), are master regulators of these pathways, herein we investigated the role of LXR in human and mouse eyes as a function of age and disease and tested the therapeutic potential of targeting LXR. We identified immunopositive LXR fragments in human extracellular early dry AMD lesions and a decrease in LXR expression within the retinal pigment epithelium (RPE) as a function of age. Aged mice lacking LXR presented with isoform-dependent ocular pathologies. Specifically, loss of the Nr1h3 isoform resulted in pathobiologies aligned with AMD, supported by compromised visual function, accumulation of native and oxidized lipids in the outer retina, and upregulation of ocular inflammatory cytokines, while absence of Nr1h2 was associated with ocular lipoidal degeneration. LXR activation not only ameliorated lipid accumulation and oxidant-induced injury in RPE cells but also decreased ocular inflammatory markers and lipid deposition in a mouse model, thereby providing translational support for pursuing LXR-active pharmaceuticals as potential therapies for dry AMD.
Collapse
Affiliation(s)
- Mayur Choudhary
- Duke Eye Center, Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Ebraheim N Ismail
- Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA
| | - Pei-Li Yao
- Duke Eye Center, Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Faryan Tayyari
- Duke Eye Center, Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Roxana A Radu
- Stein Eye Institute, Department of Ophthalmology, UCLA, Los Angeles, California, USA
| | - Steven Nusinowitz
- Stein Eye Institute, Department of Ophthalmology, UCLA, Los Angeles, California, USA
| | - Michael E Boulton
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Rajendra S Apte
- Department of Ophthalmology and Visual Sciences, Washington University in Saint Louis School of Medicine, Saint Louis, Missouri, USA
| | - Jeffrey W Ruberti
- Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA
| | - James T Handa
- Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Peter Tontonoz
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, California, USA
| | - Goldis Malek
- Duke Eye Center, Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, USA.,Department of Pathology, Duke University School of Medicine, Durham, North Carolina, USA
| |
Collapse
|
6
|
Li YJ, Zhang K, Sun T, Wang J, Guo YY, Yang L, Yang Q, Li YJ, Liu SB, Zhao MG, Wu YM. Epigenetic suppression of liver X receptor β in anterior cingulate cortex by HDAC5 drives CFA-induced chronic inflammatory pain. J Neuroinflammation 2019; 16:132. [PMID: 31255170 PMCID: PMC6599528 DOI: 10.1186/s12974-019-1507-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 05/21/2019] [Indexed: 01/30/2023] Open
Abstract
Background Liver X receptors (LXRs), including LXRα and LXRβ, are key regulators of transcriptional programs for both cholesterol homeostasis and inflammation in the brain. Here, the modes of action of LXRs and the epigenetic mechanisms regulating LXRβ expression in anterior cingulate cortex (ACC) of chronic inflammatory pain (CIP) are investigated. Methods The deficit of LXR isoform and analgesic effect of LXR activation by GW3965 were evaluated using the mouse model of CIP induced by hindpaw injection of complete Freund’s adjuvant (CFA). The mechanisms involved in GW-mediated analgesic effects were analyzed with immunohistochemical methods, ELISA, co-immunoprecipitation (Co-IP), Western blot, and electrophysiological recording. The epigenetic regulation of LXRβ expression was investigated by chromatin immunoprecipitation, quantitative real-time PCR, and sequencing. Results We revealed that CFA insult led to LXRβ reduction in ACC, which was associated with upregulated expression of histone deacetylase 5 (HDAC5), and knockdown of LXRβ by shRNA led to thermal hyperalgesia. Co-IP showed that LXRβ interacted with NF-κB p65 physically. LXRβ activation by GW3965 exerted analgesic effects by inhibiting the nuclear translocation of NF-κB, reducing the phosphorylation of mitogen-activated protein kinases (MAPKs) in ACC, and decreasing the promoted input-output and enhanced mEPSC frequency in ACC neurons after CFA exposure. In vitro experiments confirmed that HDAC5 triggered histone deacetylation on the promoter region of Lxrβ, resulting in downregulation of Lxrβ transcription. Conclusion These findings highlight an epigenetic mechanism underlying LXRβ deficits linked to CIP, and LXRβ activation may represent a potential novel target for the treatment of CIP with an alteration in inflammation responses and synaptic transmission in ACC. Electronic supplementary material The online version of this article (10.1186/s12974-019-1507-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yu-Jiao Li
- Department of Pharmacy, Precision Pharmacy & Drug Development Center, The Second Affiliated Hospital, Fourth Military Medical University, Xi'an, 710038, Shaanxi Province, People's Republic of China.,Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, People's Republic of China
| | - Kun Zhang
- Department of Pharmacy, Precision Pharmacy & Drug Development Center, The Second Affiliated Hospital, Fourth Military Medical University, Xi'an, 710038, Shaanxi Province, People's Republic of China.,Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, People's Republic of China
| | - Ting Sun
- Department of Pharmacy, Precision Pharmacy & Drug Development Center, The Second Affiliated Hospital, Fourth Military Medical University, Xi'an, 710038, Shaanxi Province, People's Republic of China
| | - Jian Wang
- Department of Ambulatorium, 94750 Army Hospital, Liancheng, 366200, FuJian Province, People's Republic of China
| | - Yan-Yan Guo
- Department of Pharmacy, Precision Pharmacy & Drug Development Center, The Second Affiliated Hospital, Fourth Military Medical University, Xi'an, 710038, Shaanxi Province, People's Republic of China
| | - Le Yang
- Department of Pharmacy, Precision Pharmacy & Drug Development Center, The Second Affiliated Hospital, Fourth Military Medical University, Xi'an, 710038, Shaanxi Province, People's Republic of China
| | - Qi Yang
- Department of Pharmacy, Precision Pharmacy & Drug Development Center, The Second Affiliated Hospital, Fourth Military Medical University, Xi'an, 710038, Shaanxi Province, People's Republic of China
| | - Yan-Jiao Li
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, People's Republic of China.,Department of Acupuncture and Moxibustion, Xi'an Hospital of Traditional Chinese Medicine, Xi'an, 710021, Shaanxi Province, People's Republic of China
| | - Shui-Bing Liu
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, People's Republic of China
| | - Ming-Gao Zhao
- Department of Pharmacy, Precision Pharmacy & Drug Development Center, The Second Affiliated Hospital, Fourth Military Medical University, Xi'an, 710038, Shaanxi Province, People's Republic of China. .,Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, People's Republic of China.
| | - Yu-Mei Wu
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, People's Republic of China.
| |
Collapse
|
7
|
Campolo M, Paterniti I, Siracusa R, Filippone A, Esposito E, Cuzzocrea S. TLR4 absence reduces neuroinflammation and inflammasome activation in Parkinson's diseases in vivo model. Brain Behav Immun 2019; 76:236-247. [PMID: 30550933 DOI: 10.1016/j.bbi.2018.12.003] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 11/30/2018] [Accepted: 12/10/2018] [Indexed: 12/18/2022] Open
Abstract
Parkinson's disease (PD) is a progressive, disabling neurodegenerative disorder. It has been shown Toll like receptor (TLR) 4-deficient mice protect against MPTP toxicity, suggesting that dopaminergic cell death is TLR4-dependent. The aim of this study was to demonstrate, in an in vivo model of PD, how TLR4 plays its important role in the pathogenesis of PD by using MPTP neurotoxin model (4 × 20 mg/kg, 2 h apart, i.p). Our experiments have demonstrated that the absence of TLR4 prevented dopamine depletion, increased tyrosine hydroxylase and dopamine transporter activities and reduced the number of α-synuclein-positive neurons. The absence of TLR4 also had an impact on inflammatory processes, modulating the transcription factors NF-κB p65 and AP-1, and reducing astrogliosis. Importantly, we demonstrated that the absence of TLR4 modulated inflammosome pathway. Moreover, it has been shown that TLR4 modulated motor and non-motor symptoms typical of PD. Our results clearly demonstrated that absence of TLR4 reduces the development of neuroinflammation associated with PD through NF-κB, AP-1 and inflammasome pathways modulation; therefore, TLR4 could be considered as an encouraging therapeutic target in neurodegenerative disorders.
