1
|
Kong L, Li J, Bai Y, Xu S, Zhang L, Chen W, Gao L, Wang F. Inhibition of soluble epoxide hydrolase enhances the dentin-pulp complex regeneration mediated by crosstalk between vascular endothelial cells and dental pulp stem cells. J Transl Med 2024; 22:61. [PMID: 38229161 PMCID: PMC10790489 DOI: 10.1186/s12967-024-04863-y] [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: 07/20/2023] [Accepted: 01/06/2024] [Indexed: 01/18/2024] Open
Abstract
BACKGROUND Revascularization and restoration of normal pulp-dentin complex are important for tissue-engineered pulp regeneration. Recently, a unique periodontal tip-like endothelial cells subtype (POTCs) specialized to dentinogenesis was identified. We have confirmed that TPPU, a soluble epoxide hydrolase (sEH) inhibitor targeting epoxyeicosatrienoic acids (EETs) metabolism, promotes bone growth and regeneration by angiogenesis and osteogenesis coupling. We hypothesized that TPPU could also promote revascularization and induce POTCs to contribute to pulp-dentin complex regeneration. Here, we in vitro and in vivo characterized the potential effect of TPPU on the coupling of angiogenesis and odontogenesis and investigated the relevant mechanism, providing new ideas for pulp-dentin regeneration by targeting sEH. METHODS In vitro effects of TPPU on the proliferation, migration, and angiogenesis of dental pulp stem cells (DPSCs), human umbilical vein endothelial cells (HUVECs) and cocultured DPSCs and HUVECs were detected using cell counting kit 8 (CCK8) assay, wound healing, transwell, tube formation and RT-qPCR. In vivo, Matrigel plug assay was performed to outline the roles of TPPU in revascularization and survival of grafts. Then we characterized the VEGFR2 + POTCs around odontoblast layer in the molar of pups from C57BL/6 female mice gavaged with TPPU. Finally, the root segments with DPSCs mixed with Matrigel were implanted subcutaneously in BALB/c nude mice treated with TPPU and the root grafts were isolated for histological staining. RESULTS In vitro, TPPU significantly promoted the migration and tube formation capability of cocultured DPSCs and HUVECs. ALP and ARS staining and RT-qPCR showed that TPPU promoted the osteogenic and odontogenic differentiation of cultured cells, treatment with an anti-TGF-β blocking antibody abrogated this effect. Knockdown of HIF-1α in HUVECs significantly reversed the effect of TPPU on the expression of angiogenesis, osteogenesis and odontogenesis-related genes in cocultured cells. Matrigel plug assay showed that TPPU increased VEGF/VEGFR2-expressed cells in transplanted grafts. TPPU contributed to angiogenic-odontogenic coupling featured by increased VEGFR2 + POTCs and odontoblast maturation during early dentinogenesis in molar of newborn pups from C57BL/6 female mice gavaged with TPPU. TPPU induced more dental pulp-like tissue with more vessels and collagen fibers in transplanted root segment. CONCLUSIONS TPPU promotes revascularization of dental pulp regeneration by enhancing migration and angiogenesis of HUVECs, and improves odontogenic differentiation of DPSCs by TGF-β. TPPU boosts the angiogenic-odontogenic coupling by enhancing VEGFR2 + POTCs meditated odontoblast maturation partly via upregulating HIF-1α, which contributes to increasing pulp-dentin complex for tissue-engineered pulp regeneration.
Collapse
Affiliation(s)
- Lingwenyao Kong
- School of Stomatology, Dalian Medical University, No. 9 West Section, Lvshun South Road, Dalian, 116044, People's Republic of China
- Academician Laboratory of Immune and Oral Development & Regeneration, Dalian Medical University, Dalian, China
| | - Juanjuan Li
- School of Stomatology, Dalian Medical University, No. 9 West Section, Lvshun South Road, Dalian, 116044, People's Republic of China
- Academician Laboratory of Immune and Oral Development & Regeneration, Dalian Medical University, Dalian, China
| | - Yuwen Bai
- School of Stomatology, Dalian Medical University, No. 9 West Section, Lvshun South Road, Dalian, 116044, People's Republic of China
- Academician Laboratory of Immune and Oral Development & Regeneration, Dalian Medical University, Dalian, China
| | - Shaoyang Xu
- School of Stomatology, Dalian Medical University, No. 9 West Section, Lvshun South Road, Dalian, 116044, People's Republic of China
| | - Lin Zhang
- School of Stomatology, Dalian Medical University, No. 9 West Section, Lvshun South Road, Dalian, 116044, People's Republic of China
| | - Weixian Chen
- School of Stomatology, Dalian Medical University, No. 9 West Section, Lvshun South Road, Dalian, 116044, People's Republic of China
- Academician Laboratory of Immune and Oral Development & Regeneration, Dalian Medical University, Dalian, China
| | - Lu Gao
- School of Stomatology, Dalian Medical University, No. 9 West Section, Lvshun South Road, Dalian, 116044, People's Republic of China.
