1
|
Katebi A, Nouri M, Behrouzi A, Ajdary S, Riazi-Rad F. The pro-inflammatory responses of innate immune cells to Leishmania RNA virus 2-infected L. major support the survival and proliferation of the parasites. Biochimie 2024:S0300-9084(24)00241-4. [PMID: 39455049 DOI: 10.1016/j.biochi.2024.10.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 09/29/2024] [Accepted: 10/21/2024] [Indexed: 10/28/2024]
Abstract
Infection of Leishmania by Leishmania RNA virus (LRV) has been proposed as a pathogenic factor that induces pro-inflammatory responses through the TLR3/TLR4 signaling pathway. We investigated the effect of L. major infection by LRV2 on innate immune cell responses (human neutrophil (HL-60) and macrophage (THP-1) cell lines). The expression levels of pro- and anti-inflammatory cytokine and chemokine genes as well as genes involved in the amino acid metabolism of arginine were then investigated by RT-qPCR. Moreover, the expression of TLR genes and their downstream signaling pathways were compared in THP-1 cells infected with the two isolates. Apoptosis was also evaluated in infected THP-1 and HL-60 cells using the PI/Annexin V flow cytometry assay. In both cell lines, the expression of pro-inflammatory cytokines increased in response to LRV2+ L. major (Lm+), and the expression of chemokines shifted toward macrophage recruitment. In contrast to LRV2- L. major (Lm-), Lm + infected THP-1 cells acquired the M2-like phenotype. The presence of LRV2 increased the gene expression of TLRs and their signaling pathways, especially TLR3 and TLR4, which was proportional to the increase in pro-inflammatory cytokines. In addition, Lm + increased the expression of IL-10 and IFN-β, which contribute to the survival and growth of the parasite in the phagolysosome. Altogether, our results showed that Lm + could stimulate pro-inflammatory responses that promote parasite replication and stabilization in the host.
Collapse
Affiliation(s)
- Asal Katebi
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
| | - Matineh Nouri
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran; Department of Microbiology, Faculty of Advanced Science and Technology, Tehran Medical Science, Islamic Azad University, Tehran, Iran
| | - Ava Behrouzi
- Department of Microbiology, Faculty of Advanced Science and Technology, Tehran Medical Science, Islamic Azad University, Tehran, Iran
| | - Soheila Ajdary
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
| | - Farhad Riazi-Rad
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
2
|
PPARβ/δ Augments IL-1β-Induced COX-2 Expression and PGE2 Biosynthesis in Human Mesangial Cells via the Activation of SIRT1. Metabolites 2022; 12:metabo12070595. [PMID: 35888719 PMCID: PMC9320509 DOI: 10.3390/metabo12070595] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 06/11/2022] [Accepted: 06/22/2022] [Indexed: 12/10/2022] Open
Abstract
Peroxisome proliferator-activated receptor β/δ (PPARβ/δ), a ligand-activated nuclear receptor, regulates lipid and glucose metabolism and inflammation. PPARβ/δ can exert an anti-inflammatory effect by suppressing proinflammatory cytokine production. Cyclooxygenase-2 (COX-2)-triggered inflammation plays a crucial role in the development of many inflammatory diseases, including glomerulonephritis. However, the effect of PPARβ/δ on the expression of COX-2 in the kidney has not been fully elucidated. The present study showed that PPARβ/δ was functionally expressed in human mesangial cells (hMCs), where its expression was increased by interleukin-1β (IL-1β) treatment concomitant with enhanced COX-2 expression and prostaglandin E2 (PGE2) biosynthesis. The treatment of hMCs with GW0742, a selective agonist of PPARβ/δ, or the overexpression of PPARβ/δ via an adenovirus-mediated approach significantly increased COX-2 expression and PGE2 production. PPARβ/δ could further augment the IL-1β-induced COX-2 expression and PGE2 production in hMCs. Moreover, both PPARβ/δ activation and overexpression markedly increased sirtuin 1 (SIRT1) expression. The inhibition or knockdown of SIRT1 significantly attenuated the effects of PPARβ/δ on the IL-1β-induced expression of COX-2 and PGE2 biosynthesis. Taken together, PPARβ/δ could augment the IL-1β-induced COX-2 expression and PGE2 production in hMCs via the SIRT1 pathway. Given the critical role of COX-2 in glomerulonephritis, PPARβ/δ may represent a novel target for the treatment of renal inflammatory diseases.
Collapse
|
3
|
Gaballah A, Genedy D, Ghayaty E, El-Hawwary AA, Elmasry A. Standardized study of atorvastatin possible osteoarthritis disease-modifying effect in a rat model of osteoarthritis. Fundam Clin Pharmacol 2021; 36:296-305. [PMID: 34612533 DOI: 10.1111/fcp.12730] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 09/23/2021] [Accepted: 10/04/2021] [Indexed: 11/29/2022]
Abstract
We studied the osteoarthritis (OA)-modifying effects of atorvastatin in an experimental OA rat model and possible underlining mechanisms. We used 62 adult male Sprague-Dawley rats (250-300 g): 32 rats were used to assess the effects of atorvastatin on surgically induced OA in the knee, and 30 rats were used to assess the potential inflammatory effects of carrageenan-induced paw edema. In the OA model, joint stiffness was assessed by measuring the knee extension angle, and pathological changes in the OA knee joint were determined by histological examination and the measurement of serum biochemical markers, including interleukin-1β (IL-1β), matrix metalloproteinase-13 (MMP-13), and reduced glutathione (GSH). In the carrageenan-induced paw edema model, both paw thickness and pain threshold were assessed in different groups. Atorvastatin significantly improved joint stiffness, pathological changes, a significant mitigation of the higher MMP-13 and IL-1β, and a significant increase of reduced GSH in OA rats. Additionally, atorvastatin significantly improved both paw thickness and pain threshold in animals. Atorvastatin is a potential OA-modifying drug that warrants further clinical investigation.
Collapse
Affiliation(s)
- Ali Gaballah
- Department of Clinical Pharmacology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Doaa Genedy
- Department of Clinical Pharmacology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Essam Ghayaty
- Department of Clinical Pharmacology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Amany A El-Hawwary
- Department of Histology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Ahlam Elmasry
- Department of Clinical Pharmacology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
4
|
Engineered liposomes targeting the gut-CNS Axis for comprehensive therapy of spinal cord injury. J Control Release 2021; 331:390-403. [PMID: 33485884 DOI: 10.1016/j.jconrel.2021.01.032] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 12/17/2020] [Accepted: 01/19/2021] [Indexed: 02/08/2023]
Abstract
Effective curative therapies for spinal cord injury (SCI), which is often accompanied by intestinal complications, are lacking. Potential therapeutic targets include astrocytes and their enteric nervous system counterpart, enteric glial cells (EGCs). Based on shared biomarkers and similar functions of both cell types, we designed an orally administered targeted delivery system in which the neuropeptide apamin, stabilized by sulfur replacement with selenium, was adopted as a targeting moiety, and the liposome surface was protected with a non-covalent cross-linked chitosan oligosaccharide lactate layer. The system effectively permeated through oral absorption barriers, targeted local EGCs and astrocytes after systemic circulation, allowing for comprehensive SCI therapy. Given the involvement of the gut-organ axis in a growing number of diseases, our research may shed light on new aspects of the oral administration route as a bypass for multiple interventions and targeted therapy.
Collapse
|
5
|
Fu D, Yu JY, Connell AR, Hookham MB, McLeese RH, Lyons TJ. Effects of Modified Low-Density Lipoproteins and Fenofibrate on an Outer Blood-Retina Barrier Model: Implications for Diabetic Retinopathy. J Ocul Pharmacol Ther 2020; 36:754-764. [PMID: 33107777 PMCID: PMC7757531 DOI: 10.1089/jop.2020.0068] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Purpose: There is a lack of treatment for early diabetic retinopathy (DR), including blood-retina barrier (BRB) breakdown. The robust clinical benefit of fenofibrate in DR provides an opportunity to explore disease mechanisms and therapeutic targets. We have previously found that modified lipoproteins contribute to DR and that fenofibrate protects the inner BRB. We now investigate (1) whether modified lipoproteins elicit outer BRB injury and (2) whether fenofibrate may alleviate such damage. Methods: Human retinal pigment epithelium ARPE-19 cells were cultured in semipermeable transwells to establish a monolayer barrier and then exposed to heavily oxidized, glycated low-density lipoprotein (HOG-LDL, 25–300 mg/L, up to 24 h) versus native (N)-LDL. Transepithelial electric resistance (TEER) and FITC-dextran permeability were measured. The effects of fenofibrate, its active metabolite fenofibric acid, and other peroxisome proliferator-activated receptor (PPARα) agonists (gemfibrozil, bezafibrate, and WY14643) were evaluated, with and without the PPARα antagonist GW6471 or the adenosine monophosphate-activated protein kinase (AMPK) inhibitor Compound C. Results: HOG-LDL induced concentration- and time-dependent barrier impairment, decreasing TEER and increasing dextran leakage, effects that were amplified by high glucose. Fenofibric acid, but not fenofibrate, gemfibrozil, bezafibrate, or WY14643, attenuated barrier impairment. This effect was reversed significantly by Compound C, but not by GW6471. Conclusions: Modified lipoproteins elicited outer BRB injury in an experimental model, which was reduced by fenofibric acid through a PPARα-independent, AMPK-mediated mechanism. These findings suggest a protective role of fenofibric acid on the outer BRB in diabetic retina.
Collapse
Affiliation(s)
- Dongxu Fu
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Northern Ireland, United Kingdom
| | - Jeremy Y Yu
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Northern Ireland, United Kingdom.,Division of Endocrinology, Diabetes and Metabolic Diseases, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Anna R Connell
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Northern Ireland, United Kingdom
| | - Michelle B Hookham
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Northern Ireland, United Kingdom
| | - Rebecca H McLeese
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Northern Ireland, United Kingdom.,Division of Endocrinology, Diabetes and Metabolic Diseases, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Timothy J Lyons
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Northern Ireland, United Kingdom.,Division of Endocrinology, Diabetes and Metabolic Diseases, Medical University of South Carolina, Charleston, South Carolina, USA.,Diabetes Free SC, BlueCross BlueShield of South Carolina, Columbia, South Carolina, USA
| |
Collapse
|
6
|
Jang HS, Noh MR, Kim J, Padanilam BJ. Defective Mitochondrial Fatty Acid Oxidation and Lipotoxicity in Kidney Diseases. Front Med (Lausanne) 2020; 7:65. [PMID: 32226789 PMCID: PMC7080698 DOI: 10.3389/fmed.2020.00065] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 02/13/2020] [Indexed: 12/31/2022] Open
Abstract
The kidney is a highly metabolic organ and uses high levels of ATP to maintain electrolyte and acid-base homeostasis and reabsorb nutrients. Energy depletion is a critical factor in development and progression of various kidney diseases including acute kidney injury (AKI), chronic kidney disease (CKD), and diabetic and glomerular nephropathy. Mitochondrial fatty acid β-oxidation (FAO) serves as the preferred source of ATP in the kidney and its dysfunction results in ATP depletion and lipotoxicity to elicit tubular injury and inflammation and subsequent fibrosis progression. This review explores the current state of knowledge on the role of mitochondrial FAO dysfunction in the pathophysiology of kidney diseases including AKI and CKD and prospective views on developing therapeutic interventions based on mitochondrial energy metabolism.
