1
|
Yuan H, Yi N, Li D, Xu C, Yin GR, Zhuang C, Wang YJ, Ni S. PPARγ regulates osteoarthritis chondrocytes apoptosis through caspase-3 dependent mitochondrial pathway. Sci Rep 2024; 14:11237. [PMID: 38755283 PMCID: PMC11099036 DOI: 10.1038/s41598-024-62116-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 05/14/2024] [Indexed: 05/18/2024] Open
Abstract
Osteoarthritis (OA) is the most prevalent form of arthritis, characterized by a complex pathogenesis. One of the key factors contributing to its development is the apoptosis of chondrocytes triggered by oxidative stress. Involvement of peroxisome proliferator-activated receptor gamma (PPARγ) has been reported in the regulation of oxidative stress. However, there remains unclear mechanisms that through which PPARγ influences the pathogenesis of OA. The present study aims to delve into the role of PPARγ in chondrocytes apoptosis induced by oxidative stress in the context of OA. Primary human chondrocytes, both relatively normal and OA, were isolated and cultured for the following study. Various assessments were performed, including measurements of cell proliferation, viability and cytotoxicity. Additionally, we examined cell apoptosis, levels of reactive oxygen species (ROS), nitric oxide (NO), mitochondrial membrane potential (MMP) and cytochrome C release. We also evaluated the expression of related genes and proteins, such as collagen type II (Col2a1), aggrecan, inducible nitric oxide synthase (iNOS), caspase-9, caspase-3 and PPARγ. Compared with relatively normal cartilage, the expression of PPARγ in OA cartilage was down-regulated. The proliferation of OA chondrocytes decreased, accompanied by an increase in the apoptosis rate. Down-regulation of PPARγ expression in OA chondrocytes coincided with an up-regulation of iNOS expression, leading to increased secretion of NO, endogenous ROS production, and decrease of MMP levels. Furthermore, we observed the release of cytochrome C, elevated caspase-9 and caspase-3 activities, and reduction of the components of extracellular matrix (ECM) Col2a1 and aggrecan. Accordingly, utilization of GW1929 (PPARγ Agonists) or Z-DEVD-FMK (caspase-3 inhibitor) can protect chondrocytes from mitochondrial-related apoptosis and alleviate the progression of OA. During the progression of OA, excessive oxidative stress in chondrocytes leads to apoptosis and ECM degradation. Activation of PPARγ can postpone OA by down-regulating caspase-3-dependent mitochondrial apoptosis pathway.
Collapse
Affiliation(s)
- Hang Yuan
- Graduate School of Bengbu Medical College, Bengbu, China
- Department of Orthopedics, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China
- Laboratory of Clinical Orthopedics, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China
| | - Ning Yi
- Department of Orthopedics, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China
- Laboratory of Clinical Orthopedics, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China
- Graduate School of Dalian Medical University, Dalian, China
| | - Dong Li
- Department of Orthopedics, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China
- Laboratory of Clinical Orthopedics, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China
| | - Chao Xu
- Department of Orthopedics, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China
- Laboratory of Clinical Orthopedics, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China
| | - Guang-Rong Yin
- Department of Orthopedics, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China
- Laboratory of Clinical Orthopedics, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China
| | - Chao Zhuang
- Graduate School of Bengbu Medical College, Bengbu, China.
- Department of Orthopedics, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China.
- Laboratory of Clinical Orthopedics, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China.
| | - Yu-Ji Wang
- Department of Orthopedics, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China.
- Laboratory of Clinical Orthopedics, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China.
| | - Su Ni
- Bone Disease Research and Clinical Rehabilitation Center, Changzhou Medical Center, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China.
| |
Collapse
|
2
|
Rayner MLD, Kellaway SC, Kingston I, Guillemot-Legris O, Gregory H, Healy J, Phillips JB. Exploring the Nerve Regenerative Capacity of Compounds with Differing Affinity for PPARγ In Vitro and In Vivo. Cells 2022; 12:cells12010042. [PMID: 36611836 PMCID: PMC9818498 DOI: 10.3390/cells12010042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/09/2022] [Accepted: 12/10/2022] [Indexed: 12/24/2022] Open
Abstract
Damage to peripheral nerves can cause debilitating consequences for patients such as lifelong pain and disability. At present, no drug treatments are routinely given in the clinic following a peripheral nerve injury (PNI) to improve regeneration and remyelination of damaged nerves. Appropriately targeted therapeutic agents have the potential to be used at different stages following nerve damage, e.g., to maintain Schwann cell viability, induce and sustain a repair phenotype to support axonal growth, or promote remyelination. The development of therapies to promote nerve regeneration is currently of high interest to researchers, however, translation to the clinic of drug therapies for PNI is still lacking. Studying the effect of PPARγ agonists for treatment of peripheral nerve injures has demonstrated significant benefits. Ibuprofen, a non-steroidal anti-inflammatory drug (NSAID), has reproducibly demonstrated benefits in vitro and in vivo, suggested to be due to its agonist action on PPARγ. Other NSAIDs have demonstrated differing levels of PPARγ activation based upon their affinity. Therefore, it was of interest to determine whether affinity for PPARγ of selected drugs corresponded to an increase in regeneration. A 3D co-culture in vitro model identified some correlation between these two properties. However, when the drug treatments were screened in vivo, in a crush injury model in a rat sciatic nerve, the same correlation was not apparent. Further differences were observed between capacity to increase axon number and improvement in functional recovery. Despite there not being a clear correlation between affinity and size of effect on regeneration, all selected PPARγ agonists improved regeneration, providing a panel of compounds that could be explored for use in the treatment of PNI.
