1
|
Mazhar MU, Naz S, Khan JZ, Azam S, Ghazanfar S, Tipu MK. Protective potential of Bacillus subtilis (NMCC-path-14) against extraarticular manifestations during acute and sub-acute phase of arthritis using mice model. Biochem Biophys Res Commun 2024; 733:150708. [PMID: 39298918 DOI: 10.1016/j.bbrc.2024.150708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 08/29/2024] [Accepted: 09/14/2024] [Indexed: 09/22/2024]
Abstract
Extra-articular manifestations (EAM), which are associated with rheumatoid arthritis (RA), affect the quality of life of patients and are one of the critical causes of early mortality. This study was aimed at investigating whether Bacillus subtilis NMCC-path-14 (1 × 108 CFU/animal/day) could serve as a valuable therapeutic agent in managing EAM using complete Freund's adjuvant (CFA) induced arthritis during acute and sub-acute phases. Arthritis was induced using intra-dermal administration of CFA in the right hind paw of mice on day 1. Dexamethasone (Dexa) (5 mg/kg/day/animal) was used as a standard treatment. Animals in Dexa and Bacillus subtilis concurrent treatment (BS-CT) received treatments on day 1. The Bacillus subtilis pre-treatment (BS-PT) group received a probiotic dose 7 days before arthritis induction. Parameters like body weight, relative organ weight, colon length, hematology, serum biochemistry, antioxidant capacity, and histopathology of liver, kidney, spleen, colon, stress-related behavioral changes, and cortisol levels were evaluated on days 7 (acute) and 14 (sub-acute). Dexa failed to manage the EAM in arthritic mice and instead exacerbated them. On the other hand, B. subtilis NMCC-path-14 significantly declined EAM with no notable side effects, highlighting its safety and effectiveness. The current data show that B. subtilis NMCC-path-14 may be an alternative option for arthritis treatment that can reduce systemic symptoms associated with arthritis. More studies are required to comprehend the underlying mechanisms of mitigating the EAM by B. subtilis NMCC-path-14.
Collapse
Affiliation(s)
- Muhammad Usama Mazhar
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.
| | - Sadaf Naz
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.
| | - Jehan Zeb Khan
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.
| | - Shahzad Azam
- Department of Pathology, Fazaia Medical College, Air University, Islamabad, Pakistan.
| | - Shakira Ghazanfar
- National Institute for Genomics and Advanced Biotechnology (NIGAB), National Agricultural Research Centre (NARC), Islamabad, Pakistan.
| | - Muhammad Khalid Tipu
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.
| |
Collapse
|
2
|
Mazhar MU, Naz S, Zulfiqar T, Khan JZ, Ghazanfar S, Tipu MK. Immunostimulant, hepatoprotective, and nephroprotective potential of Bacillus subtilis (NMCC-path-14) in comparison to dexamethasone in alleviating CFA-induced arthritis. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:3275-3299. [PMID: 37930392 DOI: 10.1007/s00210-023-02814-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 10/22/2023] [Indexed: 11/07/2023]
Abstract
To investigate and compare efficacy as well as safety of Bacillus subtilis and dexamethasone (Dexa) in complete Freund's adjuvant (CFA)-induced arthritis, we used glucocorticoid monotherapy (Dexa 5 mg/kg/day) and B. subtilis (1 × 108 CFU/animal/day p.o) as pre-treatment and concurrent treatment for a duration of 35 days. Specific emphasis was on chronic aspect of this study since long-term use of Dexa is known to produce undesirable side effects. Treatment with Dexa significantly attenuated the arthritic symptoms but produced severe side effects like weight loss, increased mortality, immunosuppression, and altered histology of liver, kidney, and spleen. Oxidative stress was also elevated by Dexa in these organs which contributed to the damage. Treatment with B. subtilis improved symptoms of arthritis without producing any deleterious side effects as seen with Dexa therapy. Immunohistochemistry (IHC) profile revealed decreased expression of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), interleukin (IL)-1β, tumor necrosis factor alpha (TNF-α), and increased nuclear factor erythroid 2-related factor 2 (Nrf-2) expression by B. subtilis and Dexa treatment in ankle joint of arthritic mice. Radiological scores were also improved by both treatments. This study concludes that B. subtilis could be an effective alternative for treating arthritis than Dexa since it does not produce life-threatening side effects on prolong treatment.
Collapse
Affiliation(s)
- Muhammad Usama Mazhar
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Sadaf Naz
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Tayyaba Zulfiqar
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Jehan Zeb Khan
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Shakira Ghazanfar
- National Institute for Genomics and Advanced Biotechnology (NIGAB), National Agricultural Research Centre (NARC), Islamabad, Pakistan
| | - Muhammad Khalid Tipu
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.
| |
Collapse
|
3
|
Soleimani AA, Shokri N, Elahimanesh M, Mohammadi P, Parvaz N, Bakhshandeh M, Najafi M. Beta arrestin-related signalling axes are influenced by dexamethasone and metformin in vascular smooth muscle cells cultured in high glucose condition. Endocrinol Diabetes Metab 2024; 7:e465. [PMID: 38102782 PMCID: PMC10782052 DOI: 10.1002/edm2.465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/27/2023] [Accepted: 12/03/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND Metformin (Met) and dexamethasone (Dexa) are known to reduce blood sugar levels and anti-inflammatory effects, respectively. Based on the acceleration of atherosclerosis process in diabetes, the β-arrestin 2 (BARR2) gene and protein expression levels were evaluated in vascular smooth muscle cells (VSMCs) treated with Met and Dexa in high glucose conditions in this study. METHODS AND MATERIALS Human VSMCs were cultured in Dulbecco's Modified Eagle Medium/Nutrient Mixture F-12 (DMEM-F12) medium and, were treated with different values of Met (1 mM, 5 mM and 7 mM) and Dexa (10-7 M, 10-6 M and 10-5 M) in 24- and 48-h periods. The BARR2 gene and protein expression levels were identified with RT-qPCR and western blotting techniques, respectively. The signalling axes were predicted from gene network made using Cytoscape software and, were annotated with Gene Ontology. RESULTS The BARR2 gene and protein expression levels reduced in VSMCs treated with Dexa and Met after 24- and 48-h periods. These results were more changed after 48 h. Furthermore, many BARR2-related signalling axes were found from the network genes. CONCLUSION Met and Dexa suppressed the BARR2 protein and gene expression levels in the VSMCs. Moreover, the gene network suggested some the cellular signalling axes related to BARR2 that may be affected by Met and Dexa.