- Academician Laboratory of Immune and Oral Development & Regeneration, Dalian Medical University, Dalian, China.
- The Affiliated Stomatological Hospital of Dalian Medical University, Dalian, China.
| | - Fu Wang
- School of Stomatology, Dalian Medical University, No. 9 West Section, Lvshun South Road, Dalian, 116044, People's Republic of China.
- Academician Laboratory of Immune and Oral Development & Regeneration, Dalian Medical University, Dalian, China.
- The Affiliated Stomatological Hospital of Dalian Medical University, Dalian, China.
| |
Collapse
|
2
|
Yang C, Wang Y, Wu X, Gong M, Li Y. Reduced levels of A20 protein prompted RIPK1-dependent apoptosis and blood-brain barrier breakdown during cerebral ischemia reperfusion injury. PLoS One 2023; 18:e0290015. [PMID: 37578944 PMCID: PMC10424866 DOI: 10.1371/journal.pone.0290015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 08/01/2023] [Indexed: 08/16/2023] Open
Abstract
Blood-brain barrier (BBB) leakage is an important cause of the exacerbation of pathological features of cerebral ischemia reperfusion injury (CIRI). However, the specific mechanism of BBB leakage is not clear. It was found that the CIRI resulted in RIPK1 activation and subsequent RIPK1-dependent apoptosis (RDA). Inhibition of RIPK1 significantly reduced BBB breakdown and brain damage. The aim of this study is to investigate the mechanism of RIPK1 in the BBB leakage during CIRI. It was discovered by proteomics that autophagy activation resulting from ischemia and reperfusion significantly downregulated the level of A20 protein. A20 is an important protein that regulates RIPK1 and RDA. It was hypothesized that activation of autophagy caused by ischemic reperfusion led to a decrease in A20 protein, which, in turn, caused the activation of RIPK1 and the occurrence of RDA, leading to leakage of the BBB. The findings in this study revealed the role of RIPK1 in the cell death and BBB leakage upon cerebral ischemia reperfusion injury, and these findings provide a novel perspective for the treatment of ischemic reperfusion.
Collapse
Affiliation(s)
- Chaonan Yang
- Department of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Yongjiao Wang
- Department of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Xiaohui Wu
- Department of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Min Gong
- Department of Pharmacy, Tianjin Medical University, Tianjin, China
- WIMM, University of Oxford, Oxford, United Kingdom
| | - Ying Li
- Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
3
|
Davis CM, Ibrahim AH, Alkayed NJ. Cytochrome P450-derived eicosanoids in brain: From basic discovery to clinical translation. ADVANCES IN PHARMACOLOGY 2023; 97:283-326. [DOI: 10.1016/bs.apha.2022.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
4
|
Chen X, Li Z, Zhang B, Liu T, Yao W, Wan L, Zhang C, Zhang Y. Antinociception role of 14,15-epoxyeicosatrienoic acid in a central post-stroke pain model in rats mediated by anti-inflammation and anti-apoptosis effect. Neurochem Int 2022; 154:105291. [PMID: 35074479 DOI: 10.1016/j.neuint.2022.105291] [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: 05/25/2021] [Revised: 11/24/2021] [Accepted: 01/20/2022] [Indexed: 11/18/2022]
Abstract
Central post stroke pain (CPSP) is an intractable neuropathic pain syndrome that occurs after the acute focal lesion of the central nervous system (CNS) due to a cerebrovascular cause. Epoxyeicosatrienoic acids (EETs) exert many pharmacological effects in vivo and in vitro, such as anti-apoptosis, anti-inflammatory, and anti-oxidative stress. Neuroinflammation and apoptosis are the potential pathophysiological mechanisms of neuropathic pain. This study aimed to investigate whether 14,15-EET has an antinociception effect on CPSP rats through its anti-inflammation and anti-apoptosis mechanisms. Rats were treated with type IV collagenase (CPSP group) or saline (Sham group) via injection with a Hamilton syringe into the