Collapse
Affiliation(s)
- Melissa L. D. Rayner
- Department of Pharmacology, School of Pharmacy, University College London, London WC1N 1AX, UK
- Centre for Nerve Engineering, University College London, London WC1N 6BT, UK
- Correspondence:
| | - Simon C. Kellaway
- Department of Pharmacology, School of Pharmacy, University College London, London WC1N 1AX, UK
- Centre for Nerve Engineering, University College London, London WC1N 6BT, UK
| | - Isabel Kingston
- Department of Pharmacology, School of Pharmacy, University College London, London WC1N 1AX, UK
- Centre for Nerve Engineering, University College London, London WC1N 6BT, UK
| | - Owein Guillemot-Legris
- Department of Pharmacology, School of Pharmacy, University College London, London WC1N 1AX, UK
- Centre for Nerve Engineering, University College London, London WC1N 6BT, UK
| | - Holly Gregory
- Department of Pharmacology, School of Pharmacy, University College London, London WC1N 1AX, UK
- Centre for Nerve Engineering, University College London, London WC1N 6BT, UK
| | - Jess Healy
- Centre for Nerve Engineering, University College London, London WC1N 6BT, UK
| | - James B. Phillips
- Department of Pharmacology, School of Pharmacy, University College London, London WC1N 1AX, UK
- Centre for Nerve Engineering, University College London, London WC1N 6BT, UK
| |
Collapse
|
3
|
Li H, Yuan W, Chen Y, Lin B, Wang S, Deng Z, Zheng Q, Li Q. Transcription and proteome changes involved in re-innervation muscle following nerve crush in rats. BMC Genomics 2022; 23:666. [PMID: 36131238 PMCID: PMC9494802 DOI: 10.1186/s12864-022-08895-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 09/16/2022] [Indexed: 02/05/2023] Open
Abstract
Severe peripheral nerve injury leads to the irreparable disruption of nerve fibers. This leads to disruption of synapses with the designated muscle, which consequently go through progressive atrophy and damage of muscle function. The molecular mechanism that underlies the re-innervation process has yet to be evaluated using proteomics or transcriptomics. In the present study, multi-dimensional data were therefore integrated with transcriptome and proteome profiles in order to investigate the mechanism of re-innervation in muscles. Two simulated nerve injury muscle models in the rat tibial nerve were compared: the nerve was either cut (denervated, DN group) or crushed but with the nerve sheath intact (re-innervated, RN group). The control group had a preserved and intact tibial nerve. At 4 weeks, the RN group showed better tibial nerve function and recovery of muscle atrophy compared to the DN group. As the high expression of Myh3, Postn, Col6a1 and Cfi, the RN group demonstrated superior re-innervation as well. Both differentially expressed genes (DEGs) and proteins (DEPs) were enriched in the peroxisome proliferator-activated receptors (PPARs) signaling pathway, as well as the energy metabolism. This study provides basic information regarding DEGs and DEPs during re-innervation-induced muscle atrophy. Furthermore, the crucial genes and proteins can be detected as possible treatment targets in the future.
Collapse
Affiliation(s)
- Haotao Li
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106, Zhongshan Road, Yuexiu District, Guangzhou, People's Republic of China
- Shantou University Medical College, Shantou, People's Republic of China
| | - Wanqiong Yuan
- Department of Orthopedics, Peking University Third Hospital, Beijing, People's Republic of China
- Beijing Key Laboratory of Spinal Disease, Beijing, People's Republic of China
- Engineering Research Center of Bone and Joint Precision Medicine, Beijing, People's Republic of China
| | - Yijian Chen
- Department of Orthopedics, Shantou Central Hospital, Shantou, Guangdong, People's Republic of China
| | - Bofu Lin
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106, Zhongshan Road, Yuexiu District, Guangzhou, People's Republic of China
- Shantou University Medical College, Shantou, People's Republic of China
| | - Shuai Wang
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106, Zhongshan Road, Yuexiu District, Guangzhou, People's Republic of China
| | - Zhantao Deng
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106, Zhongshan Road, Yuexiu District, Guangzhou, People's Republic of China
| | - Qiujian Zheng
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106, Zhongshan Road, Yuexiu District, Guangzhou, People's Republic of China
| | - Qingtian Li
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106, Zhongshan Road, Yuexiu District, Guangzhou, People's Republic of China.