Collapse
Affiliation(s)
- Ali Akbar Soleimani
- Department of Clinical Biochemistry, Faculty of MedicineTehran University of Medical SciencesTehranIran
| | - Nafiseh Shokri
- Department of Clinical Biochemistry, Faculty of MedicineIran University of Medical SciencesTehranIran
| | - Mohammad Elahimanesh
- Department of Clinical Biochemistry, Faculty of MedicineIran University of Medical SciencesTehranIran
| | - Payam Mohammadi
- Department of Clinical Biochemistry, Faculty of MedicineIran University of Medical SciencesTehranIran
| | - Najmeh Parvaz
- Department of Clinical Biochemistry, Faculty of MedicineIran University of Medical SciencesTehranIran
| | - Masoomeh Bakhshandeh
- Department of Clinical Biochemistry, Faculty of MedicineIran University of Medical SciencesTehranIran
| | - Mohammad Najafi
- Department of Clinical Biochemistry, Faculty of MedicineIran University of Medical SciencesTehranIran
- Cellular and Molecular Research CenterIran University of Medical SciencesTehranIran
- Microbial Biotechnology CenterIran University of Medical SciencesTehranIran
| |
Collapse
|
4
|
Soleimani AA, Mohammadi A, Ghasempour G, Abkenar BR, Shokri N, Najafi M. Dexamethasone suppresses the proliferation and migration of VSMCs by FAK in high glucose conditions. BMC Pharmacol Toxicol 2022; 23:63. [PMID: 35978346 PMCID: PMC9382766 DOI: 10.1186/s40360-022-00604-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 08/12/2022] [Indexed: 11/30/2022] Open
Abstract
Background High glucose conditions cause some changes in the vessels of diabetes through the signal transduction pathways. Dexamethasone and other corticosteroids have a wide range of biological effects in immunological events. In the present study, the effects of dexamethasone were investigated on the VSMC (vascular smooth muscle cell) proliferation, and migration based on the FAK gene and protein changes in high glucose conditions. Methods and materials The vascular smooth muscle cells were cultured in DMEM and were treated with dexamethasone (10–7 M, 10–6 M, and 10–5 M) for 24, and 48 h in high glucose conditions. The cell viability was estimated by MTT method. The FAK gene expression levels and pFAK protein values were determined by RT-qPCR and western blotting techniques, respectively. A scratch assay was used to evaluate cellular migration. Results The FAK gene expression levels decreased significantly dependent on dexamethasone doses at 24 and 48 h. The pFAK protein values decreased significantly with a time lag at 24- and 48-h periods as compared with gene expression levels. Conclusion The results showed that the inhibition of VSMC proliferation and migration by dexamethasone in the high glucose conditions may be related to the changes of FAK. Supplementary Information The online version contains supplementary material available at 10.1186/s40360-022-00604-3.
Collapse
Affiliation(s)
- Ali Akbar Soleimani
- Clinical Biochemistry Department, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Asghar Mohammadi
- Clinical Biochemistry Department, Faculty of Medicine, Tarbiat Modares University, Tehran, Iran
| | - Ghasem Ghasempour
- Clinical Biochemistry Department, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Borhan Rahimi Abkenar
- Clinical Biochemistry Department, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Nafiseh Shokri
- Clinical Biochemistry Department, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Najafi
- Clinical Biochemistry Department, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran. .,Microbial Biotechnology Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Wang XP, Li QL, Li W, Zhang T, Li XY, Jiao Y, Zhang XM, Jiang JJ, Zhang X, Zhang XM. Dexamethasone attenuated thoracic aortic aneurysm and dissection in vascular smooth muscle cell Tgfbr2 disrupted mice with CCL8 suppression. Exp Physiol 2022; 107:631-645. [PMID: 35344629 DOI: 10.1113/ep090190] [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: 11/01/2021] [Accepted: 03/23/2022] [Indexed: 11/08/2022]
Abstract
NEW FINDINGS What is the central question of this study? The aim of this study was to investigate the relationship of CCL8 and the thoracic aortic aneurysm and dissection (TAAD) formation in postnatal mice with vascular smooth muscle cell (VSMC) Tgfbr2 disruption and whether dexamethasone could be a potential treatment. What is the main finding and its importance? CCL8 was associated with the formation of TAAD in VSMC Tgfbr2 disrupted mice. Dexamethasone reduced TAAD formation and inhibited MAPK (p-p38) and NF-κB (p-p65) signaling pathways. CCL8 might be an important promoter in aortic inflammation. DEX provided potential therapeutic effects in TAAD treatment. ABSTRACT Aortic inflammation plays a vital role in initiation and progression of thoracic aortic aneurysm and dissection (TAAD). The disturbance of transforming growth factor-β (TGF-β) signaling pathway is believed to be one of the pathogenic mechanisms of TAAD. Initially, Myh11-CreERT2 .Tgfbr2f/f male mice were used to build TAAD mice model. And bioinformatics analyses revealed the enriched inflammatory signal pathways and upregulated chemokine CCL8. So we hypothesized that vascular smooth muscle cell (VSMC) Tgfbr2 disruption in postnatal mice resulted in aortic inflammation