Collapse
Affiliation(s)
- Michela Campolo
- Department of Chemical, Biological, Pharmacological and Environmental Sciences, University of Messina, Messina, Italy
| | - Irene Paterniti
- Department of Chemical, Biological, Pharmacological and Environmental Sciences, University of Messina, Messina, Italy
| | - Rosalba Siracusa
- Department of Chemical, Biological, Pharmacological and Environmental Sciences, University of Messina, Messina, Italy
| | - Alessia Filippone
- Department of Chemical, Biological, Pharmacological and Environmental Sciences, University of Messina, Messina, Italy
| | - Emanuela Esposito
- Department of Chemical, Biological, Pharmacological and Environmental Sciences, University of Messina, Messina, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmacological and Environmental Sciences, University of Messina, Messina, Italy; Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, USA.
| |
Collapse
|
8
|
Paterniti I, Campolo M, Cordaro M, Siracusa R, Filippone A, Esposito E, Cuzzocrea S. Effects of different natural extracts in an experimental model of benign prostatic hyperplasia (BPH). Inflamm Res 2018; 67:617-626. [PMID: 29679313 DOI: 10.1007/s00011-018-1152-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 04/11/2018] [Accepted: 04/16/2018] [Indexed: 11/28/2022] Open
Abstract
OBJECTIVE AND DESIGN To characterize the impact of inflammatory process and oxidative stress in the degree of benign prostatic hyperplasia (BPH), a common condition in which chronic inflammation plays a crucial role, we investigated the effect of different plant extract preparations in an in vivo model of BPH as new therapeutic target. MATERIAL BPH was made in rats with daily administration of testosterone propionate (3 mg/kg) for 14 days. TREATMENT Rats were randomized into different groups to receive oral administration of plant extract preparations: Serenoa repens with selenium (SeR 28.5 mg/kg associated with Se 0.005 mg/kg), Teoside (2 mg/kg), and Puryprost (14 mg/kg containing Teoside 50% 2 mg/kg and Epilobium 12 mg/kg). METHODS After 14 days, rats were killed and histological changes, prostate weight and apoptotic pathways were assayed. RESULTS The results obtained demonstrated that the association of treatments reduced prostate weight and hyperplasia, while treatment with Puryprost demonstrated a greater trend of protection compared to the other treatments. CONCLUSION Thus, our results indicate that plant extract could be considered as new useful therapy in the treatment of BPH with particular attention on Puryprost that represents a rational approach to reduce BPH through modulation of inflammatory process and anti-oxidant process.
Collapse
Affiliation(s)
- Irene Paterniti
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres No. 31, 98166, Messina, Italy
| | - Michela Campolo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres No. 31, 98166, Messina, Italy
| | - Marika Cordaro
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres No. 31, 98166, Messina, Italy
| | - Rosalba Siracusa
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres No. 31, 98166, Messina, Italy
| | - Antonio Filippone
- Department of Biomedical and Biotechnological Science, University of Catania, Catania, Italy
| | - Emanuela Esposito
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres No. 31, 98166, Messina, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres No. 31, 98166, Messina, Italy. .,Department of Pharmacological and Physiological Science, Saint Louis University, St. Louis, USA.
| |
Collapse
|
9
|
Sobrido-Cameán D, Barreiro-Iglesias A. Role of Caspase-8 and Fas in Cell Death After Spinal Cord Injury. Front Mol Neurosci 2018; 11:101. [PMID: 29666570 PMCID: PMC5891576 DOI: 10.3389/fnmol.2018.00101] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 03/15/2018] [Indexed: 01/10/2023] Open
Abstract
Spinal cord injury (SCI) causes the death of neurons and glial cells due to the initial mechanical forces (i.e., primary injury) and through a cascade of secondary molecular events (e.g., inflammation or excitotoxicity) that exacerbate cell death. The loss of neurons and glial cells that are not replaced after the injury is one of the main causes of disability after SCI. Evidence accumulated in last decades has shown that the activation of apoptotic mechanisms is one of the factors causing the death of intrinsic spinal cord (SC) cells following SCI. Although this is not as clear for brain descending neurons, some studies have also shown that apoptosis can be activated in the brain following SCI. There are two main apoptotic pathways, the extrinsic and the intrinsic pathways. Activation of caspase-8 is an important step in the initiation of the extrinsic pathway. Studies in rodents have shown that caspase-8 is activated in SC glial cells and neurons and that the Fas receptor plays a key role in its activation following a traumatic SCI. Recent work in the lamprey model of SCI has also shown the retrograde activation of caspase-8 in brain descending neurons following SCI. Here, we review our current knowledge on the role of caspase-8 and the Fas pathway in cell death following SCI. We also provide a perspective for future work on this process, like the importance of studying the possible contribution of Fas/caspase-8 signaling in the degeneration of brain neurons after SCI in mammals.
Collapse
Affiliation(s)
- Daniel Sobrido-Cameán
- Department of Functional Biology, CIBUS, Faculty of Biology, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Antón Barreiro-Iglesias
- Department of Functional Biology, CIBUS, Faculty of Biology, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| |
Collapse
|
10
|
Cordaro M, Casili G, Paterniti I, Cuzzocrea S, Esposito E. Fumaric Acid Esters Attenuate Secondary Degeneration after Spinal Cord Injury. J Neurotrauma 2017; 34:3027-3040. [DOI: 10.1089/neu.2016.4678] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Affiliation(s)
- Marika Cordaro
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Giovanna Casili
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Irene Paterniti
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
- Pharmacological and Physiological Science, Saint Louis University School of Medicine, St. Louis, Missouri
| | - Emanuela Esposito
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| |
Collapse
|
11
|
Liver X Receptor α Is Involved in Counteracting Mechanical Allodynia by Inhibiting Neuroinflammation in the Spinal Dorsal Horn. Anesthesiology 2017; 127:534-547. [PMID: 28617705 DOI: 10.1097/aln.0000000000001718] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Liver X receptors, including α and β isoforms, are ligand-activated transcription factors. Whether liver X receptor α plays a role in neuropathic pain is unknown. METHODS A spared nerve injury model was established in adult male rats and mice. Von Frey tests were performed to evaluate the neuropathic pain behavior; Western blot and immunohistochemistry were performed to understand the underlying mechanisms. RESULTS Intrathecal injection of a specific liver X receptor agonist T0901317 or GW3965 could either prevent the development of mechanical allodynia or alleviate the established mechanical allodynia, both in rats and wild-type mice. GW3965 could inhibit the activation of glial cells and the expression of tumor necrosis factor-α (mean ± SD: 196 ± 48 vs. 119 ± 57; n = 6; P < 0.01) and interleukin 1β (mean ± SD: 215 ± 69 vs. 158 ± 74; n = 6; P < 0.01) and increase the expression of interleukin 10 in the spinal dorsal horn. All of the above effects of GW3965 could be abolished by liver X receptor α mutation. Moreover, more glial cells were activated, and more interleukin 1β was released in the spinal dorsal horn in liver X receptor α knockout mice than in wild-type mice after spared nerve injury. Aminoglutethimide, a neurosteroid synthesis inhibitor, blocked the inhibitory effect of T0901317 on mechanical allodynia, on the activation of glial cells, and on the expression of cytokines. CONCLUSIONS Activation of liver X receptor α inhibits mechanical allodynia by inhibiting the activation of glial cells and rebalancing cytokines in the spinal dorsal horn via neurosteroids.