Collapse
Affiliation(s)
- Hee-Seong Jang
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Mi Ra Noh
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Jinu Kim
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States.,Department of Anatomy, Jeju National University School of Medicine, Jeju, South Korea.,Interdisciplinary Graduate Program in Advanced Convergence Technology & Science, Jeju National University, Jeju, South Korea
| | - Babu J Padanilam
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States.,Internal Medicine, Section of Nephrology, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
7
|
Duarte S, Melo T, Domingues R, de Dios Alché J, Pérez-Sala D. Insight into the cellular effects of nitrated phospholipids: Evidence for pleiotropic mechanisms of action. Free Radic Biol Med 2019; 144:192-202. [PMID: 31199965 DOI: 10.1016/j.freeradbiomed.2019.06.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 05/26/2019] [Accepted: 06/03/2019] [Indexed: 12/20/2022]
Abstract
Nitrated phospholipids have been recently identified in biological systems and showed to display anti-oxidant and anti-inflammatory potential in models of inflammation in vitro. Here, we have explored the effects of nitrated 1-palmitoyl-2-oleyl-phosphatidyl choline (NO2-POPC) in cellular models. We have observed that NO2-POPC, but not POPC, induces cellular changes consisting in cytoskeletal rearrangement and cell shrinking, and ultimately, loss of cell adhesion or impaired cell attachment. NO2-POPC releases NO in vitro and induces accumulation of NO in cells. Nevertheless, the effects of NO2-POPC are not superimposable with those of NO donors, which points to distinctive mechanisms of action. Notably, they show a stronger parallelism, although not complete overlap, with the effects of nitrated fatty acids. Interestingly, redistribution of vimentin by NO2-POPC is attenuated in a C328S mutant, thus indicating that this residue may be a target for direct or indirect modification in NO2-POPC-treated cells. Additionally, NO2-POPC interacts with several typical lipoxidation targets in vitro, including vimentin and PPARγ constructs, likely through cysteine residues. Therefore, nitrated phospholipids emerge as potential novel electrophilic lipid mediators with selective actions.
Collapse
Affiliation(s)
- Sofia Duarte
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, 28040, Madrid, Spain
| | - Tânia Melo
- Mass Spectrometry Center & QOPNA, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal; Department of Chemistry & CESAM& ECOMARE, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Rosário Domingues
- Mass Spectrometry Center & QOPNA, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal; Department of Chemistry & CESAM& ECOMARE, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Juan de Dios Alché
- Plant Reproductive Biology and Advanced Imaging Laboratory, Estación Experimental del Zaidín, Consejo Superior de Investigaciones Científicas, 18008, Granada, Spain
| | - Dolores Pérez-Sala
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, 28040, Madrid, Spain.
| |
Collapse
|
8
|
Khan MA, Alam Q, Haque A, Ashafaq M, Khan MJ, Ashraf GM, Ahmad M. Current Progress on Peroxisome Proliferator-activated Receptor Gamma Agonist as an Emerging Therapeutic Approach for the Treatment of Alzheimer's Disease: An Update. Curr Neuropharmacol 2019; 17:232-246. [PMID: 30152284 PMCID: PMC6425074 DOI: 10.2174/1570159x16666180828100002] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 03/14/2018] [Accepted: 08/21/2018] [Indexed: 01/24/2023] Open
Abstract
Alzheimer's disease (AD) is an age-related progressive neurodegenerative disorder, characterized by the deposition of amyloid-β within the brain parenchyma resulting in a significant decline in cognitive functions. The pathophysiological conditions of the disease are recognized by the perturbation of synaptic function, energy and lipid metabolism. In Addition deposition of amyloid plaques also triggers inflammation upon the induction of microglia. Peroxisome proliferatoractivated receptors (PPARs) are ligand-activated transcription factors known to play important role in the regulation of glucose absorption, homeostasis of lipid metabolism and are further known to involved in repressing the expression of genes related to inflammation. Therefore, agonists of this receptor represent an attractive therapeutic target for AD. Recently, both clinical and preclinical studies showed that use of Peroxisome proliferator-activated receptor gamma (PPARγ) agonist improves both learning and memory along with other AD related pathology. Thus, PPARγ signifies a significant new therapeutic target in treating AD. In this review, we have shed some light on the recent progress of how, PPARγ agonist selectively modulated different cellular targets in AD and its amazing potential in the treatment of AD.
Collapse
Affiliation(s)
- Mahmood Ahmad Khan
- Address correspondence to these authors at the Department of Biochemistry, University College of Medical Sciences and GTB Hospital, University of Delhi, Dilshad Garden, Delhi 110095, India; E-mail: , and King Fahd Medical Research Center, King Abdulaziz University, P.O. Box 80216, Jeddah 21589, Saudi Arabia; E-mail:
| | | | | | | | | | - Ghulam Md Ashraf
- Address correspondence to these authors at the Department of Biochemistry, University College of Medical Sciences and GTB Hospital, University of Delhi, Dilshad Garden, Delhi 110095, India; E-mail: , and King Fahd Medical Research Center, King Abdulaziz University, P.O. Box 80216, Jeddah 21589, Saudi Arabia; E-mail:
| | | |
Collapse
|
9
|
Lysophosphatidic acid enhances PGE2 to PGF2α ratio and nitric oxide level in nonpregnant buffalo uterus. Theriogenology 2018; 120:47-55. [PMID: 30092374 DOI: 10.1016/j.theriogenology.2018.07.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Revised: 07/10/2018] [Accepted: 07/16/2018] [Indexed: 01/24/2023]
Abstract
Lysophosphatidic acid (LPA) is a small ubiquitous lipid exerting diverse biological functions. Its role in reproduction in different species has created great interest in recent times. In the present study, we aimed to elucidate LPA signaling in nonpregnant buffalo uterus by in vitro studies. Standard techniques like real-time PCR (for mRNA expression of LPARs and COX-2 and iNOS), Western blot (for PPARγ protein expression), sandwich ELISA (for PGE2 and PGF2α assay) and histopathology (for assessment of endometrial architecture in culture) were used in this study. The buffalo uterine tissues were collected from the local slaughterhouse and were selected for the study on the basis of the presence of corpus luteum on the ovary (n = 5). The LPAR3 receptor was the highest expressed receptor as compared to LPAR1 and LPAR6 in non-pregnant uterine tissues after 6 h incubation in Dulbecco's Modified Eagle Medium (DMEM). 50 μM LPA increased the mRNA expressions of COX-2 and iNOS enzymes which were attenuated by the treatment of LPAR1/3 antagonist Ki16425. PPARγ antagonist GW9662 prevented the LPA-induced increase in iNOS mRNA expression but did not alter the COX-2 expression. LPA also enhanced the PGE2 to PGF2α ratio in uterine tissue homogenates which was inhibited by all the receptor antagonists as well as by the inhibitors of COX-2 and iNOS. LPA also increased the total nitrite level in tissue homogenates in LPAR1/3- and iNOS-dependent manner. Additionally, we demonstrate PPARγ mRNA and protein expressions in nonpregnant buffalo endometrium. In conclusion, the results of the present study suggest that LPA acts as a luteotropic factor during the estrus cycle in nonpregnant buffalo uterus by enhancing PGE2 to PGF2α ratio and NO level through multiple receptors.
Collapse
|
10
|
Cheng MC, Pan TM. Glyceryl 1,3-Dipalmitate Produced from Lactobacillus paracasei subspecies. paracasei NTU 101 Inhibits Oxygen-Glucose Deprivation and Reperfusion-Induced Oxidative Stress via Upregulation of Peroxisome Proliferator-Activated Receptor γ in Neuronal SH-SY5Y Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2017; 65:7926-7933. [PMID: 28829589 DOI: 10.1021/acs.jafc.7b02728] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Glyceryl 1,3-dipalmitate (GD) purified from Lactobacillus paracasei subsp. paracasei NTU 101-fermented products has been demonstrated to possess neuroprotective properties. We determined the effect of GD on oxygen-glucose deprivation and reperfusion (OGD/R)-induced SH-SY5Y neuroblastoma cell death. GD ameliorated OGD/R-induced apoptosis by elevating the protein expression of nuclear peroxisome proliferator-activated receptor γ (PPARγ) and nuclear factor erythroid 2-related factor 2 (Nrf2), thereby attenuating reactive oxygen species (ROS) generation. Pretreatment with GD reduced nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) expression from 1.54 ± 0.27 to 0.84 ± 0.46, thereby attenuating the induction of pro-inflammatory mediators, and increased the plasma membrane Ca2+ ATPase (PMCA) levels from 0.81 ± 0.02 to 1.08 ± 0.06, thus reducing the levels of cytosolic Ca2+; this also correlated with reduced cell death. We conclude that GD prevents SH-SY5Y cells from injury after OGD/R insult, possibly by modulating oxidative stress and inflammatory response.
Collapse
Affiliation(s)
- Meng-Chun Cheng
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University , Number 1, Section 4, Roosevelt Road, Taipei 10617, Taiwan
| | - Tzu-Ming Pan
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University , Number 1, Section 4, Roosevelt Road, Taipei 10617, Taiwan
| |
Collapse
|
11
|
Ravanan P, Srikumar IF, Talwar P. Autophagy: The spotlight for cellular stress responses. Life Sci 2017; 188:53-67. [PMID: 28866100 DOI: 10.1016/j.lfs.2017.08.029] [Citation(s) in RCA: 428] [Impact Index Per Article: 61.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 08/05/2017] [Accepted: 08/28/2017] [Indexed: 02/06/2023]
Abstract
Autophagy is an essential cellular mechanism which plays "housekeeping" role in normal physiological processes including removing of long lived, aggregated and misfolded proteins, clearing damaged organelles, growth regulation and aging. Autophagy is also involved in a variety of biological functions like development, cellular differentiation, defense against pathogens and nutritional starvation. The integration of autophagy into these biological functions and other stress responses is determined by the transcriptional factors that undertake the regulatory mechanism. This review discusses the machinery of autophagy, the molecular web that connects autophagy to various stress responses like inflammation, hypoxia, ER stress, and various other pathologic conditions. Defects in autophagy regulation play a central role in number of diseases, including neurodegenerative diseases, cancer, pathogen infection and metabolic diseases. Similarly, inhibiting autophagy would contribute in the treatment of cancer. However, understanding the biology of autophagy regulation requires pharmacologically active compounds which modulate the autophagy process. Inducers of autophagy are currently receiving considerable attention as autophagy upregulation may be a therapeutic benefit for certain neurodegenerative diseases (via removal of protein aggregates) while the inhibitors are being investigated for the treatment of cancers. Both induction and inhibition of autophagy have been proven to be beneficial in the treatment of cancer. This dual role of autophagy in cancers is now getting uncovered by the advancement in the research findings and development of effective autophagy modulators.