associated with CCL8 secretion. Then real-time quantitative PCR and serum ELISA results confirmed that CCL8 expression began to increase after VSMC Tgfbr2 disruption. Next, we cultured mouse thoracic aortas ex vivo, and observed that the protein expressions of CCL8 in culture supernatants were increased by ELISA. Subsequently, the co-localization of CCL8 with α-smooth muscle actin (α-SMA) orCD68 was found significantly increased by immunofluorescence. Then, dexamethasone (DEX) was used to treat TAAD in VSMC Tgfbr2 disrupted mice The results of histochemical, immunofluorescence and immunohistochemical staining indicated that DEX therapy reduced CCL8 secretion, inflammatory cell recruitment, aortic medial thickening, elastic fiber fragmentating, extracellular matrix degradation, contractile apparatus impairment, thereby ameliorated TAAD formation. Western blot showed that MAPK and NF-κB signaling pathways in aorta were overactivated after VSMC Tgfbr2 disruption, but inhibited by DEX therapy. Altogether, CCL8 might be an important promoter in TAAD formation of VSMC Tgfbr2 disrupted mice. And DEX provided potential therapeutic effects in TAAD treatment. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Xi-Peng Wang
- Department of Vascular Surgery, Peking University People's Hospital, Beijing, People's Republic of China
| | - Qing-Le Li
- Department of Vascular Surgery, Peking University People's Hospital, Beijing, People's Republic of China
| | - Wei Li
- Department of Vascular Surgery, Peking University People's Hospital, Beijing, People's Republic of China
| | - Tao Zhang
- Department of Vascular Surgery, Peking University People's Hospital, Beijing, People's Republic of China
| | - Xiao-Yan Li
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Disease, Beijing, People's Republic of China
| | - Yang Jiao
- Department of Vascular Surgery, Peking University People's Hospital, Beijing, People's Republic of China
| | - Xue-Min Zhang
- Department of Vascular Surgery, Peking University People's Hospital, Beijing, People's Republic of China
| | - Jing-Jun Jiang
- Department of Vascular Surgery, Peking University People's Hospital, Beijing, People's Republic of China
| | - Xiaoping Zhang
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Disease, Beijing, People's Republic of China
| | - Xiao-Ming Zhang
- Department of Vascular Surgery, Peking University People's Hospital, Beijing, People's Republic of China
| |
Collapse
|
6
|
Shuker E, Farhood M, Al-Qudaihi G, Fouad D. Potential Effects of Boldine on Oxidative Stress, Apoptosis, and Inflammatory Changes Induced by the Methylprednisolone Hepatotoxicity in Male Wistar Rats. Dose Response 2022; 20:15593258221082877. [PMID: 35360456 PMCID: PMC8961387 DOI: 10.1177/15593258221082877] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Accepted: 02/03/2022] [Indexed: 11/17/2022] Open
Abstract
Background Synthetic glucocorticoid therapeutic agent methylprednisolone (MPL), when used for an extended period of time at high dose, promotes the development of reactive oxygen species (ROS)-induced liver toxicity. This study investigated the role of boldine, a natural antioxidant with anti-apoptotic and anti-inflammatory properties, against MPL-induced hepatoxicity in male Wistar rats. Methods 120 rats were divided into eight equal groups: G1 (control), G2, 3, and 4 (rats orally administered 5, 10, and 50 mg boldine/kg b.w./day; respectively, for 28 days), G5 (rats intramuscularly injected with 100 mg MPL/kg b.w. only on the last three days), G6, 7, and 8 (rats administered boldine + MPL). After the last MPL injection, rats were sacrificed at intervals of 1, 24, and 48 h. Results There was a significant decrease in WBCs, RBCs count, and HGB levels, as well as an increase in PLT count, ALT, AST, TG, and LDL levels, and a decrease in HDL level in serum. Oxidative stress markers levels increased at all times, and gene expression of antioxidant enzymes increased at 24h. Immunohistochemical analysis revealed that cytochrome c levels significantly increased after MPL treatment. The COMET assay revealed detectable DNA lesions. There was no immune reactivity of IL-6 expressions as an inflammatory response marker. Conclusions Oral administration of boldine has a modulatory protective, antioxidant, and anti-apoptotic effect against free radicals.
Collapse
Affiliation(s)
- Esraa Shuker
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Manal Farhood
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Ghofran Al-Qudaihi
- Environmental Health Program, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Dalia Fouad
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
- Department of Zoology and Entomology, Faculty of Science, Helwan University, EinHelwan, Egypt
| |
Collapse
|
7
|
Liang R, Wong KH, Yang Y, Duan Y, Chen M. ROS-Responsive Dexamethasone Micelles Normalize the Tumor Microenvironment to Enhance Hypericin in Cancer Photodynamic Therapy. Biomater Sci 2022; 10:1018-1025. [DOI: 10.1039/d1bm01802g] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Efficacy of photodynamic therapy (PDT) for cancer is limited due to the abnormality of tumor microenvironment (TME), such as dysfunctional tumor vascular system leading to restrict the drug distribution in...