Collapse
|
12
|
Fessler MB. The challenges and promise of targeting the Liver X Receptors for treatment of inflammatory disease. Pharmacol Ther 2017; 181:1-12. [PMID: 28720427 DOI: 10.1016/j.pharmthera.2017.07.010] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The Liver X Receptors (LXRs) are oxysterol-activated transcription factors that upregulate a suite of genes that together promote coordinated mobilization of excess cholesterol from cells and from the body. The LXRs, like other nuclear receptors, are anti-inflammatory, inhibiting signal-dependent induction of pro-inflammatory genes by nuclear factor-κB, activating protein-1, and other transcription factors. Synthetic LXR agonists have been shown to ameliorate atherosclerosis and a wide range of inflammatory disorders in preclinical animal models. Although this has suggested potential for application to human disease, systemic LXR activation is complicated by hepatic steatosis and hypertriglyceridemia, consequences of lipogenic gene induction in the liver by LXRα. The past several years have seen the development of multiple advanced LXR therapeutics aiming to avoid hepatic lipogenesis, including LXRβ-selective agonists, tissue-selective agonists, and transrepression-selective agonists. Although several synthetic LXR agonists have made it to phase I clinical trials, none have progressed due to unforeseen adverse reactions or undisclosed reasons. Nonetheless, several sophisticated pharmacologic strategies, including structure-guided drug design, cell-specific drug targeting, as well as non-systemic drug routes have been initiated and remain to be comprehensively explored. In addition, recent studies have identified potential utility for targeting the LXRs during therapy with other agents, such as glucocorticoids and rexinoids. Despite the pitfalls encountered to date in translation of LXR agonists to human disease, it appears likely that this accelerating field will ultimately yield effective and safe applications for LXR targeting in humans.
Collapse
Affiliation(s)
- Michael B Fessler
- National Institute of Environmental Health Sciences, 111 T.W. Alexander Drive, P.O. Box 12233, MD D2-01, Research Triangle Park, NC 27709, United States.
| |
Collapse
|
13
|
Liver X receptors activation, through TO901317 binding, reduces neuroinflammation in Parkinson's disease. PLoS One 2017; 12:e0174470. [PMID: 28369131 PMCID: PMC5378346 DOI: 10.1371/journal.pone.0174470] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 03/09/2017] [Indexed: 11/23/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disease in which degeneration of nigrostriatal neurons and inflammation are key players. The aim of our study was to analyze the function of LXRs in neurodegenerative diseases as PD using in vivo, ex vivo and in vitro models of PD; for this purpose, we observed the effects of the LXR agonist, TO901317, in neuroinflammatory pathway related to PD. We performed an in vivo model of PD using the neurotoxin 1-methyl-4-phenyl-1, 2,3,6-tetrahydropyridine (MPTP) and our results clearly showed that TO901317 administration reduces all of the inflammatory markers involved in PD such as iNOS and COX2, IκB-α and NF-κB. Moreover, to confirm the neuroprotective properties of TO901317, that we obtained with the in vivo model, we performed also an ex vivo and in vitro models of PD. All the results taken, confirmed that TO901317 is able to modulate the neuroinflammatory pathway involved in PD increasing the locomotors function. Therefore, TO901317, LXR synthetic agonist, could be studied as a new target in a neurodegenerative disorder like PD.
Collapse
|
14
|
Barger SW. Gene regulation and genetics in neurochemistry, past to future. J Neurochem 2016; 139 Suppl 2:24-57. [PMID: 27747882 DOI: 10.1111/jnc.13629] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2015] [Revised: 03/01/2016] [Accepted: 03/30/2016] [Indexed: 12/14/2022]
Abstract
Ask any neuroscientist to name the most profound discoveries in the field in the past 60 years, and at or near the top of the list will be a phenomenon or technique related to genes and their expression. Indeed, our understanding of genetics and gene regulation has ushered in whole new systems of knowledge and new empirical approaches, many of which could not have even been imagined prior to the molecular biology boon of recent decades. Neurochemistry, in the classic sense, intersects with these concepts in the manifestation of neuropeptides, obviously dependent upon the central dogma (the established rules by which DNA sequence is eventually converted into protein primary structure) not only for their conformation but also for their levels and locales of expression. But, expanding these considerations to non-peptide neurotransmitters illustrates how gene regulatory events impact neurochemistry in a much broader sense, extending beyond the neurochemicals that translate electrical signals into chemical ones in the synapse, to also include every aspect of neural development, structure, function, and pathology. From the beginning, the mutability - yet relative stability - of genes and their expression patterns were recognized as potential substrates for some of the most intriguing phenomena in neurobiology - those instances of plasticity required for learning and memory. Near-heretical speculation was offered in the idea that perhaps the very sequence of the genome was altered to encode memories. A fascinating component of the intervening progress includes evidence that the central dogma is not nearly as rigid and consistent as we once thought. And this mutability extends to the potential to manipulate that code for both experimental and clinical purposes. Astonishing progress has been made in the molecular biology of neurochemistry during the 60 years since this journal debuted. Many of the gains in conceptual understanding have been driven by methodological progress, from automated high-throughput sequencing instruments to recombinant-DNA vectors that can convey color-coded genetic modifications in the chromosomes of live adult animals. This review covers the highlights of these advances, both theoretical and technological, along with a brief window into the promising science ahead. This article is part of the 60th Anniversary special issue.
Collapse
Affiliation(s)
- Steven W Barger
- Department of Geriatrics, Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA. .,Geriatric Research Education and Clinical Center, Central Arkansas Veterans Healthcare System, Little Rock, Arkansas, USA.
| |
Collapse
|
15
|
Zhao Z, Xu D, Li S, He B, Huang Y, Xu M, Ren S, Li S, Wang H, Xie W. Activation of Liver X Receptor Attenuates Oleic Acid-Induced Acute Respiratory Distress Syndrome. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:2614-22. [PMID: 27520356 PMCID: PMC5222979 DOI: 10.1016/j.ajpath.2016.06.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 05/19/2016] [Accepted: 06/23/2016] [Indexed: 01/11/2023]
Abstract
Liver X receptors (LXRs) were identified as receptors that sense oxidized cholesterol derivatives. LXRs are best known for their hepatic functions in regulating cholesterol metabolism and triglyceride synthesis, but whether and how LXRs play a role in the lung diseases is less understood. To study the function of LXRs in acute respiratory distress syndrome (ARDS), we applied the oleic acid (OA) model of ARDS to mice whose LXR was genetically or pharmacologically activated. The VP-LXRα knock-in (LXR-KI) mice, in which a constitutively activated LXRα (VP-LXRα) was inserted into the mouse LXRα locus, were used as the genetic gain-of-function model. We showed that the OA-induced lung damages, including the cytokine levels and total cell numbers and neutrophil numbers in the bronchoalveolar lavage fluid, the wet/dry weight ratio, and morphological abnormalities were reduced in the LXR-KI mice and wild-type mice treated with the LXR agonist GW3965. The pulmonoprotective effect of GW3965 was abolished in the LXR-null mice. Consistent with the pulmonoprotective effect of LXR and the induction of antioxidant enzymes by LXR, the OA-induced suppression of superoxide dismutase and catalase was attenuated in LXR-KI mice and GW3965-treated wild-type mice. Taken together, our results demonstrate that activation of LXRs can alleviate OA-induced ARDS by attenuating the inflammatory response and enhancing antioxidant capacity.