| |
Collapse
|
4
|
Metzger JM, Matsoff HN, Vu D, Zinnen AD, Jones KM, Bondarenko V, Simmons HA, Moore CF, Emborg ME. Myelin Basic Protein and Cardiac Sympathetic Neurodegeneration in Nonhuman Primates. Neurol Res Int 2021; 2021:4776610. [PMID: 34646580 PMCID: PMC8505074 DOI: 10.1155/2021/4776610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 09/22/2021] [Indexed: 11/17/2022] Open
Abstract
Minimal myelination is proposed to be a contributing factor to the preferential nigral neuronal loss in Parkinson's disease (PD). Similar to nigral dopaminergic neurons, sympathetic neurons innervating the heart have long, thin axons which are unmyelinated or minimally myelinated. Interestingly, cardiac sympathetic loss in PD is heterogeneous across the heart, yet the spatial relationship between myelination and neurodegeneration is unknown. Here, we report the mapping of myelin basic protein (MBP) expression across the left ventricle of normal rhesus macaques (n = 5) and animals intoxicated with systemic 6-OHDA (50 mg/kg iv) to model parkinsonian cardiac neurodegeneration (n = 10). A subset of 6-OHDA-treated rhesus received daily dosing of pioglitazone (5 mg/kg po; n = 5), a PPARγ agonist with neuroprotective properties. In normal animals, MBP-immunoreactivity (-ir) was identified surrounding approximately 14% of axonal fibers within nerve bundles of the left ventricle, with more myelinated nerve fibers at the base level of the left ventricle than the apex (p < 0.014). Greater MBP-ir at the base was related to a greater number of nerve bundles at that level relative to the apex (p < 0.05), as the percent of myelinated nerve fibers in bundles was not significantly different between levels of the heart. Cardiac sympathetic loss following 6-OHDA was associated with decreased MBP-ir in cardiac nerve bundles, with the percent decrease of MBP-ir greater in the apex (84.5%) than the base (52.0%). Interestingly, cardiac regions and levels with more MBP-ir in normal animals showed attenuated sympathetic loss relative to areas with less MBP-ir in 6-OHDA + placebo (r = -0.7, p < 0.014), but not in 6-OHDA + pioglitazone (r = -0.1) subjects. Our results demonstrate that myelination is present around a minority of left ventricle nerve bundle fibers, is heterogeneously distributed in the heart of rhesus macaques, and has a complex relationship with cardiac sympathetic neurodegeneration and neuroprotection.
Collapse
Affiliation(s)
- Jeanette M. Metzger
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Helen N. Matsoff
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI 53715, USA
- Occupational Therapy Program, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Don Vu
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Alexandra D. Zinnen
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Kathryn M. Jones
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Viktoriya Bondarenko
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Heather A. Simmons
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Colleen F. Moore
- Department of Psychology, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Marina E. Emborg
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI 53715, USA
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI 53715, USA
| |
Collapse
|
5
|
Repurposing Small Molecules to Target PPAR-γ as New Therapies for Peripheral Nerve Injuries. Biomolecules 2021; 11:biom11091301. [PMID: 34572514 PMCID: PMC8465622 DOI: 10.3390/biom11091301] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/08/2021] [Accepted: 08/14/2021] [Indexed: 12/21/2022] Open
Abstract
The slow rate of neuronal regeneration that follows peripheral nerve repair results in poor recovery, particularly where reinnervation of muscles is delayed, leading to atrophy and permanent loss of function. There is a clear clinical need to develop drug treatments that can accelerate nerve regeneration safely, restoring connections before the target tissues deteriorate irreversibly. The identification that the Rho/Rho-associated kinase (ROCK) pathway acts to limit neuronal growth rate is a promising advancement towards the development of drugs. Targeting Rho or ROCK directly can act to suppress the activity of this pathway; however, the pathway can also be modulated through the activation of upstream receptors; one of particular interest being peroxisome proliferator-activated receptor gamma (PPAR-γ). The connection between the PPAR-γ receptor and the Rho/ROCK pathway is the suppression of the conversion of inactive guanosine diphosphate (GDP)-Rho to active guanosine triphosphate GTP-Rho, resulting in the suppression of Rho/ROCK activity. PPAR-γ is known for its role in cellular metabolism that leads to cell growth and differentiation. However, more recently there has been a growing interest in targeting PPAR-γ in peripheral nerve injury (PNI). The localisation and expression of PPAR-γ in neural cells following a PNI has been reported and further in vitro and in vivo studies have shown that delivering PPAR-γ agonists following injury promotes nerve regeneration, leading to improvements in functional recovery. This review explores the potential of repurposing PPAR-γ agonists to treat PNI and their prospective translation to the clinic.
Collapse
|
6
|
Willems S, Zaienne D, Merk D. Targeting Nuclear Receptors in Neurodegeneration and Neuroinflammation. J Med Chem 2021; 64:9592-9638. [PMID: 34251209 DOI: 10.1021/acs.jmedchem.1c00186] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Nuclear receptors, also known as ligand-activated transcription factors, regulate gene expression upon ligand signals and present as attractive therapeutic targets especially in chronic diseases. Despite the therapeutic relevance of some nuclear receptors in various pathologies, their potential in neurodegeneration and neuroinflammation is insufficiently established. This perspective gathers preclinical and clinical data for a potential role of individual nuclear receptors as future targets in Alzheimer's disease, Parkinson's disease, and multiple sclerosis, and concomitantly evaluates the level of medicinal chemistry targeting these proteins. Considerable evidence suggests the high promise of ligand-activated transcription factors to counteract neurodegenerative diseases with a particularly high potential of several orphan nuclear receptors. However, potent tools are lacking for orphan receptors, and limited central nervous system exposure or insufficient selectivity also compromises the suitability of well-studied nuclear receptor ligands for functional studies. Medicinal chemistry efforts are needed to develop dedicated high-quality tool compounds for the therapeutic validation of nuclear receptors in neurodegenerative pathologies.