Collapse
|
8
|
Wang X, Zhang X, Qiu T, Yang Y, Li Q, Zhang X. Dexamethasone reduces the formation of thoracic aortic aneurysm and dissection in a murine model. Exp Cell Res 2021; 405:112703. [PMID: 34118251 DOI: 10.1016/j.yexcr.2021.112703] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 05/30/2021] [Accepted: 06/05/2021] [Indexed: 12/17/2022]
Abstract
Thoracic aortic aneurysm and dissection (TAAD) is a life-threatening vascular disease with no effective pharmaceutical therapies currently available. Inflammation plays a key role in the progression of aneurysms. Dexamethasone (DEX), a synthetic glucocorticoid, has showed alleviating effects on cells in vitro from TAAD patients. Here we performed a study aiming at investigating the protective role of DEX in a β-aminopropionitrile monofumarate (BAPN)-induced TAAD mouse model. DEX (dose: 0.04 mg/kg/day) treatment significantly reduced the aortic diameter and inhibited TAAD formation. DEX reduced infiltration of macrophages and neutrophils, apoptosis of vascular smooth muscle cells (VSMCs), expression of metalloproteinase 2/9, and extracellular matrix degradation in BAPN-treated TAAD mice. Furthermore, DEX therapy downregulated the expression of p-p65 in macrophages and VSMCs, which suggested that DEX might ameliorate BAPN-induced TAAD by suppressing NF-κB signaling. Therefore, DEX therapy attenuates the progression of BAPN-induced TAAD murine model and could be used as an effective adjuvant therapy for treating TAAD.
Collapse
Affiliation(s)
- Xipeng Wang
- Department of Vascular Surgery, Peking University People's Hospital, Beijing, China.
| | - Xiaoping Zhang
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Disease, Beijing, China.
| | - Tao Qiu
- Department of Vascular Surgery, Peking University People's Hospital, Beijing, China.
| | - Yang Yang
- Department of Vascular Surgery, Peking University People's Hospital, Beijing, China.
| | - Qingle Li
- Department of Vascular Surgery, Peking University People's Hospital, Beijing, China.
| | - Xiaoming Zhang
- Department of Vascular Surgery, Peking University People's Hospital, Beijing, China.
| |
Collapse
|
9
|
Glucocorticoids: Fuelling the Fire of Atherosclerosis or Therapeutic Extinguishers? Int J Mol Sci 2021; 22:ijms22147622. [PMID: 34299240 PMCID: PMC8303333 DOI: 10.3390/ijms22147622] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/13/2021] [Accepted: 07/14/2021] [Indexed: 01/21/2023] Open
Abstract
Glucocorticoids are steroid hormones with key roles in the regulation of many physiological systems including energy homeostasis and immunity. However, chronic glucocorticoid excess, highlighted in Cushing's syndrome, is established as being associated with increased cardiovascular disease (CVD) risk. Atherosclerosis is the major cause of CVD, leading to complications including coronary artery disease, myocardial infarction and heart failure. While the associations between glucocorticoid excess and increased prevalence of these complications are well established, the mechanisms underlying the role of glucocorticoids in development of atheroma are unclear. This review aims to better understand the importance of glucocorticoids in atherosclerosis and to dissect their cell-specific effects on key processes (e.g., contractility, remodelling and lesion development). Clinical and pre-clinical studies have shown both athero-protective and pro-atherogenic responses to glucocorticoids, effects dependent upon their multifactorial actions. Evidence indicates regulation of glucocorticoid bioavailability at the vasculature is complex, with local delivery, pre-receptor metabolism, and receptor expression contributing to responses linked to vascular remodelling and inflammation. Further investigations are required to clarify the mechanisms through which endogenous, local glucocorticoid action and systemic glucocorticoid treatment promote/inhibit atherosclerosis. This will provide greater insights into the potential benefit of glucocorticoid targeted approaches in the treatment of cardiovascular disease.
Collapse
|
10
|
Wei Z, Chong H, Jiang Q, Tang Y, Xu J, Wang H, Shi Y, Cui L, Li J, Zhang Y, Xue Y, Li J, Liu G, Chen X, Wang D, Zhang CY, Jiang X. Smooth Muscle Overexpression of PGC1α Attenuates Atherosclerosis in Rabbits. Circ Res 2021; 129:e72-e86. [PMID: 34162227 DOI: 10.1161/circresaha.120.317705] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Zhe Wei
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Chinese Academy of Medical Sciences Research Unit of Extracellular RNA, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), Institute of Artificial Intelligence Biomedicine, School of Life Sciences, Nanjing University, Jiangsu, China (Z.W., Y.T., J.X., H.W., Y.S., L.C., Jing Li, Y.Z., X.C., D.W., C.-Y.Z., X.J.)
| | - Hoshun Chong
- Thoracic and Cardiovascular Surgery, Nanjing Drum Tower Hospital, China (H.C., Y.X.)
| | - Qixia Jiang
- Cardiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, China (Q.J., Jutang Li)
| | - Yuhang Tang
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Chinese Academy of Medical Sciences Research Unit of Extracellular RNA, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), Institute of Artificial Intelligence Biomedicine, School of Life Sciences, Nanjing University, Jiangsu, China (Z.W., Y.T., J.X., H.W., Y.S., L.C., Jing Li, Y.Z., X.C., D.W., C.-Y.Z., X.J.)
| | - Jinhong Xu
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Chinese Academy of Medical Sciences Research Unit of Extracellular RNA, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), Institute of Artificial Intelligence Biomedicine, School of Life Sciences, Nanjing University, Jiangsu, China (Z.W., Y.T., J.X., H.W., Y.S., L.C., Jing Li, Y.Z., X.C., D.W., C.-Y.Z., X.J.)
| | - Haoquan Wang
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Chinese Academy of Medical Sciences Research Unit of Extracellular RNA, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), Institute of Artificial Intelligence Biomedicine, School of Life Sciences, Nanjing University, Jiangsu, China (Z.W., Y.T., J.X., H.W., Y.S., L.C., Jing Li, Y.Z., X.C., D.W., C.-Y.Z., X.J.)