Collapse
Affiliation(s)
- Zanmei Zhao
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Occupational Disease, Peking University Third Hospital, Beijing, China
| | - Dan Xu
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Shuqiang Li
- Department of Occupational Disease, Peking University Third Hospital, Beijing, China
| | - Bei He
- Department of Respiratory Medicine, Peking University Third Hospital, Beijing, China
| | - Yixian Huang
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Meishu Xu
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Songrong Ren
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Song Li
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Hui Wang
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Wen Xie
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|
16
|
Casili G, Impellizzeri D, Cordaro M, Esposito E, Cuzzocrea S. B-Cell Depletion with CD20 Antibodies as New Approach in the Treatment of Inflammatory and Immunological Events Associated with Spinal Cord Injury. Neurotherapeutics 2016; 13:880-894. [PMID: 27215219 PMCID: PMC5081113 DOI: 10.1007/s13311-016-0446-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Spinal cord injury (SCI) is a highly debilitating pathology that has irreversible impacts and results in functional loss. We evaluated the anti-inflammatory and immunologic role of antibody-mediated depletion of B cells through the glycoengineered anti-muCD20 antibody (18B12) in an experimental model of spinal cord compression, in vivo and ex vivo. Intraperitoneal 18B12 was administered at a dose of 30 mg/kg, 1 h and 6 h after SCI, and mice were sacrificed 24 h after trauma. We demonstrated, in vivo, that 18B12 slowed severe hindlimb motor dysfunction (Basso Mouse Scale score) and neuronal death by histological evaluation in SCI mice, as well as decreased expression of nuclear factor-kB, inducible nitric oxide synthase, cytokines, and glial fibrillary acidic protein. Also, 18B12 reduced expression of microglia, just as it lowered the expression of B and T lymphocytes. Moreover, in spinal cord organotypic cultures, pretreatment with 18B12 significantly reduced nitric oxide expression and protected cells from cell death [3-(4,5- dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay]. In this study, we showed that 18B12 treatment reduces the development of inflammation and tissue injury by alteration of the immune system associated with SCI. This study increases the current knowledge that B-cell depletion is able to exert immunomodulating actions in damaged spinal cords.
Collapse
Affiliation(s)
- Giovanna Casili
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres n°31 98166, Messina, Italy
| | - Daniela Impellizzeri
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres n°31 98166, Messina, Italy
| | - Marika Cordaro
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres n°31 98166, Messina, Italy
| | - Emanuela Esposito
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres n°31 98166, Messina, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres n°31 98166, Messina, Italy.
- Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, St. Louis, MO, 63104, USA.
| |
Collapse
|
17
|
Wu CH, Chen CC, Lai CY, Hung TH, Lin CC, Chao M, Chen SF. Treatment with TO901317, a synthetic liver X receptor agonist, reduces brain damage and attenuates neuroinflammation in experimental intracerebral hemorrhage. J Neuroinflammation 2016; 13:62. [PMID: 26968836 PMCID: PMC4788882 DOI: 10.1186/s12974-016-0524-8] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 03/03/2016] [Indexed: 11/10/2022] Open
Abstract
Background Intracerebral hemorrhage (ICH) induces a series of inflammatory processes that contribute to neuronal damage and neurological deterioration. Liver X receptors (LXRs) are nuclear receptors that negatively regulate transcriptional processes involved in inflammatory responses, but their role in the pathology following ICH remains unclear. The present study investigated the neuroprotective effects and anti-inflammatory actions of TO901317, a synthetic LXR agonist, in a model of collagenase-induced ICH and in microglial cultures. Methods Mice subjected to collagenase-induced ICH injury were injected with either TO901317 (30 mg/kg) or vehicle 10 min after ICH and subsequently daily for 2 days. Behavioral studies, histology analysis, and assessments of hematoma volumes, brain water content, and blood-brain barrier (BBB) permeability were performed. The protein expression of LXR-α, LXR-β, ATP binding cassette transporter-1 (ABCA-1), and inflammatory molecules was analyzed. The anti-inflammatory mechanism of TO901317 was investigated in cultured microglia that were stimulated with either lipopolysaccharide (LPS) or thrombin. Results ICH induced an increase in LXR-α protein levels in the hemorrhagic hemisphere at 6 h whereas LXR-β expression remained unaffected. Both LXR-α and LXR-β were expressed in neurons and microglia in the peri-ICH region and but rarely in astrocytes. TO901317 significantly attenuated functional deficits and brain damage up to 28 days post-ICH. TO901317 also reduced neuronal death, BBB disruption, and brain edema at day 4 post-ICH. These changes were associated with marked reductions in microglial activation, neutrophil infiltration, and expression levels of inflammatory mediators at 4 and 7 days. However, TO901317 had no effect on matrix metalloproteinase-9 activity. In BV2 microglial cultures, TO901317 attenuated LPS- and thrombin-stimulated nitric oxide production and reduced LPS-induced p38, JNK, MAPK, and nuclear factor-kappa B (NF-κB) signaling. Moreover, delaying administration of TO901317 to 3 h post-ICH reduced brain tissue damage and neuronal death. Conclusions Our results suggest that enhancing LXR activation may provide a potential therapy for ICH by modulating the cytotoxic functions of microglia. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0524-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Chun-Hu Wu
- Department of Physiology and Biophysics, National Defense Medical Center, Taipei, Taiwan, Republic of China.,Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Chien-Cheng Chen
- Department of Physical Medicine and Rehabilitation, Cheng Hsin General Hospital, Taipei, Taiwan, Republic of China
| | - Chai-You Lai
- Department of Physiology and Biophysics, National Defense Medical Center, Taipei, Taiwan, Republic of China.,Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Tai-Ho Hung
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital at Taipei and College of Medicine, Chang Gung University, Taipei, Taiwan, Republic of China
| | - Chao-Chang Lin
- Department of Physical Medicine and Rehabilitation, Cheng Hsin General Hospital, Taipei, Taiwan, Republic of China
| | - Min Chao
- School of Medicine, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Szu-Fu Chen
- Department of Physiology and Biophysics, National Defense Medical Center, Taipei, Taiwan, Republic of China. .,Department of Physical Medicine and Rehabilitation, Cheng Hsin General Hospital, Taipei, Taiwan, Republic of China.
| |
Collapse
|
18
|
Bao X, Cai Y, Wang Y, Zhao J, He X, Yu D, Huang J, Jing S, Du Z, Yang T, Warner M, Gustafsson JA, Fan X. Liver X Receptor β Is Involved in Formalin-Induced Spontaneous Pain. Mol Neurobiol 2016; 54:1467-1481. [PMID: 26846362 DOI: 10.1007/s12035-016-9737-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 01/20/2016] [Indexed: 12/30/2022]
Abstract
Increasing evidence indicates that the liver X receptor(LXR) β modulates inflammatory pain. However, the molecular mechanisms through which LXRβ modulates pain are unclear. Here, we found that LXRβ-null mice responded more strongly to acute noxious stimuli than wild-type (WT) littermates (in the hot plate and Hargreaves tests) and had augmented tonic inflammatory pain (in the formalin test). This increased reactivity to inflammatory pain was accompanied by enhanced formalin-evoked Fos and pERK staining of second-order nociceptive neurons. Immunohistochemistry showed that the expression of CGRP, SP, and IB4 was increased in the lamina I-II of the lumbar dorsal horns in formalin-injected LXRβ knockout (KO) mice compared with the WT controls. In addition, LXRβ deletion in the mice enhanced the formalin-induced inflammation with more activated microglia and astrocytes in the spinal cord. Furthermore, the levels of pro-inflammatory cytokines (IL-1β ,TNF-α) as well as NFκB in the formalin-injected paw were elevated by the loss of LXRβ. Taken together, these data indicate that LXRβ is involved in acute as well as inflammatory pain, and thus, it may be considered as a new target for the development of analgesics.