Collapse
Affiliation(s)
- Palaniyandi Ravanan
- Apoptosis and Cell Survival Research Laboratory, Department of Biosciences, School of Biosciences and Technology, VIT University, Vellore, Tamil Nadu-632014, India.
| | - Ida Florance Srikumar
- Apoptosis and Cell Survival Research Laboratory, Department of Biosciences, School of Biosciences and Technology, VIT University, Vellore, Tamil Nadu-632014, India
| | - Priti Talwar
- Apoptosis and Cell Survival Research Laboratory, Department of Biosciences, School of Biosciences and Technology, VIT University, Vellore, Tamil Nadu-632014, India
| |
Collapse
|
12
|
Ayala-Peña VB, Pilotti F, Volonté Y, Rotstein NP, Politi LE, German OL. Protective effects of retinoid x receptors on retina pigment epithelium cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:1134-45. [DOI: 10.1016/j.bbamcr.2016.02.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2015] [Revised: 02/04/2016] [Accepted: 02/12/2016] [Indexed: 01/01/2023]
|
13
|
Barcelos RP, Bresciani G, Rodriguez-Miguelez P, Cuevas MJ, Soares FAA, Barbosa NV, González-Gallego J. Diclofenac pretreatment effects on the toll-like receptor 4/nuclear factor kappa B-mediated inflammatory response to eccentric exercise in rat liver. Life Sci 2016; 148:247-53. [DOI: 10.1016/j.lfs.2016.02.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 10/04/2015] [Accepted: 02/03/2016] [Indexed: 01/22/2023]
|
14
|
Karuppagounder V, Arumugam S, Thandavarayan RA, Pitchaimani V, Sreedhar R, Afrin R, Harima M, Suzuki H, Suzuki K, Nakamura M, Ueno K, Watanabe K. Naringenin ameliorates daunorubicin induced nephrotoxicity by mitigating AT1R, ERK1/2-NFκB p65 mediated inflammation. Int Immunopharmacol 2015; 28:154-9. [PMID: 26072060 DOI: 10.1016/j.intimp.2015.05.050] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 05/26/2015] [Accepted: 05/31/2015] [Indexed: 12/27/2022]
Abstract
Inflammation and oxidative stress play important roles in the progression of renal damage. The natural polyphenol naringenin is known to exert potent antioxidant and anti-inflammatory effects. In this study, we have investigated the effect of naringenin on kidney dysfunction, fibrosis, endoplasmic reticulum (ER) stress, angiotensin II type I receptor (AT1R) expression and inflammation in daunorubicin (DNR) induced nephrotoxicity model. Nephrotoxicity was induced in rats by intravenous injection of DNR at a cumulative dose of 9 mg/kg. After 1 week, naringenin (20mg/kg/day. p.o) was administered daily for 6 weeks. Biochemical studies were performed to evaluate renal function. Western blotting was performed to measure the protein levels of AT1R, endothelin (ET)1, ET receptor type A (ETAR), extracellular signal-regulated kinase (ERK)1/2, nuclear factor (NF)κB p65, peroxisome proliferator activated receptor (PPAR)γ, oxidative/ER stress, apoptosis, and inflammatory markers in the kidney of DNR treated rats. Histopathological analysis was done using hemotoxylin eosin and Masson trichrome stained renal sections to investigate the structural abnormalities and fibrosis. DNR treated rats suffered from nephrotoxicity as evidenced by worsened renal function, increased blood urea nitrogen, serum creatinine levels in renal tissues and histopathogical abnormalities. Treatment with naringenin mitigated these changes. Furthermore, naringenin up regulated PPARγ and down regulated AT1R, ET1, ETAR, p-ERK1/2, p-NFκB p65, ER stress, apoptosis, and inflammatory markers. Our results suggest that naringenin has an ability to improve renal function and attenuates AT1R, ERK1/2-NFκB p65 signaling pathway in DNR induced nephrotoxicity in rats.
Collapse
Affiliation(s)
- Vengadeshprabhu Karuppagounder
- Department of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Niigata University of Pharmacy and Applied Life Sciences, Niigata 956-8603, Japan
| | - Somasundaram Arumugam
- Department of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Niigata University of Pharmacy and Applied Life Sciences, Niigata 956-8603, Japan
| | - Rajarajan Amirthalingam Thandavarayan
- Department of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Niigata University of Pharmacy and Applied Life Sciences, Niigata 956-8603, Japan; Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Vigneshwaran Pitchaimani
- Department of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Niigata University of Pharmacy and Applied Life Sciences, Niigata 956-8603, Japan
| | - Remya Sreedhar
- Department of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Niigata University of Pharmacy and Applied Life Sciences, Niigata 956-8603, Japan
| | - Rejina Afrin
- Department of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Niigata University of Pharmacy and Applied Life Sciences, Niigata 956-8603, Japan
| | - Meilei Harima
- Department of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Niigata University of Pharmacy and Applied Life Sciences, Niigata 956-8603, Japan
| | - Hiroshi Suzuki
- Department of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Niigata University of Pharmacy and Applied Life Sciences, Niigata 956-8603, Japan
| | - Kenji Suzuki
- Department of Gastroenterology, Niigata University Graduate School of Medical and Dental Sciences, Niigata City 951-8510, Japan
| | - Masahiko Nakamura
- Department of Cardiology, Yamanashi Prefectural Central Hospital, 1-1-1 Fujimi Kofu, Yamanashi 400-8506, Japan
| | - Kazuyuki Ueno
- Department of Pharmaceutical Sciences, Niigata University of Pharmacy and Applied Life Sciences, Niigata 956-8603, Japan
| | - Kenichi Watanabe
- Department of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Niigata University of Pharmacy and Applied Life Sciences, Niigata 956-8603, Japan.
| |
Collapse
|
15
|
Zhao XR, Gonzales N, Aronowski J. Pleiotropic role of PPARγ in intracerebral hemorrhage: an intricate system involving Nrf2, RXR, and NF-κB. CNS Neurosci Ther 2014; 21:357-66. [PMID: 25430543 DOI: 10.1111/cns.12350] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 10/10/2014] [Accepted: 10/11/2014] [Indexed: 12/13/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a subtype of stroke involving formation of hematoma within brain parenchyma, which accounts for 8-15% of all strokes in Western societies and 20-30% among Asian populations, and has a 1-year mortality rate >50%. The high mortality and severe morbidity make ICH a major public health problem. Only a few evidence-based targeted treatments are used for ICH management, and interventions focus primarily on supportive care and comorbidity prevention. Even in patients who survive the ictus, extravasated blood (including plasma components) and subsequent intrahematoma hemolytic products trigger a series of adverse events within the brain parenchyma, leading to secondary brain injury, edema and severe neurological deficits or death. Although the hematoma in humans gradually resolves within months, full restoration of neurological function can be slow and often incomplete, leaving survivors with devastating neurological deficits. During past years, peroxisome proliferator-activated receptor gamma (PPARγ) transcription factor and its agonists received recognition as important players in regulating not only glucose and lipid metabolism (which underlies its therapeutic effect in type 2 diabetes mellitus), and more recently, as an instrumental pleiotropic regulator of antiinflammation, antioxidative regulation, and phagocyte-mediated cleanup processes. PPARγ agonists have emerged as potential therapeutic target for stroke. The use of PPARγ as a therapeutic target appears to have particularly strong compatibility toward pathogenic components of ICH. In addition to its direct genomic effect, PPARγ may interact with transcription factor, NF-κB, which may underlie many aspects of the antiinflammatory effect of PPARγ. Furthermore, PPARγ appears to regulate expression of Nrf2, another transcription factor and master regulator of detoxification and antioxidative regulation. Finally, the synergistic costimulation of PPARγ and retinoid X receptor, RXR, may play an additional role in the therapeutic modulation of PPARγ function. In this article, we outline the main components of the role of PPARγ in ICH pathogenesis.
Collapse
Affiliation(s)
- Xiu-Rong Zhao
- Department of Neurology, Stroke Research Center, University of Texas Medical School - Houston, Houston, TX, USA
| | | | | |
Collapse
|
16
|
Study of protein haptenation by amoxicillin through the use of a biotinylated antibiotic. PLoS One 2014; 9:e90891. [PMID: 24595455 PMCID: PMC3940954 DOI: 10.1371/journal.pone.0090891] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 02/06/2014] [Indexed: 12/28/2022] Open
Abstract
Allergic reactions towards β-lactam antibiotics pose an important clinical problem. The ability of small molecules, such as a β-lactams, to bind covalently to proteins, in a process known as haptenation, is considered necessary for induction of a specific immunological response. Identification of the proteins modified by β-lactams and elucidation of the relevance of this process in allergic reactions requires sensitive tools. Here we describe the preparation and characterization of a biotinylated amoxicillin analog (AX-B) as a tool for the study of protein haptenation by amoxicillin (AX). AX-B, obtained by the inclusion of a biotin moiety at the lateral chain of AX, showed a chemical reactivity identical to AX. Covalent modification of proteins by AX-B was reduced by excess AX and vice versa, suggesting competition for binding to the same targets. From an immunological point of view, AX and AX-B behaved similarly in RAST inhibition studies with sera of patients with non-selective allergy towards β-lactams, whereas, as expected, competition by AX-B was poorer with sera of AX-selective patients, which recognize AX lateral chain. Use of AX-B followed by biotin detection allowed the observation of human serum albumin (HSA) modification by concentrations 100-fold lower that when using AX followed by immunological detection. Incubation of human serum with AX-B led to the haptenation of all of the previously identified major AX targets. In addition, some new targets could be detected. Interestingly, AX-B allowed the detection of intracellular protein adducts, which showed a cell type-specific pattern. This opens the possibility of following the formation and fate of AX-B adducts in cells. Thus, AX-B may constitute a valuable tool for the identification of AX targets with high sensitivity as well as for the elucidation of the mechanisms involved in allergy towards β-lactams.
Collapse
|
17
|
Abstract
Statins have immunomodulatory effects. Hence, they could be advantageous for different reasons. First, statins can serve as adjunct therapy in autoimmune diseases. Second, they may play a role in the prevention of accelerated atherosclerosis in the same population. In this review, we discuss the mechanisms of immune regulation by statins and review the literature for their benefit in many autoimmune diseases. In these studies, statins lead to an improvement in the disease activity scores in rheumatoid arthritis patients; a reduction in prothrombotic factors in anti-phospholipid syndrome patients; changes in vasculature, proteinuria, and cardiac events in systemic lupus erythematosus patients; changes in vasculature and proteinuria in patients with vasculitis; disease activity scores in ankylosing spondylitis; and finally vascular changes in patients with systemic sclerosis. Hence, these studies suggest that statins could be used as adjuncts to standard therapy due to their anti-inflammatory, immunomodulatory, and anti-thrombogenic effects. More evidence-based research is required to determine whether statin regimens will become the standard of care in patients with autoimmune diseases.
Collapse
|
18
|
The Proatherogenic Effect of Chronic Nitric Oxide Synthesis Inhibition in ApoE-Null Mice Is Dependent on the Presence of PPAR α. PPAR Res 2014; 2014:124583. [PMID: 24587793 PMCID: PMC3920724 DOI: 10.1155/2014/124583] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2013] [Revised: 10/14/2013] [Accepted: 10/18/2013] [Indexed: 12/17/2022] Open
Abstract
Inhibition of endothelial nitric oxide synthase (eNOS) accelerates atherosclerosis in ApoE-null mice by impairing the balance between angiotensin II (AII) and NO. Our previous data suggested a role for PPARα in the deleterious effect of the renin-angiotensin system (RAS). We tested the hypothesis that ApoE-null mice lacking PPARα (DKO mice) would be resistant to the proatherogenic effect of NOS inhibition. DKO mice fed a Western diet were immune to the 23% worsening in aortic sinus plaque area seen in the ApoE-null animals under 12 weeks of NOS inhibition with a subpressor dose of L-NAME, P = 0.002. This was accompanied by a doubling of reactive oxygen species (ROS-) generating aortic NADPH oxidase activity (a target of AII, which paralleled Nox1 expression) and by a 10-fold excess of the proatherogenic iNOS, P < 0.01. L-NAME also caused a doubling of aortic renin and angiotensinogen mRNA level in the ApoE-null mice but not in the DKO, and it upregulated eNOS in the DKO mice only. These data suggest that, in the ApoE-null mouse, PPARα contributes to the proatherogenic effect of unopposed RAS/AII action induced by L-NAME, an effect which is associated with Nox1 and iNOS induction, and is independent of blood pressure and serum lipids.