| | - Yanteng Shi
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Chinese Academy of Medical Sciences Research Unit of Extracellular RNA, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), Institute of Artificial Intelligence Biomedicine, School of Life Sciences, Nanjing University, Jiangsu, China (Z.W., Y.T., J.X., H.W., Y.S., L.C., Jing Li, Y.Z., X.C., D.W., C.-Y.Z., X.J.)
| | - Le Cui
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Chinese Academy of Medical Sciences Research Unit of Extracellular RNA, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), Institute of Artificial Intelligence Biomedicine, School of Life Sciences, Nanjing University, Jiangsu, China (Z.W., Y.T., J.X., H.W., Y.S., L.C., Jing Li, Y.Z., X.C., D.W., C.-Y.Z., X.J.)
| | - Jing Li
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Chinese Academy of Medical Sciences Research Unit of Extracellular RNA, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), Institute of Artificial Intelligence Biomedicine, School of Life Sciences, Nanjing University, Jiangsu, China (Z.W., Y.T., J.X., H.W., Y.S., L.C., Jing Li, Y.Z., X.C., D.W., C.-Y.Z., X.J.)
| | - Yujing Zhang
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Chinese Academy of Medical Sciences Research Unit of Extracellular RNA, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), Institute of Artificial Intelligence Biomedicine, School of Life Sciences, Nanjing University, Jiangsu, China (Z.W., Y.T., J.X., H.W., Y.S., L.C., Jing Li, Y.Z., X.C., D.W., C.-Y.Z., X.J.)
| | - Yunxing Xue
- Thoracic and Cardiovascular Surgery, Nanjing Drum Tower Hospital, China (H.C., Y.X.)
| | - Jutang Li
- Cardiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, China (Q.J., Jutang Li)
| | - George Liu
- Institute of Cardiovascular Science, Peking University, Beijing, China (G.L.)
| | - Xi Chen
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Chinese Academy of Medical Sciences Research Unit of Extracellular RNA, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), Institute of Artificial Intelligence Biomedicine, School of Life Sciences, Nanjing University, Jiangsu, China (Z.W., Y.T., J.X., H.W., Y.S., L.C., Jing Li, Y.Z., X.C., D.W., C.-Y.Z., X.J.)
| | - Dongjin Wang
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Chinese Academy of Medical Sciences Research Unit of Extracellular RNA, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), Institute of Artificial Intelligence Biomedicine, School of Life Sciences, Nanjing University, Jiangsu, China (Z.W., Y.T., J.X., H.W., Y.S., L.C., Jing Li, Y.Z., X.C., D.W., C.-Y.Z., X.J.)
| | - Chen-Yu Zhang
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Chinese Academy of Medical Sciences Research Unit of Extracellular RNA, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), Institute of Artificial Intelligence Biomedicine, School of Life Sciences, Nanjing University, Jiangsu, China (Z.W., Y.T., J.X., H.W., Y.S., L.C., Jing Li, Y.Z., X.C., D.W., C.-Y.Z., X.J.)
| | - Xiaohong Jiang
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, Chinese Academy of Medical Sciences Research Unit of Extracellular RNA, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), Institute of Artificial Intelligence Biomedicine, School of Life Sciences, Nanjing University, Jiangsu, China (Z.W., Y.T., J.X., H.W., Y.S., L.C., Jing Li, Y.Z., X.C., D.W., C.-Y.Z., X.J.)
| |
Collapse
|
11
|
Huang X, Liu Z, Shen L, Jin Y, Xu G, Zhang Z, Fang C, Guan W, Liu C. Augmentation of miR-202 in varicose veins modulates phenotypic transition of vascular smooth muscle cells by targeting proliferator-activated receptor-γ coactivator-1α. J Cell Biochem 2018; 120:10031-10042. [PMID: 30556158 DOI: 10.1002/jcb.28287] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 11/19/2018] [Indexed: 02/04/2023]
Abstract
In varicose veins, vascular smooth muscle cells (VSMCs) often show abnormal proliferative and migratory rates and phenotypic transition. This study aimed to investigate whether microRNA (miR)-202 and its potential target, peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α), were involved in VSMC phenotypic transition. miR-202 expression was analyzed in varicose veins and in VSMCs conditioned with platelet-derived growth factor. The effect of miR-202 on cell proliferation and migration was assessed. Furthermore, contractile marker SM-22α, synthetic markers vimentin and collagen I, and PGC-1α were analyzed by Western blot analysis. The modulation of PGC-1α expression by miR-202 was also evaluated. In varicose veins and proliferative VSMCs, miR-202 expression was upregulated, with decreased SM-22α expression and increased vimentin and collagen I expression. Transfection with a miR-202 mimic induced VSMC proliferation and migration, whereas a miR-202 inhibitor reduced cell proliferation and migration. miR-202 mimic constrained luciferase activity in HEK293 cells that were cotransfected with the PGC-1α 3'-untranslated region (3'-UTR) but not those with mutated 3'-UTR. miR-202 suppressed PGC-1α protein expression, with no influence on its messenger RNA expression. PGC-1α mediated VSMC phenotypic transition and was correlated with reactive oxygen species production. In conclusion, miR-202 affects VSMC phenotypic transition by targeting PGC-1α expression, providing a novel target for varicose vein therapy.