Collapse
Affiliation(s)
- Sabine Willems
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Strasse 9, 60438 Frankfurt, Germany
| | - Daniel Zaienne
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Strasse 9, 60438 Frankfurt, Germany
| | - Daniel Merk
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Strasse 9, 60438 Frankfurt, Germany
| |
Collapse
|
7
|
Peroxisome Proliferator-Activated Receptors as Molecular Links between Caloric Restriction and Circadian Rhythm. Nutrients 2020; 12:nu12113476. [PMID: 33198317 PMCID: PMC7696073 DOI: 10.3390/nu12113476] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 11/04/2020] [Accepted: 11/09/2020] [Indexed: 02/06/2023] Open
Abstract
The circadian rhythm plays a chief role in the adaptation of all bodily processes to internal and environmental changes on the daily basis. Next to light/dark phases, feeding patterns constitute the most essential element entraining daily oscillations, and therefore, timely and appropriate restrictive diets have a great capacity to restore the circadian rhythm. One of the restrictive nutritional approaches, caloric restriction (CR) achieves stunning results in extending health span and life span via coordinated changes in multiple biological functions from the molecular, cellular, to the whole-body levels. The main molecular pathways affected by CR include mTOR, insulin signaling, AMPK, and sirtuins. Members of the family of nuclear receptors, the three peroxisome proliferator-activated receptors (PPARs), PPARα, PPARβ/δ, and PPARγ take part in the modulation of these pathways. In this non-systematic review, we describe the molecular interconnection between circadian rhythm, CR-associated pathways, and PPARs. Further, we identify a link between circadian rhythm and the outcomes of CR on the whole-body level including oxidative stress, inflammation, and aging. Since PPARs contribute to many changes triggered by CR, we discuss the potential involvement of PPARs in bridging CR and circadian rhythm.
Collapse
|
8
|
Peroxisome Proliferator-Activated Receptors and Caloric Restriction-Common Pathways Affecting Metabolism, Health, and Longevity. Cells 2020; 9:cells9071708. [PMID: 32708786 PMCID: PMC7407644 DOI: 10.3390/cells9071708] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/14/2020] [Accepted: 07/14/2020] [Indexed: 02/06/2023] Open
Abstract
Caloric restriction (CR) is a traditional but scientifically verified approach to promoting health and increasing lifespan. CR exerts its effects through multiple molecular pathways that trigger major metabolic adaptations. It influences key nutrient and energy-sensing pathways including mammalian target of rapamycin, Sirtuin 1, AMP-activated protein kinase, and insulin signaling, ultimately resulting in reductions in basic metabolic rate, inflammation, and oxidative stress, as well as increased autophagy and mitochondrial efficiency. CR shares multiple overlapping pathways with peroxisome proliferator-activated receptors (PPARs), particularly in energy metabolism and inflammation. Consequently, several lines of evidence suggest that PPARs might be indispensable for beneficial outcomes related to CR. In this review, we present the available evidence for the interconnection between CR and PPARs, highlighting their shared pathways and analyzing their interaction. We also discuss the possible contributions of PPARs to the effects of CR on whole organism outcomes.
Collapse
|
9
|
The roles played by TLR4 in the pathogenesis of multiple sclerosis; A systematic review article. Immunol Lett 2020; 220:63-70. [PMID: 32032617 DOI: 10.1016/j.imlet.2020.02.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 01/19/2020] [Accepted: 02/03/2020] [Indexed: 12/15/2022]
Abstract
Multiple sclerosis (MS) is a world-wide pro-inflammatory based disease, which is prevalent among young individuals. The etiology of the disease and its related complications are yet to be clarified. It has been hypothesized that environmental factors, including pathogen-associated molecular patterns (PAMPs) and the internal factors such as damage-associated molecular patterns (DAMPs), may be the most important inducers/stimulators of the disorder and its related complications. Previous investigations proved that pathogen recognition receptors (PRRs) are the main sensors for the PAMPs and DAMPs. Therefore, it seems that the PRRs have been considered to be the plausible molecules participating in the etiology of MS. Toll-like receptors (TLRs) have been the widely studied PRRs and their roles have been documented in human-related diseases. TLR4 is the main PRR expressed on the cell surface of several immune cells including macrophages and dendritic cells. Several investigations reported that TLR4 to be the main molecule involved in the pathogenesis of pro-inflammatory based diseases. Thus, it has been hypothesized that TLR4 may be a part of the MS puzzle. This review article discusses the role of TLR4 in the MS pathogenesis using recent in vitro and in vivo investigations.
Collapse
|
10
|
Abstract
OBJECTIVES The aim of this study is to investigate the role of peroxisome proliferator-activated receptor-gamma isoform (PPARγ), in trigeminal neuropathic pain utilizing a novel mouse trigeminal inflammatory compression (TIC) injury model. RESULTS The study determined that the PPARγ nuclear receptor plays a significant role in trigeminal nociception transmission, evidenced by: 1) Intense PPARγ immunoreactivity is expressed 3 weeks after TIC nerve injury in the spinal trigeminal caudalis, the termination site of trigeminal nociceptive nerve fibers. 2) Systemic administration of a PPARγ agonist, pioglitazone (PIO), attenuates whisker pad mechanical allodynia at doses of 300 mg/kg i.p. and 600 mg/kg p.o. 3) Administration of a PPARγ antagonist, GW9662 (30 mg/kg i.p.), prior to providing the optimal dose of PIO (300 mg/kg i.p.) blocked the analgesic effect of PIO. DISCUSSION This is the first study localizing PPARγ immunoreactivity throughout the brainstem trigeminal sensory spinal nucleus (spV) and its increase three weeks after TIC nerve injury. This is also the first study to demonstrate that activation of PPARγ attenuates trigeminal hypersensitivity in the mouse TIC nerve injury model. The findings presented here suggest the possibility of utilizing the FDA approved diabetic treatment drug, PIO, as a new therapeutic that targets PPARγ for treatment of patients suffering from orofacial neuropathic pain.