Collapse
|
19
|
Beltrán A, Isabel Burguete M, Abánades DR, Pérez-Sala D, Luis SV, Galindo F. Turn-on fluorescent probes for nitric oxide sensing based on the ortho-hydroxyamino structure showing no interference with dehydroascorbic acid. Chem Commun (Camb) 2014; 50:3579-81. [DOI: 10.1039/c3cc49555h] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
20
|
Javadi S, Ejtemaeimehr S, Keyvanfar HR, Moghaddas P, Aminian A, Rajabzadeh A, Mani AR, Dehpour AR. Pioglitazone potentiates development of morphine-dependence in mice: possible role of NO/cGMP pathway. Brain Res 2013; 1510:22-37. [PMID: 23399681 DOI: 10.1016/j.brainres.2012.12.035] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Revised: 11/15/2012] [Accepted: 12/13/2012] [Indexed: 01/23/2023]
Abstract
Peroxizome proliferator-activated receptor gamma (PPARγ) is highly expressed in the central nervous system where it modulates numerous gene transcriptions. Nitric oxide synthase (NOS) expression could be modified by simulation of PPARγ which in turn activates nitric oxide (NO)/soluble guanylyl-cyclase (sGC)/cyclic guanosine mono phosphate (cGMP) pathway. It is well known that NO/cGMP pathway possesses pivotal role in the development of opioid dependence and this study is aimed to investigate the effect of PPARγ stimulation on opioid dependence in mice as well as human glioblastoma cell line. Pioglitazone potentiated naloxone-induced withdrawal syndrome in morphine dependent mice in vivo. While selective inhibition of PPARγ, neuronal NOS or GC could reverse the pioglitazone-induced potentiation of morphine withdrawal signs; sildenafil, a phosphodiesterase-5 inhibitor amplified its effect. We also showed that nitrite levels in the hippocampus were significantly elevated in pioglitazone-treated morphine dependent mice. In the human glioblastoma (U87) cell line, rendered dependent to morphine, cAMP levels did not show any alteration after chronic pioglitazone administration while cGMP measurement revealed a significant rise. We were unable to show a significant alteration in neuronal NOS mRNA expressions by pioglitazone in mice hippocampus or U87 cells. Our results suggest that pioglitazone has the ability to enhance morphine-dependence and to augment morphine withdrawal signs. The possible pathway underlying this effect is through activation of NO/GC/cGMP pathway.
Collapse
Affiliation(s)
- Shiva Javadi
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, P.O. Box 13145-784, Tehran, Iran
| | | | | | | | | | | | | | | |
Collapse
|
21
|
The regulatory effect of veratric acid on NO production in LPS-stimulated RAW264.7 macrophage cells. Cell Immunol 2012; 280:164-70. [DOI: 10.1016/j.cellimm.2012.12.007] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Revised: 12/10/2012] [Accepted: 12/25/2012] [Indexed: 11/23/2022]
|
22
|
Kouroumichakis I, Papanas N, Zarogoulidis P, Liakopoulos V, Maltezos E, Mikhailidis DP. Fibrates: therapeutic potential for diabetic nephropathy? Eur J Intern Med 2012; 23:309-16. [PMID: 22560376 DOI: 10.1016/j.ejim.2011.12.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2011] [Revised: 12/04/2011] [Accepted: 12/18/2011] [Indexed: 12/30/2022]
Abstract
Despite intensive glucose-lowering treatment and advanced therapies for cardiovascular risk factors, such as hypertension and dyslipidaemia, diabetes mellitus with its macro- and microvascular complications remains a major health problem. Especially diabetic nephropathy is a leading cause of morbidity and mortality, and its prevalence is increasing. Peroxisome proliferator-activated receptor-α (PPAR-α), a member of a large nuclear receptor superfamily, is expressed in several tissues including the kidney. Recently, experimental data have suggested that PPAR-α activation plays a pivotal role in the regulation of fatty acid oxidation, lipid metabolism, inflammatory and vascular responses, and might regulate various metabolic and intracellular signalling pathways that lead to diabetic microvascular complications. This review examines the role of PPAR-α activation in diabetic nephropathy and summarises data from experimental and clinical studies on the emerging therapeutic potential of fibrates in diabetic nephropathy.
Collapse
Affiliation(s)
- I Kouroumichakis
- Outpatient Clinic of Obesity, Diabetes and Metabolism, Second Department of Internal Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | | | | | | | | | | |
Collapse
|
23
|
PPARγ agonist rosiglitazone ameliorates LPS-induced inflammation in vascular smooth muscle cells via the TLR4/TRIF/IRF3/IP-10 signaling pathway. Cytokine 2011; 55:409-19. [DOI: 10.1016/j.cyto.2011.05.020] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2010] [Revised: 05/18/2011] [Accepted: 05/26/2011] [Indexed: 02/06/2023]
|
24
|
|
25
|
WY-14643, a Potent Peroxisome Proliferator Activator Receptor-α PPAR-α Agonist Ameliorates the Inflammatory Process Associated to Experimental Periodontitis. PPAR Res 2010; 2010:193019. [PMID: 21253492 PMCID: PMC3022213 DOI: 10.1155/2010/193019] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2010] [Revised: 11/05/2010] [Accepted: 11/24/2010] [Indexed: 01/14/2023] Open
Abstract
We have investigated the effects of WY14643, a potent peroxisome proliferator activator receptor-α (PPAR-α) agonist, in a rat model of ligature-induced periodontitis.
Male Sprague-Dawley rats were lightly anaesthetized with pentobarbitone (35 mg/kg). Sterile, 2-0 black braided silk thread was placed around the cervix of the lower left first molar and knotted medially. Animals received WY14643 (1 mg/kg i.p, daily for eight days). Eighths days after placement of the ligature, we evaluated several markers of inflammation such us (1) myeloperoxidase activity, (2) a cytokines and adhesion molecules expression, (3) NF-κB expression, (4) iNOS expression, (5) the nitration of tyrosine residues, (6) activation of the nuclear enzyme poly(ADP-ribose) polymerase, (7) apoptosis, and (8) the degree of gingivomucosal tissues injury. Administration of WY14643 significantly decreased all of the parameters of inflammation as described above. These results demonstrate that WY14643 exerts an anti-inflammatory role during experimental periodontitis and is able to ameliorate the tissue damage.
Collapse
|
26
|
Rakkestad KE, Holme JA, Paulsen RE, Schwarze PE, Becher R. Mono(2-ethylhexyl) phthalate induces both pro- and anti-inflammatory responses in rat alveolar macrophages through crosstalk between p38, the lipoxygenase pathway and PPARalpha. Inhal Toxicol 2010; 22:140-50. [PMID: 19938896 DOI: 10.3109/08958370903019885] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Airway inflammation is important in asthma pathogenesis. Recent epidemiological data have indicated an association between asthma symptoms in children and exposure to di(2-ethylhexyl) phthalate (DEHP). Thus, we have studied inflammatory responses in primary rat alveolar macrophages (AMs) after exposure to mono(2-ethylhexyl) phthalate (MEHP), the major primary metabolite of DEHP. First, we show that MEHP induces a dose-dependent release of the pro-inflammatory tumour necrosis factor-alpha (TNF-alpha) in AMs, giving a maximal (5-fold) increase at 0.7 mM. This concentration also induced some cell death. MEHP also induced phosphorylation of MAPK p38, while the p38 inhibitor SB 202190 reduced MEHP-induced TNF-alpha, suggesting a p38-dependent cytokine production. Next, we elucidated possible effects of MEHP on the 5-lipoxygenase (5-LO) pathway and found that MEHP caused increased leukotriene (LTB(4)) release. Further, we found that the 5-LO inhibitor nordihydrogualaretic acid (NDGA) significantly reduced both MEHP-induced TNF-alpha release and MEHP-induced formation of reactive oxygen species (ROS), supporting an involvement of the 5-LO pathway in MEHP induced inflammatory reactions. Last, we found that MK-886, a known inhibitor of peroxisome proliferator-activated receptor alpha (PPARalpha), increased the MEHP-induced TNF-alpha response. This indicates that MEPH-PPARalpha binding mediates an anti-inflammatory signal.
Collapse
Affiliation(s)
- Kirsten E Rakkestad
- Department of Air Pollution and Noise, Division of Environmental Medicine, Norwegian Institute of Public Health, Oslo, Norway
| | | | | | | | | |
Collapse
|
27
|
Mao XR, Moerman-Herzog AM, Chen Y, Barger SW. Unique aspects of transcriptional regulation in neurons--nuances in NFkappaB and Sp1-related factors. J Neuroinflammation 2009; 6:16. [PMID: 19450264 PMCID: PMC2693111 DOI: 10.1186/1742-2094-6-16] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2009] [Accepted: 05/18/2009] [Indexed: 12/11/2022] Open
Abstract
The unique physiology and function of neurons create differences in their cellular physiology, including their regulation of gene expression. We began several years ago exploring the relationships between the NFκB transcription factor, neuronal survival, and glutamate receptor activation in telencephalic neurons. These studies led us to conclude that this population of cells is nearly incapable of activating the NFκB that is nonetheless expressed at reasonable levels. A subset of the κB cis elements are instead bound by members of the Sp1 family in neurons. Also surprising was our discovery that Sp1 itself, typically described as ubiquitous, is severely restricted in expression within forebrain neurons; Sp4 seems to be substituted during neuronal differentiation. These findings and their implications for neuronal differentiation – as well as potential dedifferentiation during degenerative processes – are discussed here.
Collapse
Affiliation(s)
- Xianrong R Mao
- Department of Anesthesiology, Washington University School of Medicine, St Louis, MO 63110, USA.
| | | | | | | |
Collapse
|
28
|
Syversen U, Stunes AK, Gustafsson BI, Obrant KJ, Nordsletten L, Berge R, Thommesen L, Reseland JE. Different skeletal effects of the peroxisome proliferator activated receptor (PPAR)alpha agonist fenofibrate and the PPARgamma agonist pioglitazone. BMC Endocr Disord 2009; 9:10. [PMID: 19331671 PMCID: PMC2678137 DOI: 10.1186/1472-6823-9-10] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2008] [Accepted: 03/30/2009] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND All the peroxisome proliferator activated receptors (PPARs) are found to be expressed in bone cells. The PPARgamma agonist rosiglitazone has been shown to decrease bone mass in mice and thiazolidinediones (TZDs) have recently been found to increase bone loss and fracture risk in humans treated for type 2 diabetes mellitus. The aim of the study was to examine the effect of the PPARalpha agonist fenofibrate (FENO) and the PPARgamma agonist pioglitazone (PIO) on bone in intact female rats. METHODS Rats were given methylcellulose (vehicle), fenofibrate or pioglitazone (35 mg/kg body weight/day) by gavage for 4 months. BMC, BMD, and body composition were measured by DXA. Histomorphometry and biomechanical testing of excised femurs were performed. Effects of the compounds on bone cells were studied. RESULTS The FENO group had higher femoral BMD and smaller medullary area at the distal femur; while trabecular bone volume was similar to controls. Whole body BMD, BMC, and trabecular bone volume were lower, while medullary area was increased in PIO rats compared to controls. Ultimate bending moment and energy absorption of the femoral shafts were reduced in the PIO group, while similar to controls in the FENO group. Plasma osteocalcin was higher in the FENO group than in the other groups. FENO stimulated proliferation and differentiation of, and OPG release from, the preosteoblast cell line MC3T3-E1. CONCLUSION We show opposite skeletal effects of PPARalpha and gamma agonists in intact female rats. FENO resulted in significantly higher femoral BMD and lower medullary area, while PIO induced bone loss and impairment of the mechanical strength. This represents a novel effect of PPARalpha activation.