Collapse
Affiliation(s)
- Xianchen Huang
- Department of Gastrointestinal Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Vascular Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| | - Zhao Liu
- Department of Vascular Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Liming Shen
- Department of Vascular Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| | - Yiqi Jin
- Department of Vascular Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| | - Guoxiong Xu
- Department of Vascular Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| | - Zhixuan Zhang
- Department of Vascular Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| | - Changwen Fang
- Department of Vascular Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| | - Wenxian Guan
- Department of Gastrointestinal Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Changjian Liu
- Department of Vascular Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
12
|
Gao W, Du X, Lei L, Wang H, Zhang M, Wang Z, Li X, Liu G, Li X. NEFA-induced ROS impaired insulin signalling through the JNK and p38MAPK pathways in non-alcoholic steatohepatitis. J Cell Mol Med 2018; 22:3408-3422. [PMID: 29602237 PMCID: PMC6010831 DOI: 10.1111/jcmm.13617] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 02/21/2018] [Indexed: 12/11/2022] Open
Abstract
The aim of this study was to investigate the changes in hepatic oxidative phosphorylation (OXPHOS) complexes (COs) in patients and cows with non‐alcoholic steatohepatitis (NASH) and to investigate the mechanism that links mitochondrial dysfunction and hepatic insulin resistance induced by non‐esterified fatty acids (NEFAs). Patients and cows with NASH displayed high blood NEFAs, TNF‐α and IL‐6 concentrations, mitochondrial dysfunction and insulin resistance. The protein levels of peroxisome proliferator‐activated receptor‐γ coactivator‐1α (PGC‐1α), mitofusin‐2 (Mfn‐2) and OXPHOS complexes (human: COI and COIII; cow: COI‐IV) were significantly decreased in patients and cows with NASH. NEFA treatment significantly impaired mitochondrial function and, increased reactive oxygen species (ROS) production, and excessive ROS overactivated the JNK and p38MAPK pathways and induced insulin resistance in cow hepatocytes. PGC‐1α and Mfn‐2 overexpression significantly decreased the NEFA‐induced ROS production and TNF‐α and IL‐6 mRNA expressions, reversed the inhibitory effect of NEFAs on mitochondrial function and attenuated the overactivation of the ROS‐JNK/p38MAPK pathway, alleviated insulin resistance induced by NEFAs in cow hepatocytes and HepG2 cells. These findings indicate that NEFAs induce mitochondrial dysfunction and insulin resistance mediated by the ROS‐JNK/p38MAPK pathway. PGC‐1α or Mfn‐2 overexpression reversed the lipotoxicity of NEFAs on mitochondrial dysfunction and insulin resistance. Our study clarified the mechanism that links hepatic mitochondrial dysfunction and insulin resistance in NASH.
Collapse
Affiliation(s)
- Wenwen Gao
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, China
| | - Xiliang Du
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, China
| | - Lin Lei
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, China
| | - Heyuan Wang
- Department of Endocrinology and Metabolism, The first Hospital, Jilin University, Changchun, Jilin Province, China
| | - Min Zhang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, China
| | - Zhe Wang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, China
| | - Xiaobing Li
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, China
| | - Guowen Liu
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, China
| | - Xinwei Li
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, China
| |
Collapse
|
13
|
Li W, Zhi W, Liu F, He Z, Wang X, Niu X. Atractylenolide I restores HO-1 expression and inhibits Ox-LDL-induced VSMCs proliferation, migration and inflammatory responses in vitro. Exp Cell Res 2017; 353:26-34. [PMID: 28274716 DOI: 10.1016/j.yexcr.2017.02.040] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 02/21/2017] [Accepted: 02/27/2017] [Indexed: 12/25/2022]
Abstract
Pathogenesis of atherosclerosis is characterized by the proliferation and migration of vascular smooth muscle cells (VSMCs) and inflammatory lesions. The aim of this study is to elucidate the effect of atractylenolide I (AO-I) on smooth muscle cell inflammation, proliferation and migration induced by oxidized modified low density lipoprotein (Ox-LDL). Here, We found that atractylenolide I inhibited Ox-LDL-induced VSMCs proliferation and migration in a dose-dependent manner, and decreased the production of inflammatory cytokines and the expression of monocyte chemoattractant protein-1 (MCP-1) in VSMCs. The study also identified that AO-I prominently inhibited p38-MAPK and NF-κB activation. More importantly, the specific heme oxygenase-1 (HO-1) inhibitor zinc protoporphyrin (ZnPP) IX partially abolished the beneficial effects of atractylenolide I on Ox-LDL-induced VSMCs. Furthermore, atractylenolide I blocked the foam cell formation in macrophages induced by Ox-LDL. In summary, inhibitory roles of AO-I in VSMCs proliferation and migration, lipid peroxidation and subsequent inflammatory responses might contribute to the anti-atherosclerotic property of AO-I.
Collapse
Affiliation(s)
- Weifeng Li
- School of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, PR China.
| | - Wenbing Zhi
- School of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, PR China
| | - Fang Liu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, PR China
| | - Zehong He
- School of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, PR China
| | - Xiuei Wang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, PR China
| | - Xiaofeng Niu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, PR China.