Collapse
|
11
|
Isaac C, Mauborgne A, Grimaldi A, Ade K, Pohl M, Limatola C, Boucher Y, Demangel C, Guenin-Macé L. Mycolactone displays anti-inflammatory effects on the nervous system. PLoS Negl Trop Dis 2017; 11:e0006058. [PMID: 29149212 PMCID: PMC5693295 DOI: 10.1371/journal.pntd.0006058] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 10/20/2017] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Mycolactone is a macrolide produced by the skin pathogen Mycobacterium ulcerans, with cytotoxic, analgesic and immunomodulatory properties. The latter were recently shown to result from mycolactone blocking the Sec61-dependent production of pro-inflammatory mediators by immune cells. Here we investigated whether mycolactone similarly affects the inflammatory responses of the nervous cell subsets involved in pain perception, transmission and maintenance. We also investigated the effects of mycolactone on the neuroinflammation that is associated with chronic pain in vivo. METHODOLOGY/ PRINCIPLE FINDINGS Sensory neurons, Schwann cells and microglia were isolated from mice for ex vivo assessment of mycolactone cytotoxicity and immunomodulatory activity by measuring the production of proalgesic cytokines and chemokines. In all cell types studied, prolonged (>48h) exposure to mycolactone induced significant cell death at concentrations >10 ng/ml. Within the first 24h treatment, nanomolar concentrations of mycolactone efficiently suppressed the cell production of pro-inflammatory mediators, without affecting their viability. Notably, mycolactone also prevented the pro-inflammatory polarization of cortical microglia. Since these cells critically contribute to neuroinflammation, we next tested if mycolactone impacts this pathogenic process in vivo. We used a rat model of neuropathic pain induced by chronic constriction of the sciatic nerve. Here, mycolactone was injected daily for 3 days in the spinal canal, to ensure its proper delivery to spinal cord. While this treatment failed to prevent injury-induced neuroinflammation, it decreased significantly the local production of inflammatory cytokines without inducing detectable cytotoxicity. CONCLUSION/ SIGNIFICANCE The present study provides in vitro and in vivo evidence that mycolactone suppresses the inflammatory responses of sensory neurons, Schwann cells and microglia, without affecting the cell viability. Together with previous studies using peripheral blood leukocytes, our work implies that mycolactone-mediated analgesia may, at least partially, be explained by its anti-inflammatory properties.
Collapse
Affiliation(s)
- Caroline Isaac
- Institut Pasteur, Unité d’Immunobiologie de l’Infection, Paris, France
- INSERM U1221, Paris, France
| | - Annie Mauborgne
- Centre de Psychiatrie et Neurosciences, Inserm U894, Paris, France
| | - Alfonso Grimaldi
- Pasteur Institute Rome, Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Kemy Ade
- Institut Pasteur, Unité d’Immunobiologie de l’Infection, Paris, France
- INSERM U1221, Paris, France
| | - Michel Pohl
- Centre de Psychiatrie et Neurosciences, Inserm U894, Paris, France
| | - Cristina Limatola
- Pasteur Institute Rome, Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
- IRCCS Neuromed, Pozzilli, Italy
| | - Yves Boucher
- Centre de Psychiatrie et Neurosciences, Inserm U894, Paris, France
- Groupe Hospitalier Pitié Salpétrière, UFR Odontologie Université Paris Diderot, Paris, France
| | - Caroline Demangel
- Institut Pasteur, Unité d’Immunobiologie de l’Infection, Paris, France
- INSERM U1221, Paris, France
| | - Laure Guenin-Macé
- Institut Pasteur, Unité d’Immunobiologie de l’Infection, Paris, France
- INSERM U1221, Paris, France
- * E-mail:
| |
Collapse
|
12
|
Yuan J, Ge H, Liu W, Zhu H, Chen Y, Zhang X, Yang Y, Yin Y, Chen W, Wu W, Yang Y, Lin J. M2 microglia promotes neurogenesis and oligodendrogenesis from neural stem/progenitor cells via the PPARγ signaling pathway. Oncotarget 2017; 8:19855-19865. [PMID: 28423639 PMCID: PMC5386728 DOI: 10.18632/oncotarget.15774] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 01/23/2017] [Indexed: 12/20/2022] Open
Abstract
Neural stem/progenitor cells (NSPCs) are an important source of cells for cell replacement therapy after nerve injury. How to induce NSPCs differentiation towards neurons and oligodendrocytes is a challenging issue in neuroscience research. In the present study, we polarized microglia into M1 and M2 phenotype, used their supernatants to induce NSPCs differentiation, and investigated the effects of different microglia phenotypes on NSPCs differentiation and their mechanisms. We discovered that, after exposure to M1 phenotype supernatant, NSPCs differentiated into fewer Tuj-1+ and Olig2+ cells, but more GFAP+ cells. Meanwhile, a significantly increased number of Tuj-1+ and Olig2+ cells and smaller number of GFAP+ cells were generated by M2 microglia supernatant-induced NSPCs differentiation. We also observed that 15d-PGJ2, an endogenous ligand of PPARγ, was elevated in M2 phenotype supernatant and could activate PPARγ expression in NSPCs, whereas use of the PPARγ inhibitor GW9662, could reduce the percentage of differentiated neurons and oligodendrocytes. Our study results confirm that M2 microglia supernatant can activate the PPARγ signaling pathway and promote neurogenesis and oligodendrogenesis from NSPCs differentiation. The present study provides a further theoretical basis for induction of NSPCs oriented differentiation.