Collapse
Affiliation(s)
- Unni Syversen
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Endocrinology, St Olav's University Hospital HF, Trondheim, Norway
| | - Astrid K Stunes
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Björn I Gustafsson
- Department of Gastroenterology, St Olav's University Hospital HF, Trondheim, Norway
- Department of Surgery, Yale University School of Medicine, New Haven, USA
| | - Karl J Obrant
- Department of Orthopaedics, Malmø University Hospital, Malmø, Sweden
| | - Lars Nordsletten
- Department of Orthopaedics, Ullevål University Hospital, Oslo, Norway
| | - Rolf Berge
- Section of Medical Biochemistry, Institute of Medicine, University of Bergen, Norway
| | - Liv Thommesen
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Sør-Trøndelag University College, Faculty of Food Science and Medical Technology, Trondheim, Norway
| | - Janne E Reseland
- Department of Biomaterials, Institute for Clinical Dentistry, University of Oslo, Blindern, Oslo, Norway
| |
Collapse
|
29
|
Napimoga MH, Vieira SM, Dal-Secco D, Freitas A, Souto FO, Mestriner FL, Alves-Filho JC, Grespan R, Kawai T, Ferreira SH, Cunha FQ. Peroxisome proliferator-activated receptor-gamma ligand, 15-deoxy-Delta12,14-prostaglandin J2, reduces neutrophil migration via a nitric oxide pathway. THE JOURNAL OF IMMUNOLOGY 2008; 180:609-17. [PMID: 18097063 DOI: 10.4049/jimmunol.180.1.609] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Ligands for peroxisome proliferator-activated receptor gamma (PPAR-gamma), such as 15-deoxy-Delta12,14-prostaglandin J2 (15d-PGJ2) have been implicated as a new class of anti-inflammatory compounds with possible clinical applications. Based on this concept, this investigation was designed to determine the effect of 15d-PGJ2-mediated activation of PPAR-gamma ligand on neutrophil migration after an inflammatory stimulus and clarify the underlying molecular mechanisms using a mouse model of peritonitis. Our results demonstrated that 15d-PGJ2 administration decreases leukocyte rolling and adhesion to the inflamed mesenteric tissues by a mechanism dependent on NO. Specifically, pharmacological inhibitors of NO synthase remarkably abrogated the 15d-PGJ2-mediated suppression of neutrophil migration to the inflammatory site. Moreover, inducible NOS-/- mice were not susceptible to 15d-PGJ2-mediated suppression of neutrophil migration to the inflammatory sites when compared with their wild type. In addition, 15d-PGJ2-mediated suppression of neutrophil migration appeared to be independent of the production of cytokines and chemokines, since their production were not significantly affected in the carrageenan-injected peritoneal cavities. Finally, up-regulation of carrageenan-triggered ICAM-1 expression in the mesenteric microcirculation vessels was abrogated by pretreatment of wild-type mice with 15d-PGJ2, whereas 15d-PGJ2 inhibited F-actin rearrangement process in neutrophils. Taken together these findings demonstrated that 15d-PGJ2 suppresses inflammation-initiated neutrophil migration in a mechanism dependent on NO production in mesenteric tissues.
Collapse
Affiliation(s)
- Marcelo H Napimoga
- Laboratory of Molecular Biology, University of Uberaba, Minas Gerais, Brazil.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Li J, Liu NF, Wei Q. Effect of Rosiglitazone on Cardiac Fibroblast Proliferation, Nitric Oxide Production and Connective Tissue Growth Factor Expression Induced by Advanced Glycation End-products. J Int Med Res 2008; 36:329-35. [PMID: 18380944 DOI: 10.1177/147323000803600216] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
This study investigated the effects of the peroxisome proliferator-activated receptor-γ agonist rosiglitazone (RGZ) on cardiac fibroblast proliferation, nitric oxide content and connective tissue growth factor (CTGF) expression following incubation with advanced glycation end-products (AGEs). Cultured neonatal rat cardiac fibroblasts were incubated with various concentrations of AGEs for 48 h. Cells were also incubated with 200 mg/l AGEs plus various concentrations of RGZ. Cardiac fibroblast proliferation and cell cycle status were detected using a 3-(4, 5dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) assay and flow cytometry. Western blotting was used to measure the expression of CTGF and nitric oxide content was evaluated using a nitrate reductase assay. AGEs significantly accelerated proliferation, increased CTGF expression and decreased nitric oxide production in cardiac fibroblasts. These effects of AGEs were inhibited by RGZ in a dose-dependent manner. Treatment with RGZ could be a valuable therapeutic approach in diabetic patients with myocardial fibrosis.
Collapse
Affiliation(s)
- J Li
- Department of Geriatrics, Zhongda Hospital of Southeast University, Nanjing 21009, Jiangsu Province, People's Republic of China
| | - NF Liu
- Department of Cardiology, Zhongda Hospital of Southeast University, Nanjing 21009, Jiangsu Province, People's Republic of China
| | - Q Wei
- Department of Cardiology, Zhongda Hospital of Southeast University, Nanjing 21009, Jiangsu Province, People's Republic of China
| |
Collapse
|
31
|
Chen YJ, Quilley J. Fenofibrate treatment of diabetic rats reduces nitrosative stress, renal cyclooxygenase-2 expression, and enhanced renal prostaglandin release. J Pharmacol Exp Ther 2007; 324:658-63. [PMID: 17993607 DOI: 10.1124/jpet.107.129197] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Renal cyclooxygenase (COX)-2 expression is increased in the diabetic rat and has been linked to increased glomerular filtration rate (GFR) and renal injury. Our studies indicate that oxidative stress in the form of peroxynitrite (ONOO) may be the stimulus for induction of COX-2. In this study, we addressed the effects of a peroxisome proliferator-activated receptor alpha agonist on renal COX-2 expression as fibrates exert renal protective effects. Forty-eight hours after the induction of diabetes with streptozotocin in male Wistar rats, fenofibrate treatment (100 mg/kg/day) was started, and the effects were compared with untreated diabetic rats and treated and untreated age-matched control rats (n = 5 per group). After 12 to 14 weeks of treatment, the right kidney was perfused to determine prostaglandin release in response to arachidonic acid (AA), and the left kidney was used to examine the expression of COX-2 and nitrotyrosine, an index of ONOO formation. Release of prostaglandin (PG) E(2) in response to AA was enhanced in the diabetic rat kidney compared with control (4.8 +/- 0.7 versus 1.9 +/- 0.7 ng/min) and reduced by fenofibrate to 0.6 +/- 0.2 ng/min. A similar pattern was obtained for AA-stimulated release of 6-ketoPGF(1alpha). The effects of fenofibrate were associated with reduced renal expression of COX-2 and nitrotyrosine in diabetic rats. We used creatinine clearance as an index of GFR, which was increased in the diabetic rat, 3.09 +/- 0.4 versus 1.15 +/- 0.1 ml/min for control, and reduced by fenofibrate treatment to 1.87 +/- 0.3 ml/min. These results show that fenofibrate treatment of diabetic rats decreases renal COX-2 expression, possibly by reducing nitrosative stress, and is associated with a reduction of the enhanced GFR.
Collapse
Affiliation(s)
- Yu-Jung Chen
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, USA
| | | |
Collapse
|
32
|
Paukkeri EL, Leppänen T, Sareila O, Vuolteenaho K, Kankaanranta H, Moilanen E. PPARalpha agonists inhibit nitric oxide production by enhancing iNOS degradation in LPS-treated macrophages. Br J Pharmacol 2007; 152:1081-91. [PMID: 17891158 PMCID: PMC2095111 DOI: 10.1038/sj.bjp.0707477] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND AND PURPOSE Nitric oxide (NO) production through the inducible nitric oxide synthase (iNOS) pathway is increased in response to pro-inflammatory cytokines and bacterial products. In inflammation, NO has pro-inflammatory and regulatory effects. Peroxisome proliferator-activated receptors (PPARs), members of the nuclear steroid receptor superfamily, regulate not only metabolic but also inflammatory processes. The aim of the present study was to investigate the role of PPARalpha in the regulation of NO production and iNOS expression in activated macrophages. EXPERIMENTAL APPROACH The effects of PPARalpha agonists were investigated on iNOS mRNA and protein expression, on NO production and on the activation of transcription factors NF-kappaB and STAT1 in J774 murine macrophages exposed to bacterial lipopolysaccharide (LPS). KEY RESULTS PPARalpha agonists GW7647 and WY14643 reduced LPS-induced NO production in a dose-dependent manner as measured by the accumulation of nitrite into the culture medium. However, PPARalpha agonists did not alter LPS-induced iNOS mRNA expression or activation of NF-kappaB or STAT1 which are important transcription factors for iNOS. Nevertheless, iNOS protein levels were reduced by PPARalpha agonists in a time-dependent manner. The reduction was markedly greater after 24 h incubation than after 8 h incubation. Treatment with the proteasome inhibitors, lactacystin or MG132, reversed the decrease in iNOS protein levels caused by PPARalpha agonists. CONCLUSIONS AND IMPLICATIONS The results suggest that PPARalpha agonists reduce LPS-induced iNOS expression and NO production in macrophages by enhancing iNOS protein degradation through the proteasome pathway. The results offer an additional mechanism underlying the anti-inflammatory effects of PPARalpha agonists.
Collapse
Affiliation(s)
- E-L Paukkeri
- The Immunopharmacology Research Group, Medical School, University of Tampere and Research Unit, Tampere University Hospital Tampere, Finland
| | - T Leppänen
- The Immunopharmacology Research Group, Medical School, University of Tampere and Research Unit, Tampere University Hospital Tampere, Finland
| | - O Sareila
- The Immunopharmacology Research Group, Medical School, University of Tampere and Research Unit, Tampere University Hospital Tampere, Finland
| | - K Vuolteenaho
- The Immunopharmacology Research Group, Medical School, University of Tampere and Research Unit, Tampere University Hospital Tampere, Finland
| | - H Kankaanranta
- The Immunopharmacology Research Group, Medical School, University of Tampere and Research Unit, Tampere University Hospital Tampere, Finland
| | - E Moilanen
- The Immunopharmacology Research Group, Medical School, University of Tampere and Research Unit, Tampere University Hospital Tampere, Finland
- Author for correspondence:
| |
Collapse
|
33
|
Risé P, Ghezzi S, Carissimi R, Mastromauro F, Petroni A, Galli C. Delta5 desaturase mRNA levels are increased by simvastatin via SREBP-1 at early stages, not via PPARalpha, in THP-1 cells. Eur J Pharmacol 2007; 571:97-105. [PMID: 17655842 DOI: 10.1016/j.ejphar.2007.06.021] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2006] [Revised: 06/05/2007] [Accepted: 06/07/2007] [Indexed: 10/23/2022]
Abstract
In addition to inhibiting cholesterol biosynthesis, statins increase the conversion of linoleic acid to its derivatives, in particular to arachidonic acid, both in vivo and in vitro. Desaturases are the rate-limiting enzymes in this metabolic process and statins markedly enhance delta5 desaturase activity. To evaluate the delta5 desaturase gene expression and the transcription factors involved, THP-1 cells (a monocytic cell line) were incubated with 5 microM simvastatin for different time periods. The activity of the enzyme, evaluated as product/precursor ratio in the metabolic pathway (starting from [1-(14)C] linoleic acid), increased in treated cells with respect to controls after 24 h, whereas, mRNA levels of the delta5 desaturase increased after 12 h of incubation with simvastatin. Fatty acid desaturase genes are regulated by both sterol regulatory element binding proteins (SREBPs) and peroxisome proliferators activated receptors (PPARs). Both PPARalpha (WY 14643 and fenofibrate) and PPARgamma (ciglitazone) agonists did not affect linoleic acid conversion and the delta5 desaturase activity at any time considered (8-48 h), but they increased the delta5 desaturase mRNA levels, after 48 h; only fenofibrate showed a synergistic effect with simvastatin at this time, with a concomitantly increase in PPARalpha expression and beta-oxidation. Simvastatin alone increased SREBP-1 levels with respect to controls, starting from 8 h of incubation, whereas PPARalpha and linoleic acid beta-oxidation (a PPARalpha mediated process) were not affected after 48 h of incubation. These results taken together suggest that SREBP-1 is involved in the early regulation of delta5 desaturase gene by simvastatin, in THP-1 cells.