Collapse
Affiliation(s)
- Xiaohang Bao
- Department of Anesthesiology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400038, People's Republic of China.,Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing, 400038, China
| | - Yulong Cai
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing, 400038, China
| | - Ying Wang
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing, 400038, China
| | - Jinghui Zhao
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing, 400038, China
| | - Xie He
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing, 400038, China
| | - Dan Yu
- Department of Anesthesiology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400038, People's Republic of China
| | - Jing Huang
- Department of Anesthesiology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400038, People's Republic of China
| | - Sheng Jing
- Department of Anesthesiology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400038, People's Republic of China
| | - Zhiyong Du
- Department of Anesthesiology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400038, People's Republic of China
| | - Tiande Yang
- Department of Anesthesiology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400038, People's Republic of China.
| | - Margaret Warner
- Department of Biology and Biochemistry, Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX, 77054, USA
| | - Jan-Ake Gustafsson
- Department of Biology and Biochemistry, Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX, 77054, USA. .,Department of Biosciences and Nutrition, Karolinska Institute, Novum, Novum, 141 86, Sweden.
| | - Xiaotang Fan
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing, 400038, China.
| |
Collapse
|
19
|
Kruczek D, Clarner T, Beyer C, Kipp M, Mey J. Activation of Nuclear Receptors RAR, RXR, and LXR Does Not Reduce Cuprizone-Induced Demyelination in Mice. NUCLEAR RECEPTOR RESEARCH 2015. [DOI: 10.11131/2015/101163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
| | - Tim Clarner
- Institut für Neuroanatomie, Universitätsklinikum Aachen, Germany
| | - Cordian Beyer
- Institut für Neuroanatomie, Universitätsklinikum Aachen, Germany
| | - Markus Kipp
- Institut für Neuroanatomie, Universitätsklinikum Aachen, Germany
- Lehrstuhl II – Neuroanatomie, Ludwig-Maximilians-Universität München, Germany
| | - Jörg Mey
- Institut für Biologie II, RWTH Aachen, Germany
- Laboratorio Regeneraci73243;n Nerviosa, Hospital Nacional de Parapléjicos, Toledo, Spain
- EURON Graduate School of Neuroscience, Maastricht University, Netherlands
| |
Collapse
|
20
|
Han M, Liang L, Liu LR, Yue J, Zhao YL, Xiao HP. Liver X receptor gene polymorphisms in tuberculosis: effect on susceptibility. PLoS One 2014; 9:e95954. [PMID: 24788534 PMCID: PMC4006844 DOI: 10.1371/journal.pone.0095954] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 04/02/2014] [Indexed: 01/06/2023] Open
Abstract
Objectives The Liver X receptors (LXRs), Liver X receptor A (LXRA) and Liver X receptor B (LXRB), regulate lipid metabolism and antimicrobial response. LXRs have a crucial role in the control of Mycobacterium tuberculosis (M.tb). Lacking LXRs mice is more susceptibility to infection M.tb, developing higher bacterial burdens and an increase in the size and number of granulomatous lesions. We aimed to assess the associations between single nucleotide polymorphisms (SNPs) in LXRs and risk of tuberculosis. Methods We sequenced the LXRs genes to detect SNPs and to examine genotypic frequencies in 600 patients and 620 healthy controls to investigate for associations with tuberculosis (TB) in the Chinese Han population. DNA re-sequencing revealed eight common variants in the LXRs genes. Results The G allele of rs1449627 and the T allele of rs1405655 demonstrated an increased risk of developing TB (p<0.001, p = 0.002), and the T allele of rs3758673, the T allele of rs2279238, and the C allele of rs1449626 in LXRA and the C allele of rs17373080, the G allele of rs2248949, and the C allele of rs1052677 in LXRB were protective against TB patients compared to healthy controls (p = 0.0002, p = 0.006, p<0.001, p = 0.004, p = 0.008, p = 0.003, respectively). All SNP genotypes were significantly associated with TB. An estimation of the frequencies of haplotypes revealed two potential risk haplotypes,GGCG in LXRB (p = 0.004,) and TTCG in LXRA (p<0.001, p = 0.004). Moreover, three protective haplotypes, TTAT and CCAT in LXRA and CATC in LXRB, were significantly “protective” (p = 0.008, p<0.001, p = 0.031) for TB. Furthermore, we determined that the LXRs SNPs were nominally associated with the clinical pattern of disease. Conclusions Our study data supported that LXRs play a fundamental role in the genetic susceptibility to TB and to different clinical patterns of disease. Thus, further investigation is required in larger populations and in additional areas.
Collapse
Affiliation(s)
- Min Han
- Shanghai Key Laboratory of Mycobacterium Tuberculosis, Shanghai Pulmonary Hospital Affiliated to Tongji University School of Medicine, Shanghai, P.R. China
| | - Li Liang
- Shanghai Key Laboratory of Mycobacterium Tuberculosis, Shanghai Pulmonary Hospital Affiliated to Tongji University School of Medicine, Shanghai, P.R. China
| | - Li-rong Liu
- Shanghai Key Laboratory of Mycobacterium Tuberculosis, Shanghai Pulmonary Hospital Affiliated to Tongji University School of Medicine, Shanghai, P.R. China
| | - Jun Yue
- Shanghai Key Laboratory of Mycobacterium Tuberculosis, Shanghai Pulmonary Hospital Affiliated to Tongji University School of Medicine, Shanghai, P.R. China
- * E-mail:
| | - Yan-lin Zhao
- National Center for Tuberculosis Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, P.R. China
| | - He-ping Xiao
- Shanghai Key Laboratory of Mycobacterium Tuberculosis, Shanghai Pulmonary Hospital Affiliated to Tongji University School of Medicine, Shanghai, P.R. China
| |
Collapse
|
21
|
Paterniti I, Impellizzeri D, Di Paola R, Esposito E, Gladman S, Yip P, Priestley JV, Michael-Titus AT, Cuzzocrea S. Docosahexaenoic acid attenuates the early inflammatory response following spinal cord injury in mice: in-vivo and in-vitro studies. J Neuroinflammation 2014; 11:6. [PMID: 24405628 PMCID: PMC3895696 DOI: 10.1186/1742-2094-11-6] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 12/30/2013] [Indexed: 12/30/2022] Open
Abstract
Background Two families of polyunsaturated fatty acid (PUFA), omega-3 (ω-3) and omega-6 (ω-6), are required for physiological functions. The long chain ω-3 PUFAs, eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), have significant biological effects. In particular, DHA is a major component of cell membranes in the brain. It is also involved in neurotransmission. Spinal cord injury (SCI) is a highly devastating pathology that can lead to catastrophic dysfunction, with a significant reduction in the quality of life. Previous studies have shown that EPA and DHA can exert neuroprotective effects in SCI in mice and rats. The aim of this study was to analyze the mechanism of action of ω-3 PUFAs, such as DHA, in a mouse model of SCI, with a focus on the early pathophysiological processes. Methods In this study, SCI was induced in mice by the application of an aneurysm clip onto the dura mater via a four-level T5 to T8 laminectomy. Thirty minutes after compression, animals received a tail vein injection of DHA at a dose of 250 nmol/kg. All animals were killed at 24 h after SCI, to evaluate various parameters implicated in the spread of the injury. Results Our results in this in-vivo study clearly demonstrate that DHA treatment reduces key factors associated with spinal cord trauma. Treatment with DHA significantly reduced: (1) the degree of spinal cord inflammation and tissue injury, (2) pro-inflammatory cytokine expression (TNF-α), (3) nitrotyrosine formation, (4) glial fibrillary acidic protein (GFAP) expression, and (5) apoptosis (Fas-L, Bax, and Bcl-2 expression). Moreover, DHA significantly improved the recovery of limb function. Furthermore, in this study we evaluated the effect of oxidative stress on dorsal root ganglion (DRG) cells using a well-characterized in-vitro model. Treatment with DHA ameliorated the effects of oxidative stress on neurite length and branching. Conclusions Our results, in vivo and in vitro, clearly demonstrate that DHA treatment reduces the development of inflammation and tissue injury associated with spinal cord trauma.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Salvatore Cuzzocrea
- Department of Biological and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres, 98166 Messina, Italy.