Collapse
Affiliation(s)
- Jichao Yuan
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University, Chongqing 400038, China.,Department of Neurology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Hongfei Ge
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Wei Liu
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Haitao Zhu
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Yaxing Chen
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Xuan Zhang
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Yang Yang
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Yi Yin
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Weixiang Chen
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Wanjiang Wu
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Yunfeng Yang
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Jiangkai Lin
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| |
Collapse
|
13
|
Li J, Csakai A, Jin J, Zhang F, Yin H. Therapeutic Developments Targeting Toll-like Receptor-4-Mediated Neuroinflammation. ChemMedChem 2016; 11:154-65. [PMID: 26136385 PMCID: PMC4983275 DOI: 10.1002/cmdc.201500188] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Indexed: 02/06/2023]
Abstract
Toll-like receptors (TLRs) have been shown to play an important role in the immune system, which warrants study of their remarkable potential as pharmacological targets. Activation of TLRs requires participation from specific pathogen-associated molecular patterns (PAMPs) and accessory proteins such as myeloid differentiation protein 2 (MD2), lipopolysaccharide binding protein (LBP), and cluster differentiation antigen 14 (CD14). Assembly of the TLR4-MD2-LPS complex is essential in TLR4 activation. Recent studies have revealed that TLR4 activation is a significant trigger of signal transmission pathways in the nervous system, which could result in chronic pain as well as opioid tolerance and dependence. Researchers of the molecular structure of TLRs and their accessory proteins have opened a door to syntheses of TLRs agonists and antagonists, such as eritoran. Small-molecule modulators of TLR4, such as MD2-I and tricyclic antidepressants, offer more promising prospects than peptides, given their convenience in oral administration and lower cost. Herein we mainly discuss the mechanisms and clinical prospects of TLR4 agonists and antagonists.
Collapse
Affiliation(s)
- Jing Li
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100032, China
| | - Adam Csakai
- Department of Chemistry and Biochemistry and BioFrontiers Institute, University of Colorado, Boulder, CO, 80309-0596, USA
| | - Jialin Jin
- Center of Basic Molecular Science, Department of Chemistry, Tsinghua University, Beijing, 100082, China
- Physikalisch-Astronomische Fakultät, Abbe School of Photonics, Jena, 07743, Germany
| | - Fengchun Zhang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100032, China.
| | - Hang Yin
- Department of Chemistry and Biochemistry and BioFrontiers Institute, University of Colorado, Boulder, CO, 80309-0596, USA.
- Center of Basic Molecular Science, Department of Chemistry, Tsinghua University, Beijing, 100082, China.
| |
Collapse
|
14
|
Darehgazani R, Peymani M, Hashemi MS, Omrani MD, Movafagh A, Ghaedi K, Nasr-Esfahani MH. PPARγ ameliorated LPS induced inflammation of HEK cell line expressing both human Toll-like receptor 4 (TLR4) and MD2. Cytotechnology 2015. [PMID: 26224481 DOI: 10.1007/s10616-015-9893-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
TLR4 is transmembrane pattern-recognition receptor that initiates signals in response to diverse pathogen-associated molecular patterns especially LPS. Recently, there have been an increasing number of studies about the role of TLRs in the pathogenesis of several disorders as well as the therapeutic potential of TLR intervention in such diseases. Peroxisome proliferator-activated receptor-gamma (PPARγ) is a ligand-activated transcription factor with numerous biological effects. PPARγ has been shown to exert a potential anti-inflammatory effect through suppression of TLR4-mediated inflammation. Therefore, PPARγ agonists may have a potential to combat inflammatory conditions in pathologic states. The current study aims to show the decrease of inflammation by overexpression of PPARγ in a cell reporter model. To reach this goal, recombinant pBudCE4.1 (+) containing encoding sequences of human TLR4 and MD2 was constructed and used to transfect HEK cells. Subsequently, inflammation was induced by LPS treatment as control group. In the treatment group, overexpression of PPARγ prior to inflammation was performed and the expression of inflammatory markers was assessed in this condition. The expression of inflammatory markers (TNFα and iNOS) was defined by quantitative real time PCR and the amount of phosphorylated NF-κB was measured by western blot. Data indicated expression of TNFα and iNOS increased in LPS induced inflammation of stably transformed HEK cells with MD2 and TLR4. In this cell reporter model overexpression of PPARγ dramatically prevented LPS-induced inflammation through the blocking of TLR4/NF-κB signaling. PPARγ was shown to negatively regulate TLR4 activity and therefore exerts its anti-inflammatory action against LPS induced inflammation.