Collapse
Affiliation(s)
- Patrizia Risé
- Department of Pharmacological Sciences, University of Milan, via Balzaretti 9, 20133 Milan, Italy.
| | | | | | | | | | | |
Collapse
|
34
|
Woster AP, Combs CK. Differential ability of a thiazolidinedione PPARgamma agonist to attenuate cytokine secretion in primary microglia and macrophage-like cells. J Neurochem 2007; 103:67-76. [PMID: 17573821 DOI: 10.1111/j.1471-4159.2007.04706.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Peroxisome proliferator-activated receptor gamma (PPARgamma) agonists are known to inhibit select pro-inflammatory changes in models of CNS and systemic inflammation. Recent reports suggest that these anti-inflammatory effects are due to mechanisms other than canonical nuclear receptor-mediated transcriptional alteration. Using primary microglia and the monocytic cell line, THP-1, we demonstrate that rosiglitazone, a PPARgamma-activating thiazolidinedione, decreases pro-inflammatory cytokine secretion as measured by ELISA. Cells were pre-treated with various thiazolidinediones, including rosiglitazone, prior to stimulation with lipopolysaccharide or phorbol 12-myristate 13-acetate (PMA) to stimulate cytokine production. Tumor necrosis factor alpha (TNFalpha) secretion was significantly inhibited in both primary microglia and THP-1 cells differentiated for 72 h in the presence of PMA to induce a macrophage-like phenotype. No reduction in TNFalpha secretion was observed in undifferentiated THP-1 cells with rosiglitazone pre-treatment. Electrophoretic mobility shift assay revealed no significant difference in PPARgamma activation between PMA-differentiated and undifferentiated THP-1 cells. When PMA-differentiated and undifferentiated THP-1 cells were treated with the irreversible PPARgamma antagonist, GW 9662, a significant, dose-dependent decrease in TNFalpha secretion was observed. These results suggest that the anti-inflammatory benefit of PPARgamma ligands occur independently of classical PPARgamma activation.
Collapse
Affiliation(s)
- Andrew P Woster
- Department of Pharmacology, Physiology & Therapeutics, University of North Dakota, School of Medicine and Health Sciences, Grand Forks, North Dakota, USA
| | | |
Collapse
|
35
|
Yousefipour Z, Hercule H, Truong L, Oyekan A, Newaz M. Ciglitazone, a Peroxisome Proliferator-Activated Receptor γ Inducer, Ameliorates Renal Preglomerular Production and Activity of Angiotensin II and Thromboxane A2 in Glycerol-Induced Acute Renal Failure. J Pharmacol Exp Ther 2007; 322:461-8. [PMID: 17494862 DOI: 10.1124/jpet.107.122473] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Peroxisome proliferator-activated receptor gamma (PPARgamma), a nuclear transcription factor, modulates vascular responses to angiotensin II (AII) or thromboxane A(2) (TxA(2)) via regulation of their gene/receptor. Increased vasoconstriction and deteriorating renal function in glycerol-induced acute renal failure (ARF) may be attributed to down-regulation of PPARgamma. In this study, we investigated the effect of ciglitazone (CG), a PPARgamma inducer, on AII and TxA(2) production and activity in glycerol-induced ARF. Vascular responses to AII or 9,11-dideoxy-11alpha,9alpha-epoxymethano prostaglandin F(2alpha) (U46619), a TxA(2) mimetic, were determined in preglomerular vessels following induction of ARF with glycerol. Renal damage and function were assessed in CG-treated (9 nmol/kg for 21 days) rats. PPARgamma protein expression and activity, which were significantly lower in ARF rats, were enhanced by CG (26 and 30%). CG also increased PPARgamma mRNA by 67 +/- 6%, which was reduced in ARF. In ARF, there was significant tubular necrosis and apoptosis, a 5-fold increase in proteinuria and a 2-fold enhancement in vasoconstriction to AII and U46619. CG reduced proteinuria (49 +/- 3%), enhanced Na(+) (124 +/- 35%) and creatinine excretion (92 +/- 25%), markedly diminished tubular necrosis, and reduced ARF-induced increase in AII (40 +/- 3%) and TxA(2) (39 +/- 2%) production, the attending increase in vasoconstriction to AII (36 +/- 2%) and U46619 (50 +/- 11%), and the increase in angiotensin receptor-1 (AT(1)) (23 +/- 3%) or thromboxane prostaglandin (TP) receptor (13 +/- 1%). CG reduced free radical generation by 55 +/- 14% while elevating nitrite excretion (65 +/- 13%). Our results suggest that enhanced activity of AII and TxA(2), increased AT(1) or TP receptor expression, and renal injury in glycerol-induced ARF are consequent to down-regulation of PPARgamma gene. CG ameliorated glycerol-induced effects through maintaining PPARgamma gene.
Collapse
Affiliation(s)
- Zivar Yousefipour
- Center for Cardiovascular Diseases, Texas Southern University, 3100 Cleburne Avenue, Houston, TX 77004, USA
| | | | | | | | | |
Collapse
|
36
|
Zhao X, Zhang Y, Strong R, Zhang J, Grotta JC, Aronowski J. Distinct patterns of intracerebral hemorrhage-induced alterations in NF-κB subunit, iNOS, and COX-2 expression. J Neurochem 2006; 101:652-63. [PMID: 17250675 DOI: 10.1111/j.1471-4159.2006.04414.x] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Transcription factor nuclear factor-kappaB (NF-kappaB), plays a key role in regulating inflammation in brain pathologies. The goal of this study was to characterize temporal changes in NF-kappaB activation, NF-kappaB subunit expression, and expression of selected NF-kappaB-regulated gene products [inducible form of nitric oxide synthase (iNOS) and cyclooxygenase-2], at the transcriptional and translational level in the brain after intracerebral hemorrhage (ICH). Employing the intrastriatal injection of autologous blood in rats to model ICH, we demonstrated, using NF-kappaB-DNA binding assay, a robust and prolonged NF-kappaB activation, starting as early as 15 min after the onset of ICH. Consequently, we demonstrated that the mRNA and protein for p50, p52, p65, c-Rel, and RelB NF-kappaB subunits, as well as IkappaBalpha were all up-regulated, with a time course ranging from minutes to days following ICH, depending on the subunit. Using reverse transcription-polymerase chain reaction to analyze mRNA and immunoblotting to analyze protein in ICH-affected tissue, we found robust induction of iNOS at both mRNA and protein levels that followed a time-course similar to changes in p65, p52, and RelB mRNA. Oddly, in contrast to iNOS, cyclooxygenase-2 mRNA and protein following an early transient increase demonstrated significant reduction in response to ICH. In summary, NF-kappaB activation occurs within minutes and persists for at least a week in response to ICH. This reaction utilizes different NF-kappaB regulatory subunits and is associated with the expression of selected target genes.
Collapse
Affiliation(s)
- Xiurong Zhao
- Department of Neurology, Stroke Program, University of Texas-Houston Medical School, Houston, Texas 77030, USA
| | | | | | | | | | | |
Collapse
|
37
|
Klopotek A, Hirche F, Eder K. PPAR gamma ligand troglitazone lowers cholesterol synthesis in HepG2 and Caco-2 cells via a reduced concentration of nuclear SREBP-2. Exp Biol Med (Maywood) 2006; 231:1365-72. [PMID: 16946405 DOI: 10.1177/153537020623100810] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Cholesterol synthesis in animal cells is regulated by sterol regulatory element-binding protein (SREBP)-2. The objective of this study was to investigate whether activation of peroxisome proliferator-activatedreceptor (PPAR)-gamma influences the SREBP-2 dependent cholesterol synthesis in liver and intestinal cells. Therefore, HepG2 and Caco-2 cells were incubated with and without 10 or 30 microM of troglitazone, a synthetic PPAR gamma agonist, for 4 hrs. Incubation with 10 or 30 microM of troglitazone caused a significant, dose-dependent reduction of cholesterol synthesis in both HepG2 and Caco-2 cells (P < 0.05). HepG2 and Caco-2 cells incubated with 10 or 30 microM of troglitazone had also lower mRNA concentrations and lower nuclear protein concentrations of SREBP-2 than untreated control cells (P < 0.05). mRNA concentrations of the SREBP-2 target genes HMG-CoA reductase and LDL receptor were also reduced in HepG2 and Caco-2 cells treated with 30 microM of troglitazone compared to control cells (P < 0.05). In conclusion, this study shows that PPAR gamma activation by troglitazone lowers the cholesterol synthesis in HepG2 and Caco-2 cells by reducing the concentration of nuclear SREBP-2 and successive downregulation of its target genes involved in cholesterol synthesis.
Collapse
Affiliation(s)
- Anett Klopotek
- Institute of Nutritional Sciences, Martin-Luther-University Halle-Wittenberg, D-06108 Halle/Saale, Germany
| | | | | |
Collapse
|
38
|
Ou Z, Zhao X, Labiche LA, Strong R, Grotta JC, Herrmann O, Aronowski J. Neuronal expression of peroxisome proliferator-activated receptor-gamma (PPARγ) and 15d-prostaglandin J2—Mediated protection of brain after experimental cerebral ischemia in rat. Brain Res 2006; 1096:196-203. [PMID: 16725118 DOI: 10.1016/j.brainres.2006.04.062] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2006] [Revised: 04/04/2006] [Accepted: 04/10/2006] [Indexed: 12/29/2022]
Abstract
Existing experimental evidence suggests that PPARgamma may play a beneficial role in neuroprotection from various brain pathologies. Here we found that focal cerebral ischemia induced by middle cerebral/common carotid arteries occlusion (MCA/CCAo) induced up-regulation of PPARgamma messenger RNA in the ischemic hemisphere as early as 6 h after the ischemic event. The increased PPARgamma mRNA expression was primarily associated with neurons in the ischemic penumbra, suggesting an important role for PPARgamma in neurons after ischemia. Intraventricular injection of 15d-Delta(12,14)-prostaglandin J(2) (15d-PGJ(2)), a proposed endogenous PPARgamma agonist, into the ischemic rat brains significantly increased the PPARgamma-DNA-binding activity and reduced infarction volume at 24 h after reperfusion. We propose that PPARgamma up-regulation in response to ischemia may contribute to PPARgamma activation in the presence of PPARgamma agonists. Activation of PPARgamma in neurons at an early stage after ischemia may represent a pro-survival mechanism against ischemic injury.