| |
Collapse
|
22
|
Fandel D, Wasmuht D, Avila-Martín G, Taylor JS, Galán-Arriero I, Mey J. Spinal cord injury induced changes of nuclear receptors PPARα and LXRβ and modulation with oleic acid/albumin treatment. Brain Res 2013; 1535:89-105. [PMID: 23958344 DOI: 10.1016/j.brainres.2013.08.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Revised: 08/09/2013] [Accepted: 08/10/2013] [Indexed: 11/19/2022]
Abstract
In previous studies with animal models of spinal cord injury (SCI) pharmacological activation of peroxisome proliferator activated receptors (PPAR) and liver X receptors (LXR) were used to reduce tissue damage and promote behavioral recovery in animal models. We have studied the endogenous expression of the transcription factors PPARα and LXRβ in the chronic stage after SCI in rats. The immunohistochemical investigation revealed a long lasting increase in the level of PPARα in white matter in the vicinity of the lesion site. The source of this signal was identified in a subpopulation of astrocytes outside of the glial scar area. Intrathecal injections of oleic acid/albumin reduced the lesion-induced PPARα immunoreactivity. In addition, ependymal cells displayed a prominent PPARα signal in the non-injured spinal cord, and continued to express the receptor as they proliferated and migrated within the damaged tissue. The nuclear receptor LXRβ was detected at similar levels after SCI as in sham operated animals. We found high levels of immunoreactivity in the gray matter, while in the white matter it was present in subpopulations of astrocytes and oligodendrocytes. Macrophages that had accumulated within the center of the lesion contained LXRβ in their cell nuclei. Possible endogenous functions of PPARα and LXRβ after SCI are discussed, specifically the control of fatty acid and cholesterol metabolism and the regulation of inflammatory reactions.
Collapse
Affiliation(s)
- Daniel Fandel
- Laboratorio Regeneración Nerviosa, Hospital Nacional de Parapléjicos, Servicio de Salud de Castilla la Mancha, Toledo, Spain
| | | | | | | | | | | |
Collapse
|
23
|
Shackleford G, Makoukji J, Grenier J, Liere P, Meffre D, Massaad C. Differential regulation of Wnt/beta-catenin signaling by Liver X Receptors in Schwann cells and oligodendrocytes. Biochem Pharmacol 2013; 86:106-14. [DOI: 10.1016/j.bcp.2013.02.036] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Revised: 02/27/2013] [Accepted: 02/27/2013] [Indexed: 11/16/2022]
|
24
|
Effect of infliximab combined with methylprednisolone on expressions of NF-κB, TRADD, and FADD in rat acute spinal cord injury. Spine (Phila Pa 1976) 2013; 38:E861-9. [PMID: 23574812 DOI: 10.1097/brs.0b013e318294892c] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
STUDY DESIGN The possibility to prevent acute spinal cord injury (ASCI) by selective infliximab combined with methylprednisolone (MP) was assessed in experimental ASCI. OBJECTIVE To investigate the effects of infliximab, MP, and the combination of these 2 agents on expressions of NF-κB (nuclear factor Kappa B), TRADD (tumor necrosis factor receptor associated death domain), and FADD (fas associated death domain) in a rat model of acute spinal cord injury (ASCI), and to confirm the therapeutic efficacy and possible mechanism of infliximab combined with MP in the treatment of ASCI. SUMMARY OF BACKGROUND DATA The theory that SCI can induce tumor necrosis factor-α expression at the injury site has been evaluated. However, there are few data to confirm the therapeutic efficacy of infliximab combinated with MP in the treatment of rat SCI METHODS: One hundred eighty adult male Sprague Dawley rats with 280 to 300 g body weight were allocated randomly and accordingly. We applied Basso, Beattie, Bresnahan locomotor rating scale to assess the hindlimb motor functional score (10 rats × 6 groups), the hematoxylin and eosin stain and immunohistochemistry stain (10 rats × 6 groups) to assay the morphological changes of spinal cord, the arrangement and expressions of NF-κB, TRADD and FADD, and the RT-PCR (10 rats × 6 groups) to evaluate the messenger RNA expressions of NF-κB, TRADD, and FADD. RESULTS The results showed that both infliximab and MP could lower the expressions of NF-κB, TRADD, and FADD 24 hours after the ASCI, and increased Basso, Beattie, Bresnahan score on the 14th and the 21st days after ASCI, suggesting possible neuroprotective effectiveness on attenuating the severity of neurological deficits and improving the locomotor function in the rat ASCI model. Moreover, infliximab combined with MP exhibited the more powerful ability to this amelioration. CONCLUSION Infliximab combined with methylprednisolone may be an effective treatment for the recovery of ASCI. Further study is needed to determine if this neuroprotective effect is seen for long-term outcomes especially in human ASCI.