Collapse
Affiliation(s)
- Reyhaneh Darehgazani
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Velenjak, 1985717443, Tehran, Iran.,Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, Royan Street, Salman Street, Khorsagan, 816513-1378, Isfahan, Iran
| | - Maryam Peymani
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, Royan Street, Salman Street, Khorsagan, 816513-1378, Isfahan, Iran.,Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Sahrekord, Iran
| | - Motahare-Sadat Hashemi
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, Royan Street, Salman Street, Khorsagan, 816513-1378, Isfahan, Iran
| | - Mir Davood Omrani
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Velenjak, 1985717443, Tehran, Iran.
| | - Abolfazl Movafagh
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Velenjak, 1985717443, Tehran, Iran
| | - Kamran Ghaedi
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, Royan Street, Salman Street, Khorsagan, 816513-1378, Isfahan, Iran. .,Biology Department, School of Sciences, University of Isfahan, Isfahan, Iran.
| | - Mohammad Hossein Nasr-Esfahani
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, Royan Street, Salman Street, Khorsagan, 816513-1378, Isfahan, Iran.
| |
Collapse
|
15
|
PPAR-α agonist fenofibrate protects against the damaging effects of MPTP in a rat model of Parkinson's disease. Prog Neuropsychopharmacol Biol Psychiatry 2014; 53:35-44. [PMID: 24593945 DOI: 10.1016/j.pnpbp.2014.02.009] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2013] [Revised: 02/18/2014] [Accepted: 02/21/2014] [Indexed: 01/23/2023]
Abstract
Parkinson's disease (PD) is a chronic neurodegenerative disorder characterized by progressive loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc). The etiology and pathogenesis of PD are still unknown, however, many evidences suggest a prominent role of oxidative stress, inflammation, apoptosis, mitochondrial dysfunction and proteosomal dysfunction. The peroxisome proliferator-activated receptor (PPAR) ligands, a member of the nuclear receptor family, have anti-inflammatory activity over a variety of rodent's models for acute and chronic inflammation. PPAR-α agonists, a subtype of the PPAR receptors, such as fenofibrate, have been shown a major role in the regulation of inflammatory processes. Animal models of PD have shown that neuroinflammation is one of the most important mechanisms involved in dopaminergic cell death. In addition, anti-inflammatory drugs are able to attenuate toxin-induced parkinsonism. In this study we evaluated the effects of oral administration of fenofibrate 100mg/kg 1h after infusion of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) in the SNpc. First, we assessed the motor behavior in the open field for 24h, 7, 14 and 21 days after MPTP. Twenty-two days after surgery, the animals were tested for two-way active avoidance and forced swimming for evaluation regarding cognitive and depressive parameters, respectively. Twenty-three days after infusion of the toxin, we quantified DA and turnover and evaluated oxidative stress through the measurement of GSH (glutathione peroxidase), SOD (superoxide dismutase) and LOOH (hydroperoxide lipid). The data show that fenofibrate was able to decrease hypolocomotion caused by MPTP 24h after injury, depressive-like behavior 22 days after the toxin infusion, and also protected against decreased level of DA and excessive production of reactive oxygen species (ROS) 23 days after surgery. Thus, fenofibrate has shown a neuroprotective effect in the MPTP model of Parkinson's disease.
Collapse
|
16
|
Lactobacillus casei Shirota protects from fructose-induced liver steatosis: a mouse model. J Nutr Biochem 2012; 24:531-8. [PMID: 22749137 DOI: 10.1016/j.jnutbio.2012.01.014] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Revised: 01/23/2012] [Accepted: 01/27/2012] [Indexed: 02/06/2023]
Abstract
To test the hypothesis that Lactobacillus casei Shirota (Lcs) protects against the onset of non-alcoholic fatty liver disease (NAFLD) in a mouse model of fructose-induced steatosis, C57BL/6J mice were either fed tap water or 30% fructose solution +/- Lcs for 8 weeks. Chronic consumption of 30% fructose solution led to a significant increase in hepatic steatosis as well as plasma alanine-aminotransferase (ALT) levels, which was attenuated by treatment with Lcs. Protein levels of the tight junction protein occludin were found to be markedly lower in both fructose treated groups in the duodenum, whereas microbiota composition in this part of the intestine was not affected. Lcs treatment markedly attenuated the activation of the Toll-like receptor (TLR) 4 signalling cascade found in the livers of mice only treated with fructose. Moreover, in livers of fructose fed mice treated with Lcs peroxisome proliferator-activated receptor (PPAR)-γ activity was markedly higher than in mice only fed fructose. Taken together, the results of the present study suggest that the dietary intake of Lcs protects against the onset of fructose-induced NAFLD through mechanisms involving an attenuation of the TLR-4-signalling cascade in the liver.