Collapse
Affiliation(s)
- Zhishuo Ou
- University of Texas Health Science Center-Houston, Medical School, Department of Neurology, Stroke Program, Houston, 77030, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
Zhao X, Zhang Y, Strong R, Grotta JC, Aronowski J. 15d-Prostaglandin J2 activates peroxisome proliferator-activated receptor-gamma, promotes expression of catalase, and reduces inflammation, behavioral dysfunction, and neuronal loss after intracerebral hemorrhage in rats. J Cereb Blood Flow Metab 2006; 26:811-20. [PMID: 16208315 DOI: 10.1038/sj.jcbfm.9600233] [Citation(s) in RCA: 187] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Peroxisome proliferator-activated receptor-gamma (PPARgamma) is a transcription factor that regulates the expression of various gene products that are essential in lipid and glucose metabolism, as well as that of the peroxisome-enriched antioxidant enzyme, catalase. Activation of PPARgamma is linked to anti-inflammatory activities and is beneficial for cardiovascular diseases. However, little is known about its role in intracerebral hemorrhage (ICH). 15-Deoxy-Delta(12,14)-prostaglandin J2 (15d-PGJ2) acts as a physiologic agonist for PPARgamma. In this study, we found that injection of 15d-PGJ2 into the locus of striatal hematoma increased PPARgamma-deoxyribonucleic acid (DNA) binding activity and the expression of catalase messenger ribonucleic acid (mRNA) and protein in the perihemorrhagic area. Additionally, 15d-PGJ2 significantly reduced nuclear factor-kappaB (NF-kappaB) activation and prevented neutrophil infiltration measured by myeloperoxidase (MPO) immunoassay, and also reduced cell apoptosis measured by terminal deoxynucleotide transferase dUTP nick-end labeling (TUNEL). In addition, 15d-PGJ2 reduced behavioral dysfunction produced by the ICH. Altogether, our findings indicate that injection of 15d-PGJ2 at the onset of ICH is associated with activation of PPARgamma and elevation of catalase expression, suppression of NF-kappaB activity, and restricted neutrophil infiltration. All these events predicted reduced behavioral deficit and neuronal damage.
Collapse
Affiliation(s)
- Xiurong Zhao
- Stroke Program, Department of Neurology, University of Texas - Houston Medical School, Houston, Texas 77030, USA
| | | | | | | | | |
Collapse
|
40
|
Gagné F, Blaise C, Fournier M, Hansen PD. Effects of selected pharmaceutical products on phagocytic activity in Elliptio complanata mussels. Comp Biochem Physiol C Toxicol Pharmacol 2006; 143:179-86. [PMID: 16533621 DOI: 10.1016/j.cbpc.2006.01.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2005] [Revised: 01/16/2006] [Accepted: 01/22/2006] [Indexed: 10/24/2022]
Abstract
Municipal wastewaters are recognized as a major source of pharmaceutical and personal care products to the aquatic environment, thereby exposing biota to unknown chronic effects. This study sought to examine the immunotoxic effects of pharmaceutical and urban waste products on the freshwater mussel Elliptio complanata. Hemolymph samples were collected and treated in vitro with increasing concentrations of various drugs (bezafibrate, carbamazepine, fluoxetine, gemfibrozil, morphine, naproxen, novobiocin, oxytetracycline, sulfamethazole, sulfapyridine and trimethoprim) and urban waste related chemicals (coprostanol, caffeine, cotinine) for 24 h at 15 degrees C. In a parallel experiment, mussels were caged and placed in two final aeration lagoons for the treatment of domestic wastewaters. At the end of the exposure period, hemolymphs were tested for phagocytic activity, intracellular esterase activity, cell adherence and lipid peroxidation (LPO). The products that most increased phagocytosis were bezafibrate, gemfibrozil and trimethoprim, while novobiocin and morphine reduced its activity. Intracellular esterase activity was reduced most strongly with sulfamethazole, novobiocin, gemfibrozil, bezafibrate and carbamazepine. Cell adherence was decreased by oxytetracycline, novobiocin and naproxen, and increased by gemfibrozil, bezafibrate and sulfapyridine. Exposure to these products also modulated LPO in hemocytes. Coprostanol and naproxen were more potent to reduce LPO while novobiocin and sulfapyridine were the most potent to induce LPO. The potential to induce LPO was positively correlated with the number of functional groups on the molecule (i.e., its nucleophilicity). Mussels exposed to domestic wastewater treatment plant aeration lagoons had decreased intracellular esterase and phagocytic activity as well, suggesting immunosuppression. PPCPs (pharmaceuticals and personal care products) that are recognized to disrupt cytokine signalling network by the nitric oxide pathway and cell permeability were generally the most potent ones. The data suggest that PPCPs have the potential to cause adverse effects on the immune system of bivalves.
Collapse
Affiliation(s)
- F Gagné
- Environment Canada, St. Lawrence Centre, Montreal, Quebec.
| | | | | | | |
Collapse
|
41
|
Dobrian AD. The complex role of PPARgamma in renal dysfunction in obesity: managing a Janus-faced receptor. Vascul Pharmacol 2006; 45:36-45. [PMID: 16716756 DOI: 10.1016/j.vph.2006.01.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2006] [Revised: 01/01/2006] [Accepted: 01/01/2006] [Indexed: 01/11/2023]
Abstract
Obesity is frequently accompanied by insulin resistance, type II diabetes, hypertension and atherosclerosis, a cluster of pathologies that are the major components of the metabolic syndrome. Obesity is a known cause for renal dysfunction that leads to two major renal pathologies: hypertension and glomerular and tubulointerstitial injury. Peroxizome proliferator activated receptors (PPARs) are transcription factors belonging to the nuclear hormone receptor superfamily with important functions in the regulation of metabolism. The role of PPARgamma isoforms in adipogenesis and vascular inflammation associated to obesity has been vastly studied and is well recognized, albeit not completely mechanistically understood. Also, the effect of various PPARgamma agonists on blood pressure reduction in different forms of hypertension, including obesity related hypertension has been reported, but the mechanisms involved are only beginning to be studied. Even less clear is the concurrent beneficial effect of PPARgamma agonists thiazolinendiones (TZD) on blood pressure reduction in different forms of hypertension and, at the same time, in some cases, the significant water retention leading to edema and heart failure. The occurrence of both these apparently opposite effects on the renal water and sodium handling suggests a complex role of PPARgamma in the kidney that is likely related to the metabolic state. Also, PPARgamma activation leads to a reduction in mesangial cell proliferation while stimulating apoptosis. TZD treatment reduces albuminuria in obese and diabetic humans and rodent models suggesting protective effects against renal tubuloglomerular injury. The focus of this review is to present and critically discuss the recent findings on the roles of PPARgamma in the kidney in direct relation to renal function and renal injury in obesity and obesity-initiated diabetes.
Collapse
Affiliation(s)
- Anca Dana Dobrian
- Department of Physiological Sciences, Eastern Virginia Medical School, 700W Olney Road, Lewis Hall, Room 2027, Norfolk, VA 23507, USA.
| |
Collapse
|
42
|
Park CW, Zhang Y, Zhang X, Wu J, Chen L, Cha DR, Su D, Hwang MT, Fan X, Davis L, Striker G, Zheng F, Breyer M, Guan Y. PPARα agonist fenofibrate improves diabetic nephropathy in db/db mice. Kidney Int 2006; 69:1511-7. [PMID: 16672921 DOI: 10.1038/sj.ki.5000209] [Citation(s) in RCA: 152] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Peroxisome proliferator-activated receptor alpha (PPARalpha) is a member of the ligand-activated nuclear receptor superfamily, and plays an important role in lipid metabolism and glucose homeostasis. The purpose of this study is to determine whether the activation of PPARalpha by fenofbrate would improve diabetes and its renal complications in type II diabetes mellitus. Male C57 BLKS db/db mice and db/m controls at 8 weeks of age were divided to receive either a regular diet chow (db/db, n=8; db/m, n=6) or a diet containing fenofibrate (db/db, n=8; db/m, n=7). Mice were followed for 8 weeks. Fenofibrate treatment dramatically reduced fasting blood glucose (P<0.001) and HbA1c levels (P<0.001), and was associated with decreased food intake (P<0.01) and slightly reduced body weight. Fenofibrate also ameliorated insulin resistance (P<0.001) and reduced plasma insulin levels (P<0.05) in db/db mice. Hypertrophy of pancreatic islets was decreased and insulin content markedly increased (P<0.05) in fenofibrate-treated diabetic animals. In addition, fenofibrate treatment significantly reduced urinary albumin excretion (P<0.001). This was accompanied by dramatically reduced glomerular hypertrophy and mesangial matrix expansion. Furthermore, the addition of fenofibrate to cultured mesangial cells, which possess functional active PPARalpha, decreased type I collagen production. Taken together, the PPARalpha agonist fenofibrate dramatically improves hyperglycemia, insulin resistance, albuminuria, and glomerular lesions in db/db mice. The activation of PPARalpha by fenofibrate in mesangial cells may partially contribute to its renal protection. Thus, fenofibrate may serve as a therapeutic agent for type II diabetes and diabetic nephropathy.
Collapse
Affiliation(s)
- C W Park
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Zhao X, Quigley JE, Yuan J, Wang MH, Zhou Y, Imig JD. PPAR-alpha activator fenofibrate increases renal CYP-derived eicosanoid synthesis and improves endothelial dilator function in obese Zucker rats. Am J Physiol Heart Circ Physiol 2006; 290:H2187-95. [PMID: 16501022 DOI: 10.1152/ajpheart.00937.2005] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Previous studies have shown that the synthesis of renal cytochrome P-450 (CYP)-derived eicosanoids is downregulated in genetic or high-fat diet-induced obese rats. Experiments were designed to determine whether fenofibrate, a peroxisome proliferator-activated receptor (PPAR)-alpha agonist, would induce renal eicosanoid synthesis and improve endothelial function in obese Zucker rats. Administration of fenofibrate (150 mg.kg(-1).day(-1) for 4 wk) significantly reduced plasma insulin, triglyceride, and total cholesterol levels in obese Zucker rats. CYP2C11 and CYP2C23 proteins were downregulated in renal vessels of obese Zucker rats. Consequently, renal vascular epoxygenase activity decreased by 15% in obese Zucker rats compared with lean controls. Chronic fenofibrate treatment significantly increased renal cortical and vascular CYP2C11 and CYP2C23 protein levels in obese Zucker rats, whereas it had no effect on epoxygenase protein and activity in lean Zucker rats. Renal cortical and vascular epoxygenase activities were consequently increased by 54% and 18%, respectively, in fenofibrate-treated obese rats. In addition, acetylcholine (1 microM)-induced vasodilation was significantly reduced in obese Zucker kidneys (37% +/- 11%) compared with lean controls (67% +/- 9%). Chronic fenofibrate administration increased afferent arteriolar responses to 1 microM of acetylcholine in obese Zucker rats (69% +/- 4%). Inhibition of the epoxygenase pathway with 6-(2-propargyloxyphenyl)hexanoic acid attenuated afferent arteriolar diameter responses to acetylcholine to a greater extent in lean compared with obese Zucker rats. These results demonstrate that the PPAR-alpha agonist fenofibrate increased renal CYP-derived eicosanoids and restored endothelial dilator function in obese Zucker rats.