| |
Collapse
|
14
|
Fang Z, Li P, Jia W, Jiang T, Wang Z, Xiang Y. miR-696 plays a role in hepatic gluconeogenesis in ob/ob mice by targeting PGC-1α. Int J Mol Med 2016; 38:845-52. [DOI: 10.3892/ijmm.2016.2659] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 06/14/2016] [Indexed: 11/06/2022] Open
|
15
|
Zhou JJ, Ma JD, Mo YQ, Zheng DH, Chen LF, Wei XN, Dai L. Down-regulating peroxisome proliferator-activated receptor-gamma coactivator-1 beta alleviates the proinflammatory effect of rheumatoid arthritis fibroblast-like synoviocytes through inhibiting extracellular signal-regulated kinase, p38 and nuclear factor-kappaB activation. Arthritis Res Ther 2014; 16:472. [PMID: 25367151 PMCID: PMC4237730 DOI: 10.1186/s13075-014-0472-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 10/16/2014] [Indexed: 11/28/2022] Open
Abstract
Introduction Rheumatoid arthritis (RA) is a chronic inflammatory disease leading to joint destruction and disability. Peroxisome proliferator-activated receptor-gamma coactivator-1beta (PGC-1β) is a transcriptional coactivator that plays important roles in regulating multiple aspects of energy metabolism and cytokine signaling pathways. PGC-1β overexpression leads to the attenuation of macrophage-mediated inflammation. In this study, we aimed to determine the expression of PGC-1β in RA synovium and fibroblast-like synoviocytes (FLS), and explore the mechanisms of PGC-1β on both the proinflammatory effects and apoptosis in RA-FLS. Methods Synovium was obtained from 31 patients with active RA, as well as 13 osteoarthritis (OA) and 10 orthopedic arthropathies (Orth.A) as “less inflamed” disease controls. FLS were then isolated and cultured. Synovial PGC-1β expression was determined by immunohistochemistry staining, while FLS PGC-1β expression was detected by immunofluorescence staining, quantitative real-time PCR (qPCR) assay and western blot. PGC-1β was depleted by lentivirus sh-RNA, and up-regulated by pcDNA3.1- PGC-1β. The expression of proinflammatory cytokines, matrix metalloproteinases and receptor activator of nuclear factor-kappaB ligand was analyzed by qPCR, cytometric bead array and western blot. The expression of mitogen-activated protein kinases and nuclear factor-kappaB (NF-κB) was determined by qPCR and western blot. Besides, cell apoptosis was examined using flow cytometry. The interaction between PGC-1β and NF-κB was performed by dual-luciferase reporter gene assays. Results (A) Synovial PGC-1β was over-expressed in RA patients compared with OA or Orth.A patients. (B) PGC-1β expression significantly increased in RA-FLS compared with OA-FLS. (C) PGC-1β mediated the expression of proinflammatory cytokines and apoptosis through extracellular signal-regulated kinase (ERK), p38 and NF-κB in RA-FLS. (D) PGC-1β mediated NF-κB transcription in RA-FLS, but did not affect ERK and p38. Conclusion The results indicate that PGC-1β may play important roles in the proinflammatory effects and apoptosis of RA-FLS.
Collapse
|
16
|
Foletta VC, Brown EL, Cho Y, Snow RJ, Kralli A, Russell AP. Ndrg2 is a PGC-1α/ERRα target gene that controls protein synthesis and expression of contractile-type genes in C2C12 myotubes. BIOCHIMICA ET BIOPHYSICA ACTA 2013; 1833:3112-3123. [PMID: 24008097 DOI: 10.1016/j.bbamcr.2013.08.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Revised: 06/17/2013] [Accepted: 08/09/2013] [Indexed: 01/02/2023]
Abstract
The stress-responsive, tumor suppressor N-myc downstream-regulated gene 2 (Ndrg2) is highly expressed in striated muscle. In response to anabolic and catabolic signals, Ndrg2 is suppressed and induced, respectively, in mouse C2C12 myotubes. However, little is known about the mechanisms regulating Ndrg2 expression in muscle, as well as the biological role for Ndrg2 in differentiated myotubes. Here, we show that Ndrg2 is a target of a peroxisome proliferator-activated receptor-gamma coactivator-1α (PGC-1α) and estrogen-related receptor alpha (ERRα) transcriptional program and is induced in response to endurance exercise, a physiological stress known also to increase PGC-1α/ERRα activity. Analyses of global gene and protein expression profiles in C2C12 myotubes with reduced levels of NDRG2, suggest that NDRG2 affects muscle growth, contractile properties, MAPK signaling, ion and vesicle transport and oxidative phosphorylation. Indeed, suppression of NDRG2 in myotubes increased protein synthesis and the expression of fast glycolytic myosin heavy chain isoforms, while reducing the expression of embryonic myosin Myh3, other contractile-associated genes and the MAPK p90 RSK1. Conversely, enhanced expression of NDRG2 reduced protein synthesis, and furthermore, partially blocked the increased protein synthesis rates elicited by a constitutively active form of ERRα. In contrast, suppressing or increasing levels of NDRG2 did not affect mRNA expression of genes involved in mitochondrial biogenesis that are regulated by PGC-1α or ERRα. This study shows that in C2C12 myotubes Ndrg2 is a novel PGC-1α/ERRα transcriptional target, which influences protein turnover and the regulation of genes involved in muscle contraction and function.
Collapse
Affiliation(s)
- Victoria C Foletta
- Centre for Physical Activity and Nutrition Research, School of Exercise and Nutrition Sciences, Deakin University, Burwood 3125, Australia.
| | - Erin L Brown
- Centre for Physical Activity and Nutrition Research, School of Exercise and Nutrition Sciences, Deakin University, Burwood 3125, Australia
| | - Yoshitake Cho
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Rod J Snow
- Centre for Physical Activity and Nutrition Research, School of Exercise and Nutrition Sciences, Deakin University, Burwood 3125, Australia
| | - Anastasia Kralli
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Aaron P Russell
- Centre for Physical Activity and Nutrition Research, School of Exercise and Nutrition Sciences, Deakin University, Burwood 3125, Australia
| |
Collapse
|
17
|
McCarthy C, Lieggi NT, Barry D, Mooney D, de Gaetano M, James WG, McClelland S, Barry MC, Escoubet-Lozach L, Li AC, Glass CK, Fitzgerald DJ, Belton O. Macrophage PPAR gamma Co-activator-1 alpha participates in repressing foam cell formation and atherosclerosis in response to conjugated linoleic acid. EMBO Mol Med 2013; 5:1443-57. [PMID: 23964012 PMCID: PMC3799497 DOI: 10.1002/emmm.201302587] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Revised: 06/27/2013] [Accepted: 07/02/2013] [Indexed: 12/31/2022] Open
Abstract
Conjugated linoleic acid (CLA) has the unique property of inducing regression of pre-established murine atherosclerosis. Understanding the mechanism(s) involved may help identify endogenous pathways that reverse human atherosclerosis. Here, we provide evidence that CLA inhibits foam cell formation via regulation of the nuclear receptor coactivator, peroxisome proliferator-activated receptor (PPAR)-γ coactivator (PGC)-1α, and that macrophage PGC-1α plays a role in atheroprotection in vivo. PGC-1α was identified as a hub gene within a cluster in the aorta of the apoE−/− mouse in the CLA-induced regression model. PGC-1α was localized to macrophage/foam cells in the murine aorta where its expression was increased during CLA-induced regression. PGC-1α expression was also detected in macrophages in human atherosclerosis and was inversely linked to disease progression in patients with the disease. Deletion of PGC-1α in bone marrow derived macrophages promoted, whilst over expression of the gene inhibited foam cell formation. Importantly, macrophage specific deletion of PGC-1α accelerated atherosclerosis in the LDLR−/− mouse in vivo. These novel data support a functional role for PGC-1α in atheroprotection.