Collapse
|
25
|
Xu P, Li D, Tang X, Bao X, Huang J, Tang Y, Yang Y, Xu H, Fan X. LXR Agonists: New Potential Therapeutic Drug for Neurodegenerative Diseases. Mol Neurobiol 2013; 48:715-28. [DOI: 10.1007/s12035-013-8461-3] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Accepted: 04/09/2013] [Indexed: 01/04/2023]
|
26
|
Mohammadi E, Ghaedi K, Esmailie A, Rahgozar S. Gene expression profiling of liver X receptor α and Bcl-2-associated X protein in experimental transection spinal cord-injured rats. J Spinal Cord Med 2013; 36:66-71. [PMID: 23433337 PMCID: PMC3555109 DOI: 10.1179/2045772312y.0000000032] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Study of molecular responses to central nervous system injury would be helpful for controlling the harmful pathways post-injury and triggering the useful pathways required for the treatment of injury. OBJECTIVE To investigate the expression level of liver X receptor α (LXRα) which has anti-inflammatory effects and pro-apoptotic Bcl-2-associated X protein (Bax) upon spinal cord injury (SCI). DESIGN To induce SCI, transection was carried out at T9 level of male Wister rats. Approximately 8 mm of rostral, caudal, and epicenter tissues of injured sites in treated rats were chosen for quantitative real-time polymerase chain reaction at the 6, 24, and 72 hours, and 7 and 10 days post-surgery. RESULTS Our results showed a complicated temporal and spatial pattern of alteration in LXRα and Bax mRNA expression levels after SCI. LXRα expression level followed a homologues pattern (additive and subtractive wave) with a difference in time at three areas of studied. Rostral, caudal, and epicenter expression patterns of Bax were dissimilar in these areas. Gradual increase in the expression of Bax without any decrease at the rostral area was observed, presumably indicating the active transcription process of this gene, regardless of its protein situation. CONCLUSION A time lapse significant change in Bax expression level was observed only in the epicenter of injury, emphasizing that apoptotic responses are limited to this area. Furthermore, an increase in LXRα transcription level was observed first in rostral area and then extended to epicentral and caudal areas, implying that inflammation responses extended from rostral to caudal areas.
Collapse
Affiliation(s)
| | - Kamran Ghaedi
- Correspondence to: Kamran Ghaedi (Ph.D.), Biology Department, School of Sciences, University of Isfahan, Hezar jerib Ave., Azadi Sq., Postal Code 73441-81746 Isfahan, Iran. Tel No.: +98-311-7932479; Fax No.: +98-311-7932456.
| | | | | |
Collapse
|
27
|
Robertson Remen KM, Lerner UH, Gustafsson JÅ, Andersson G. Activation of the liver X receptor-β potently inhibits osteoclastogenesis from lipopolysaccharide-exposed bone marrow-derived macrophages. J Leukoc Biol 2013; 93:71-82. [DOI: 10.1189/jlb.0712339] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
28
|
Biological Roles of Liver X Receptors in Immune Cells. Arch Immunol Ther Exp (Warsz) 2012; 60:235-49. [DOI: 10.1007/s00005-012-0179-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Accepted: 03/16/2012] [Indexed: 12/17/2022]
|
29
|
Jakobsson T, Treuter E, Gustafsson JÅ, Steffensen KR. Liver X receptor biology and pharmacology: new pathways, challenges and opportunities. Trends Pharmacol Sci 2012; 33:394-404. [PMID: 22541735 DOI: 10.1016/j.tips.2012.03.013] [Citation(s) in RCA: 245] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Revised: 03/05/2012] [Accepted: 03/26/2012] [Indexed: 01/12/2023]
Abstract
Nuclear receptors (NRs) are master regulators of transcriptional programs that integrate the homeostatic control of almost all biological processes. Their direct mode of ligand regulation and genome interaction is at the core of modern pharmacology. The two liver X receptors LXRα and LXRβ are among the emerging newer drug targets within the NR family. LXRs are best known as nuclear oxysterol receptors and physiological regulators of lipid and cholesterol metabolism that also act in an anti-inflammatory way. Because LXRs control diverse pathways in development, reproduction, metabolism, immunity and inflammation, they have potential as therapeutic targets for diseases as diverse as lipid disorders, atherosclerosis, chronic inflammation, autoimmunity, cancer and neurodegenerative diseases. Recent insights into LXR signaling suggest future targeting strategies aiming at increasing LXR subtype and pathway selectivity. This review discusses the current status of our understanding of LXR biology and pharmacology, with an emphasis on the molecular aspects of LXR signaling that constitute the potential of LXRs as drug targets.
Collapse
Affiliation(s)
- Tomas Jakobsson
- Center for Biosciences, Department of Biosciences and Nutrition, Karolinska Institutet, S-14183 Stockholm, Sweden
| | | | | | | |
Collapse
|
30
|
Cui W, Sun Y, Wang Z, Xu C, Peng Y, Li R. Liver X receptor activation attenuates inflammatory response and protects cholinergic neurons in APP/PS1 transgenic mice. Neuroscience 2012; 210:200-10. [PMID: 22425753 DOI: 10.1016/j.neuroscience.2012.02.047] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2012] [Accepted: 02/25/2012] [Indexed: 01/26/2023]
Abstract
Alzheimer's disease (AD) is associated with beta-amyloid deposition, glial activation, and increased levels of the cytokines, as well as cholinergic dysfunction. Liver X receptor (LXR) has been found to inhibit the expression of pro-inflammatory genes. However, the effects of LXR activation on inflammatory response and on cholinergic system in AD are not yet clear. The present results revealed that LXR activation markedly attenuated several inflammatory markers and decreased microglial activation and reactive astrocytes in amyloid precursor protein (APP)/PS1 transgenic mice. Additionally, LXR activation significantly increased the number of cholinergic neurons in the medial septal regions and the basal nucleus of Meynert (NBM), and attenuated cognitive impairment. Furthermore, we observed that LXR activation inhibited the production of COX-2 and iNOS from Aβ(25-35)-induced microglia. LXR activation and nuclear factor kappa B (NF-κB) inhibitor PDTC both attenuated Aβ(25-35) induction of NF-κB activation. These results suggest that LXR agonists suppress the production of pro-inflammatory molecules, at least in part, by modulating NF-κB-signaling pathway. Collectively, these studies suggest that LXR agonists may have therapeutic significance in AD.
Collapse
Affiliation(s)
- W Cui
- Department of Human Anatomy, Xinxiang Medical University, Xinxiang 453003, China
| | | | | | | | | | | |
Collapse
|
31
|
König R, Stillfried M, Aperdannier P, Clarner T, Beyer C, Kipp M, Mey J. Expression of retinoid X receptor beta is induced in astrocytes during corpus callosum demyelination. J Chem Neuroanat 2012; 43:120-32. [DOI: 10.1016/j.jchemneu.2012.01.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Revised: 01/16/2012] [Accepted: 01/16/2012] [Indexed: 10/14/2022]
|
32
|
Liver X receptor α activation with the synthetic ligand T0901317 reduces lung injury and inflammation after hemorrhage and resuscitation via inhibition of the nuclear factor κB pathway. Shock 2011; 35:367-74. [PMID: 20926989 DOI: 10.1097/shk.0b013e3181f7d742] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Liver X receptor α (LXRα) is a nuclear transcription factor that regulates lipid metabolism. Recently, it has been shown that activation of LXRα with synthetic ligands has anti-inflammatory effects in atherosclerosis and chemical-induced dermatitis. We investigated the effect of the LXRα agonist, T0901317, on lung inflammation in a rodent model of hemorrhagic shock. Hemorrhagic shock was induced in male rats by withdrawing blood to a goal mean arterial blood pressure of 50 mmHg. Blood pressure was maintained at this level for 3 h, at which point rats were rapidly resuscitated with shed blood. Animals were then treated with T0901317 (50 mg · kg) or vehicle i.p. and sacrificed at 1, 2, and 3 h after resuscitation. Treatment with T0901317 significantly improved the cardiac and stroke volume indices as well as the heart rate of rats during the resuscitation period as compared with vehicle-treated rats. The T0901317-treated animals showed significant improvement in the plasma level of lactate, whereas base deficit and bicarbonate levels both trended toward improvement. The T0901317-treated animals also showed lower levels of plasma cytokines and chemokines monocyte chemoattractant protein 1, macrophage inflammatory protein 1α, TNF-α, KC, and IL-6. Lung injury and neutrophil infiltration were reduced by treatment with T0901317, as evaluated by histology and myeloperoxidase assay. At molecular analysis, treatment with T0901317 increased nuclear LXRα expression and DNA binding while also inhibiting activation of nuclear factor κB, a proinflammatory transcription factor, in the lung. Thus, our data suggest that LXRα is an important modulator of the inflammatory response and lung injury after severe hemorrhagic shock, likely through the inhibition of the nuclear factor κB pathway.