Collapse
|
17
|
Polvani S, Tarocchi M, Galli A. PPARγ and Oxidative Stress: Con(β) Catenating NRF2 and FOXO. PPAR Res 2012; 2012:641087. [PMID: 22481913 PMCID: PMC3317010 DOI: 10.1155/2012/641087] [Citation(s) in RCA: 185] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Revised: 11/05/2011] [Accepted: 11/17/2011] [Indexed: 12/22/2022] Open
Abstract
Peroxisome-proliferator activator receptor γ (PPARγ) is a nuclear receptor of central importance in energy homeostasis and inflammation. Recent experimental pieces of evidence demonstrate that PPARγ is implicated in the oxidative stress response, an imbalance between antithetic prooxidation and antioxidation forces that may lead the cell to apoptotic or necrotic death. In this delicate and intricate game of equilibrium, PPARγ stands out as a central player devoted to the quenching and containment of the damage and to foster cell survival. However, PPARγ does not act alone: indeed the nuclear receptor is at the point of interconnection of various pathways, such as the nuclear factor erythroid 2-related factor 2 (NRF2), Wnt/β-catenin, and forkhead box proteins O (FOXO) pathways. Here we reviewed the role of PPARγ in response to oxidative stress and its interaction with other signaling pathways implicated in this process, an interaction that emerged as a potential new therapeutic target for several oxidative-related diseases.
Collapse
Affiliation(s)
- Simone Polvani
- Gastroenterology Unit, Department of Clinical Pathophysiology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy
| | - Mirko Tarocchi
- Gastroenterology Unit, Department of Clinical Pathophysiology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy
| | - Andrea Galli
- Gastroenterology Unit, Department of Clinical Pathophysiology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy
| |
Collapse
|
18
|
Davanian H, Båge T, Lindberg J, Lundeberg J, Q. Concha H, Sällberg Chen M, Yucel-Lindberg T. Signaling pathways involved in the regulation of TNFα-induced toll-like receptor 2 expression in human gingival fibroblasts. Cytokine 2012; 57:406-16. [DOI: 10.1016/j.cyto.2011.12.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2011] [Revised: 11/16/2011] [Accepted: 12/12/2011] [Indexed: 11/25/2022]
|
19
|
Changes of peroxisome proliferator-activated receptor-γ on crushed rat sciatic nerves and differentiated primary Schwann cells. J Mol Neurosci 2011; 47:380-8. [PMID: 22094441 DOI: 10.1007/s12031-011-9662-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2011] [Accepted: 09/30/2011] [Indexed: 12/14/2022]
Abstract
Peroxisome proliferator-activated receptor-γ (PPAR-γ) has been found to play an essential role in cell proliferation, but whether it was involved in Schwann cells differentiation has never been studied. We have found in sciatic nerve injury that expression of PPAR-γ decreases mainly in Schwann cells, and it was also increased in differentiated Schwann cells. Further, activated PPAR-γ by the endogenous ligand 15 d-PGJ(2) increased expressions of PPAR-γ level and Schwann cell differentiation, and this effect may be protected by its antagonist GDW9662. These results indicate that PPAR-γ could promote Schwann cell differentiation, which plays an important role in peripheral nerve injury and regeneration.
Collapse
|
20
|
Wang P, Zhu F, Konstantopoulos K. Interleukin-6 synthesis in human chondrocytes is regulated via the antagonistic actions of prostaglandin (PG)E2 and 15-deoxy-Δ(12,14)-PGJ2. PLoS One 2011; 6:e27630. [PMID: 22096605 PMCID: PMC3214064 DOI: 10.1371/journal.pone.0027630] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Accepted: 10/20/2011] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Elevated levels of interleukin-6 (IL-6), prostaglandin (PG)E(2), PGD(2) and its dehydration end product 15-deoxy-Δ(12,14)-PGJ(2) (15d-PGJ(2)) have been detected in joint synovial fluids from patients with rheumatoid arthritis (RA). PGE(2) directly stimulates IL-6 production in human articular chondrocytes. However, the effects of PGD(2) and 15d-PGJ(2) in the absence or presence of PGE(2) on IL-6 synthesis in human chondrocytes have yet to be determined. It is believed that dysregulated overproduction of IL-6 is responsible for the systemic inflammatory manifestations and abnormal laboratory findings in RA patients. METHODOLOGY/PRINCIPAL FINDINGS Using the T/C-28a2 chondrocyte cell line as a model system, we report that exogenous PGE(2) and PGD(2)/15d-PGJ(2) exert antagonistic effects on IL-6 synthesis in human T/C-28a2 chondrocytes. Using a synthesis of sophisticated molecular biology techniques, we determined that PGE(2) stimulates Toll-like receptor 4 (TLR4) synthesis, which is in turn responsible for the activation of the ERK1/2, PI3K/Akt and PKA/CREB pathways that phosphorylate the NF-κB p65 subunit leading to NF-κB activation. Binding of the activated NF-κB p65 subunit to IL-6 promoter induces IL-6 synthesis in human T/C28a2 chondrocytes. PGD(2) or 15d-PGJ(2) concurrently downregulates TLR4 and upregulates caveolin-1, which in turn inhibit the PGE(2)-dependent ERK1/2, PI3-K and PKA activation, and ultimately with NF-κB-dependent IL-6 synthesis in chondrocytes. CONCLUSIONS/SIGNIFICANCE We have delineated the signaling cascade by which PGE(2) and PGD(2)/15d-PGJ(2) exert opposing effects on IL-6 synthesis in human chondrocytes. Elucidation of the molecular pathway of IL-6 synthesis and secretion by chondrocytes will provide insights for developing strategies to reduce inflammation and pain in RA patients.
Collapse
Affiliation(s)
- Pu Wang
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Fei Zhu
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Konstantinos Konstantopoulos
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland, United States of America
- Johns Hopkins Physical Sciences in Oncology Center and Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, Maryland, United States of America
- Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, Maryland, United States of America
| |
Collapse
|