Collapse
Affiliation(s)
- Cathal McCarthy
- School of Biomolecular and Biomedical Science, UCD Conway Institute, UCD, Dublin, Ireland
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Li YH, Wang QX, Zhou JW, Chu XM, Man YL, Liu P, Ren BB, Sun TR, An Y. Effects of rosuvastatin on expression of angiotensin-converting enzyme 2 after vascular balloon injury in rats. JOURNAL OF GERIATRIC CARDIOLOGY : JGC 2013; 10:151-8. [PMID: 23888175 PMCID: PMC3708055 DOI: 10.3969/j.issn.1671-5411.2013.02.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2012] [Revised: 02/19/2013] [Accepted: 04/22/2013] [Indexed: 11/18/2022]
Abstract
Objective To investigate the effects and mechanisms of rosuvastatin on angiotensin -converting enzyme 2 (ACE2) in the process of neointimal formation after vascular balloon injury in rats, and to explore the effects of ACE2 and rosuvastatin in restenosis. Methods Thirty-six Wistar rats were randomly allocated into three groups: control group (n = 12), surgery group (n = 12), and statin group (n = 12). Aortic endothelial denudation of rats was performed using 2F balloon catheters. At days 14 and 28 after injury, aortic arteries were harvested to examine the following. Intimal thickening was examined by hematoxylin and eosin staining. We measured angiotensin II (Ang II) and angiotensin 1-7 (Ang-[1–7]) levels by a radioimmunological method or enzyme-linked immunosorbent assay. Protein and mRNA expression of ACE2 and Ang II type 1 receptor (AT1) were investigated by immunohistochemistry, Western blots, and Reverse transcriptase-polymerase chain reaction (RT-PCR). We measured changes in proliferating cell nuclear antigen (PCNA) by immunohistochemistry. The level of phosphorylated extracellular signal regulated kinase 1/2 (P-ERK1/2) was evaluated by Western blotting. Results Proliferation of vascular smooth muscle cells (VSMC) and intimal thickening were higher at day 14 after vascular balloon injury in the surgery group compared with the control group. Proliferation of VSMC was decreased by day 28 after injury, while intimal thickening continued. With rosuvastatin treatment, the extent of VSMC proliferation and intimal thickening was reduced at day 14 and 28 after injury. Ang II and P-ERK levels were significantly increased, Ang-(1–7) levels were significantly decreased, mRNA and protein expressions of ACE2 were significantly decreased, and AT1 expression was significantly increased at days 14 and 28 after vascular balloon injury in the surgery group compared with the control group. PCNA expression was higher in the surgery group than in the control group, and it was significantly decreased after being given rosuvastatin. Expression of ACE2 mRNA and protein, and Ang-(1–7) levels were significantly increased, while AT1 expression and levels of Ang II and P-ERK were significantly decreased in the statin group compared with the surgery group. Conclusions Expression of ACE2 mRNA and protein is decreased in the process of intimal thickening after balloon injury. The inhibitory effect of rosuvastatin on intimal thickening is related to upregulation of ACE2, an increase in Ang-(1–7), downregulation of AT1, and activation of the P-ERK pathway.
Collapse
Affiliation(s)
- Yong-Hong Li
- Department of Cardiology, Affiliated Hospital of Medical College, Qingdao University, Qingdao 266003, China
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Xu W, Hou D, Jiang X, Lu Z, Guo T, Liu Y, Wang D, Zen K, Yu B, Zhang CY. The protective role of peroxisome proliferator-activated receptor γ coactivator-1α in hyperthyroid cardiac hypertrophy. J Cell Physiol 2012; 227:3243-53. [DOI: 10.1002/jcp.24015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
20
|
Patten IS, Arany Z. PGC-1 coactivators in the cardiovascular system. Trends Endocrinol Metab 2012; 23:90-7. [PMID: 22047951 DOI: 10.1016/j.tem.2011.09.007] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Revised: 09/25/2011] [Accepted: 09/29/2011] [Indexed: 01/16/2023]
Abstract
The beating heart consumes more ATP per weight than any other organ. The machineries required for this are many and complex. Fuel and oxygen must be transported via the vasculature, absorbed by cardiomyocytes, broken down, and regulated to match cellular demands. Much of this occurs in mitochondria, which comprise fully one third of cardiac mass. The PGC-1 proteins are transcriptional coactivators that have emerged as powerful orchestrators of these numerous processes, ensuring their proper coregulation in response to intracellular and extracellular cues. An important role for PGC-1s in cardiac function has been revealed over the past few years, and more recently interest in their role in the vasculature has been burgeoning. We review this literature, focusing on recent developments.
Collapse
Affiliation(s)
- Ian S Patten
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | | |
Collapse
|