Collapse
|
33
|
Loane DJ, Washington PM, Vardanian L, Pocivavsek A, Hoe HS, Duff KE, Cernak I, Rebeck GW, Faden AI, Burns MP. Modulation of ABCA1 by an LXR agonist reduces β-amyloid levels and improves outcome after traumatic brain injury. J Neurotrauma 2011; 28:225-36. [PMID: 21175399 PMCID: PMC3037807 DOI: 10.1089/neu.2010.1595] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Traumatic brain injury (TBI) increases brain beta-amyloid (Aβ) in humans and animals. Although the role of Aβ in the injury cascade is unknown, multiple preclinical studies have demonstrated a correlation between reduced Aβ and improved outcome. Therefore, therapeutic strategies that enhance Aβ clearance may be beneficial after TBI. Increased levels of ATP-binding cassette A1 (ABCA1) transporters can enhance Aβ clearance through an apolipoprotein E (apoE)-mediated pathway. By measuring Aβ and ABCA1 after experimental TBI in C57BL/6J mice, we found that Aβ peaked early after injury (1-3 days), whereas ABCA1 had a delayed response (beginning at 3 days). As ABCA1 levels increased, Aβ levels returned to baseline levels-consistent with the known role of ABCA1 in Aβ clearance. To test if enhancing ABCA1 levels could block TBI-induced Aβ, we treated TBI mice with the liver X-receptor (LXR) agonist T0901317. Pre- and post-injury treatment increased ABCA1 levels at 24 h post-injury, and reduced the TBI-induced increase in Aβ. This reduction in Aβ was not due to decreased amyloid precursor protein processing, or a shift in the solubility of Aβ, indicating enhanced clearance. T0901317 also limited motor coordination deficits in injured mice and reduced brain lesion volume. These data indicate that activation of LXR can reduce Aβ accumulation after TBI, and is accompanied by improved functional recovery.
Collapse
Affiliation(s)
- David J. Loane
- Department of Anesthesiology and Center for Shock Trauma and Anesthesiology Research, National Study Center for Trauma and EMS, University of Maryland School of Medicine, Baltimore, Maryland
| | | | - Lilit Vardanian
- Department of Neuroscience, Georgetown University Medical Center, Washington, D.C
| | - Ana Pocivavsek
- Department of Neuroscience, Georgetown University Medical Center, Washington, D.C
| | - Hyang-Sook Hoe
- Department of Neuroscience, Georgetown University Medical Center, Washington, D.C
| | - Karen E. Duff
- Department of Pathology, Taub Institute for Alzheimer Disease Research, and Integrative Neuroscience New York State Psychiatric Institute, Columbia University Medical Center, New York, New York
| | - Ibolja Cernak
- Johns Hopkins University, Applied Physics Laboratory, Laurel, Maryland
| | - G. William Rebeck
- Department of Neuroscience, Georgetown University Medical Center, Washington, D.C
| | - Alan I. Faden
- Department of Anesthesiology and Center for Shock Trauma and Anesthesiology Research, National Study Center for Trauma and EMS, University of Maryland School of Medicine, Baltimore, Maryland
| | - Mark P. Burns
- Department of Neuroscience, Georgetown University Medical Center, Washington, D.C
| |
Collapse
|
34
|
Calabrese V, Cornelius C, Dinkova-Kostova AT, Calabrese EJ, Mattson MP. Cellular stress responses, the hormesis paradigm, and vitagenes: novel targets for therapeutic intervention in neurodegenerative disorders. Antioxid Redox Signal 2010; 13:1763-811. [PMID: 20446769 PMCID: PMC2966482 DOI: 10.1089/ars.2009.3074] [Citation(s) in RCA: 634] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2009] [Revised: 04/27/2010] [Accepted: 05/01/2010] [Indexed: 12/22/2022]
Abstract
Despite the capacity of chaperones and other homeostatic components to restore folding equilibrium, cells appear poorly adapted for chronic oxidative stress that increases in cancer and in metabolic and neurodegenerative diseases. Modulation of endogenous cellular defense mechanisms represents an innovative approach to therapeutic intervention in diseases causing chronic tissue damage, such as in neurodegeneration. This article introduces the concept of hormesis and its applications to the field of neuroprotection. It is argued that the hormetic dose response provides the central underpinning of neuroprotective responses, providing a framework for explaining the common quantitative features of their dose-response relationships, their mechanistic foundations, and their relationship to the concept of biological plasticity, as well as providing a key insight for improving the accuracy of the therapeutic dose of pharmaceutical agents within the highly heterogeneous human population. This article describes in mechanistic detail how hormetic dose responses are mediated for endogenous cellular defense pathways, including sirtuin and Nrf2 and related pathways that integrate adaptive stress responses in the prevention of neurodegenerative diseases. Particular attention is given to the emerging role of nitric oxide, carbon monoxide, and hydrogen sulfide gases in hormetic-based neuroprotection and their relationship to membrane radical dynamics and mitochondrial redox signaling.
Collapse
|
35
|
Systemic but not local administration of retinoic acid reduces early transcript levels of pro-inflammatory cytokines after experimental spinal cord injury. Neurosci Lett 2010; 485:21-5. [PMID: 20736048 DOI: 10.1016/j.neulet.2010.08.051] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2010] [Revised: 08/13/2010] [Accepted: 08/16/2010] [Indexed: 11/21/2022]
Abstract
Early rises of pro-inflammatory cytokines play a key role in tissue damage and has detrimental consequences for functional outcome after spinal cord injury (SCI). All-trans retinoic acid (RA) has been shown to be a therapeutic agent reducing cytokine expression in vitro, but its use may be limited due to adverse side effects associated with systemic delivery. Local delivery of RA may circumvent adverse side effects, but may simultaneously reduce the therapeutic benefits of the therapy. Here, we investigated whether local or systemic RA treatment differentially affected pro-inflammatory cytokine expression early after rat SCI. Pro-inflammatory cytokines IL-1β, IL-6 and TNFα were investigated at 6h after moderate contusion injury of the thoracic (T9) spinal cord, when mRNA levels are known to peak. Rats were either treated with intrathecal RA (0, 2.5, 10, or 100ng) or received an intraperitoneal injection of RA (15mg/kg bodyweight). Surprisingly intrathecal RA up to amounts of 100ng did not attenuate SCI-induced increases in gene-expression of pro-inflammatory cytokines. In contrast, intraperitoneal RA rendered a 60%, 35% and 58% reduction of IL-1β, IL-6 and TNFα mRNA levels, respectively. Although local doses higher than 100ng RA may reduce pro-inflammatory cytokine gene-expression, such doses precipitate and possibly increase risks of adverse side effects. We conclude that in contrast to systemic delivery, intrathecal administration of RA up to doses of 100ng is ineffective in reducing early pro-inflammatory cytokine gene-expression. Future studies are required to investigate the effects of single intraperitoneal RA treatment on long-term SCI outcome.
Collapse
|