Collapse
Affiliation(s)
- Xueying Zhao
- Vascular Biology Center, Medical College of Georgia, Augusta, GA 30912-2500.
| | | | | | | | | | | |
Collapse
|
44
|
Zhao X, Ou Z, Grotta JC, Waxham N, Aronowski J. Peroxisome-proliferator-activated receptor-gamma (PPARgamma) activation protects neurons from NMDA excitotoxicity. Brain Res 2006; 1073-1074:460-9. [PMID: 16442504 DOI: 10.1016/j.brainres.2005.12.061] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2005] [Revised: 12/01/2005] [Accepted: 12/05/2005] [Indexed: 10/25/2022]
Abstract
A growing body of evidence indicates that the transcription factor PPARgamma plays a beneficial role in various neurological diseases. The postulated principal mechanism underlying the beneficial effects of PPARgamma is due to its anti-inflammatory properties. However, PPARgamma exists in neurons where it may provide additional effects that regulate neuronal vulnerability. In the present study, we employed in vitro and in vivo models of excitotoxic neuronal injury to test hypothesis on the neuroprotective role of PPARgamma. The endogenous PPARgamma ligand, 15d-Delta(12,14)-Prostaglandin J2 (15d-PGJ2), and a selective thiazolidinedione PPARgamma agonist, ciglitazone, significantly reduced neuronal death in response to glutamate and NMDA-mediated, but not kainate-mediated toxicity. This neuroprotective effect of 15d-PGJ2 and ciglitazone was linked to increased PPARgamma DNA binding activity as it was fully reversed by the pretreatment of neurons with selective PPARgamma antagonists and anti-PPARgamma antibody. It was not due to the blockade of NMDA-receptor-mediated Ca++ entry. Our data demonstrate that PPARgamma activation may represent a potential target for treatment of numerous acute and chronic neurological diseases with pathologies that involve excitotoxic damage.
Collapse
Affiliation(s)
- Xiurong Zhao
- Stroke Program, Department of Neurology, University of Texas-Houston Medical School, 6431 Fannin, Rm. 7.044, Houston, TX 77030, USA
| | | | | | | | | |
Collapse
|
45
|
Abeles AM, Pillinger MH. Statins as antiinflammatory and immunomodulatory agents: A future in rheumatologic therapy? ACTA ACUST UNITED AC 2006; 54:393-407. [PMID: 16447216 DOI: 10.1002/art.21521] [Citation(s) in RCA: 110] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Aryeh M Abeles
- New York University School of Medicine, the Hospital for Joint Diseases, New York, New York, USA
| | | |
Collapse
|
46
|
Johann AM, von Knethen A, Lindemann D, Brüne B. Recognition of apoptotic cells by macrophages activates the peroxisome proliferator-activated receptor-gamma and attenuates the oxidative burst. Cell Death Differ 2005; 13:1533-40. [PMID: 16341123 DOI: 10.1038/sj.cdd.4401832] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
It is appreciated that phagocytosis of apoptotic cells (AC) is an immunological relevant process that shapes the pro- versus anti-inflammatory macrophage phenotype. It was our intention to study the respiratory burst, a prototype marker of macrophage activation, under the impact of AC. Following incubation of RAW264.7 macrophages with AC, we noticed attenuated production of reactive oxygen species (ROS) in response to PMA treatment, and observed a correlation between attenuated ROS formation and suppression of protein kinase Calpha (PKCalpha) activation. EMSA analysis demonstrated an immediate activation of peroxisome proliferator-activated receptor-gamma (PPARgamma) following supplementation of AC to macrophages. In macrophages carrying a dominant-negative PPARgamma mutant, recognition of AC no longer suppressed PKCalpha activation, and the initial phase of ROS formation was largely restored. Interference with actin polymerization and transwell experiments suggest that recognition of AC by macrophages suffices to attenuate the early phase of ROS formation that is attributed to PPARgamma activation.
Collapse
Affiliation(s)
- A M Johann
- Institute of Biochemistry I, Faculty of Medicine, Johann Wolfgang Goethe-University Frankfurt, Theodor-Stern-Kai 7, Frankfurt 60590, Germany
| | | | | | | |
Collapse
|
47
|
Abstract
Nuclear receptors are transcription factors that are essential in embryonic development, maintenance of differentiated cellular phenotypes, metabolism, and apoptosis. Dysfunction of nuclear receptor signaling leads to a wide spectra of proliferative, reproductive, and metabolic diseases, including cancers, infertility, obesity, and diabetes. In addition, many proteins have been identified as coregulators which can be recruited by DNA-binding nuclear receptors to affect transcriptional regulation. The cellular level of coregulators is crucial for nuclear receptor-mediated transcription and many coregulators have been shown to be targets for diverse intracellular signaling pathways and posttranslational modifications. This review provides a general overview of the roles and mechanism of action of nuclear receptors and their coregulators. Since progression of renal diseases is almost always associated with inflammatory processes and/or involve metabolic disorders of lipid and glucose, cell proliferation, hypertrophy, apoptosis, and hypertension, the importance of nuclear receptors and their coregulators in these contexts will be addressed.
Collapse
Affiliation(s)
- Xiong Z Ruan
- Centre for Nephrology, Royal Free and University College Medical School, University College London, Royal Free Campus, Rowland Hill Street, London, United Kingdom.
| | | | | | | |
Collapse
|
48
|
Stamatakis K, Sánchez-Gómez FJ, Pérez-Sala D. Identification of novel protein targets for modification by 15-deoxy-Delta12,14-prostaglandin J2 in mesangial cells reveals multiple interactions with the cytoskeleton. J Am Soc Nephrol 2005; 17:89-98. [PMID: 16291835 DOI: 10.1681/asn.2005030329] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
The cyclopentenone prostaglandin 15-deoxy-Delta12,14-PGJ2 (15d-PGJ2) has been shown to display protective effects against renal injury or inflammation. In cultured mesangial cells (MC), 15d-PGJ2 inhibits the expression of proinflammatory genes and modulates cell proliferation. Therefore, cyclopentenone prostaglandins (cyPG) have been envisaged as a promise in the treatment of renal disease. The effects of 15d-PGJ2 may be dependent on or independent from its role as a peroxisome proliferator-activated receptor agonist. It was shown recently that an important determinant for the peroxisome proliferator-activated receptor-independent effects of 15d-PGJ2 is the capacity to modify proteins covalently and alter their function. However, a limited number of protein targets have been identified to date. Herein is shown that a biotinylated derivative of 15d-PGJ2 recapitulates the effects of 15d-PGJ2 on the stress response and inhibition of inducible nitric oxide synthase levels and forms stable adducts with proteins in intact MC. Biotinylated 15d-PGJ2 was then used to identify proteins that potentially are involved in cyPG biologic effects. Extracts from biotinylated 15d-PGJ2-treated MC were separated by two-dimensional electrophoresis, and the spots of interest were analyzed by mass spectrometry. Identified targets include proteins that are regulated by oxidative stress, such as heat-shock protein 90 and nucleoside diphosphate kinase, as well as proteins that are involved in cytoskeletal organization, such as actin, tubulin, vimentin, and tropomyosin. Biotinylated 15d-PGJ2 binding to several targets was confirmed by avidin pull-down. Consistent with these findings, 15d-PGJ2 induced early reorganization of vimentin and tubulin in MC. The cyclopentenone moiety and the presence of cysteine were important for vimentin rearrangement. These studies may contribute to the understanding of the mechanism of action and therapeutic potential of cyPG.
Collapse
Affiliation(s)
- Konstantinos Stamatakis
- Departamento de Estructura y Función de Proteínas, Centro de Investigaciones Biológicas, C.S.I.C., Ramiro de Maeztu 9, 28040 Madrid, Spain
| | | | | |
Collapse
|
49
|
Ren S, Xin C, Beck KF, Saleem MA, Mathieson P, Pavenstädt H, Pfeilschifter J, Huwiler A. PPARalpha activation upregulates nephrin expression in human embryonic kidney epithelial cells and podocytes by a dual mechanism. Biochem Biophys Res Commun 2005; 338:1818-24. [PMID: 16288986 DOI: 10.1016/j.bbrc.2005.10.158] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2005] [Accepted: 10/21/2005] [Indexed: 11/18/2022]
Abstract
Nephrin is an important member of the glomerular ultrafiltration complex and changes in its expression are associated with severe proteinuria. In this study, we show that synthetic PPARalpha agonists, but not PPARgamma agonists, stimulate an increased nephrin mRNA and protein expression in cultures of human podocytes and A293 human embryonic kidney epithelial cells which are blocked by the PPARalpha antagonist Ru486. Furthermore, the PPARalpha agonists have an additive effect on the interleukin-1beta (IL-1beta)-induced nephrin upregulation. Luciferase-reporter assays reveal that human nephrin promoter activity is stimulated by the PPARalpha agonists. Neither IL-1beta nor TNFalpha alone has an effect on nephrin promoter activity suggesting that additional posttranscriptional regulatory events might be operative. The role of nephrin mRNA stability regulation was evaluated in cells treated with actinomycin D to stop further RNA transcription. In the presence of PPARalpha agonists, IL-1beta or TNFalpha, the decay of nephrin mRNA was drastically reduced thus arguing for an additional posttranscriptional mode of action. In summary, these data show that PPARalpha activation causes an increased nephrin expression by a dual action, on the one hand by stimulating nephrin promoter activity and on the other hand by reducing nephrin mRNA degradation. These findings may have importance for treatment strategies of renal diseases affecting the expression of nephrin and subsequently the proper action of the glomerular filtration apparatus.
Collapse
Affiliation(s)
- Shuyu Ren
- Pharmazentrum Frankfurt/ZAFES, Klinikum der Johann Wolfgang Goethe-Universität, Theodor-Stern-Kai 7, D-60590 Frankfurt am Main, Germany
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Kim J, Oh YS, Shinn SH. Troglitazone reverses the inhibition of nitric oxide production by high glucose in cultured bovine retinal pericytes. Exp Eye Res 2005; 81:65-70. [PMID: 15978256 DOI: 10.1016/j.exer.2005.01.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2004] [Revised: 01/12/2005] [Accepted: 01/16/2005] [Indexed: 12/01/2022]
Abstract
In the retinal microcirculation, there is a basal release of nitric oxide (NO) which maintains the retinal blood flow. The proportions of endothelial cells and pericytes in the retinal capillaries are almost equal, so pericytes appear to play a important role in the regulation of microcirculatory hemodynamics in the retina. It has been suggested that the pathogenesis of early diabetic retinopathy may involve a reduced bioavailability or diminished production of NO. In this study, we investigated the role of troglitazone, a potent agonist of peroxisome proliferator activated receptor-gamma (PPARgamma) used for the treatment of diabetes, on the NO release and the effect of exposure to high glucose on the production of NO in cultured bovine retinal pericytes. Troglitazone significantly increased NO production and iNOS expression after 24hr in a dose-and PPARgamma-dependent manner. Elevation of D-glucose, but not L-glucose, from 5.5 to 30 mm for 24 hr decreased NO production, but co-treatment with troglitazone reversed high glucose-induced inhibition of NO production as well as iNOS expression. In conclusion, high glucose inhibits iNOS expression and subsequently NO synthesis in cultured bovine retinal pericytes, and troglitazone restores the NO production.
Collapse
Affiliation(s)
- Jaetaek Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Chung-Ang University, Seoul, South Korea.
| | | | | |
Collapse
|