ventral posterior lateral nucleus (VPL) according to the stereotaxic coordinates. We first tested the mechanical withdrawal threshold, as well as neuroinflammation- and apoptosis-related protein expressions in the per-lesion site of CPSP and Sham rats. Sprague-Dawley rats were randomly divided into five groups, as follows: vehicle; EET at 0.025, 0.05, and 0.1 μg; and EET (0.1 μg) + EEZE (3.25 ng). EET or and vehicle were administered into VPL nuclei three consecutive days after hemorrhagic stroke. Immunostaining, ELISA, and Western blot were performed to evaluate neuroinflammation and apoptosis. Hemorrhagic stroke induced mechanical allodynia, glial activation, neuroinflammation, and apoptosis-related protein upregulation. However, early treatment with 14,15-EET inhibited glial cell activation, decreased proinflammatory cytokines and apoptosis-related protein, and alleviated the pain behavior of CPSP rats. Our results provided strong evidence that antinociception produced by 14,15-EET is partly mediated by the inhibition of neuroinflammation and apoptosis.
Collapse
Affiliation(s)
- Xuhui Chen
- Department of Anaesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zuofan Li
- Department of Anaesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Bo Zhang
- Department of Anaesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Tongtong Liu
- Department of Anaesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wenlong Yao
- Department of Anaesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Li Wan
- Department of Anaesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Chuanhan Zhang
- Department of Anaesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yue Zhang
- Department of Anaesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
5
|
Xu Y, Liu Y, Li K, Yuan D, Yang S, Zhou L, Zhao Y, Miao S, Lv C, Zhao J. COX-2/PGE2 Pathway Inhibits the Ferroptosis Induced by Cerebral Ischemia Reperfusion. Mol Neurobiol 2022; 59:1619-1631. [PMID: 35013936 DOI: 10.1007/s12035-021-02706-1] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 12/15/2021] [Indexed: 12/21/2022]
Abstract
Cerebral ischemia reperfusion (I/R) injury easily develops in ischemic stroke, resulting in more serious injury. Ferroptosis is involved in cerebral I/R injury, but the mechanism remains unclear. Prostaglandin E2 (PGE2) is potential to regulate ferroptosis. This study mainly explored the regulation effects of PGE2 on ferroptosis induced by cerebral I/R. We first detected PGE2 levels and ferroptosis status in 11 human brain tissues. Then, we induced a cerebral I/R animal model to examine ferroptosis status in cerebral I/R. We further injected a ferroptosis inhibitor to define the response of the PGE2 pathway to ferroptosis. Finally, we injected PGE2 and pranoprofen to explore the regulation of the cyclooxygenases 2 (COX-2)/PGE2 pathway on ferroptosis in cerebral I/R. We found that PGE2 release was correlated with the levels of reactive oxygen species, malondialdehyde, glutathione peroxidase 4, COX-2, and Spermidine/spermine N1-acetyltransferase 1. Ferroptosis can be induced by cerebral I/R, while inhibition of ferroptosis induced by cerebral I/R can inactivate PGE2 synthases, degrade enzyme, and parts of PGE2 receptors, and reduce cerebral infarct volume. In turn, PGE2 inhibited ferroptosis through the reduction of Fe2+, glutathione oxidation, and lipid peroxidation, while pranoprofen, one of the COX inhibitors, played an opposite role. In conclusion, PGE2 was positively correlated with ferroptosis, inhibition of ferroptosis induced by cerebral I/R can inactivate COX-2/PGE2 pathway, and PGE2 inhibited ferroptosis induced by cerebral I/R, possibly via PGE2 receptor 3 and PGE2 receptor 4. Graphical abstract Inhibition of ferroptosis inactivates the COX-2/PGE2 pathway. Cerebral ischemia reperfusion injury induces the secretion of PGE2. After the inhibition of ferroptosis by Fer-1, the expression of cyclooxygenases (COX-1 and COX-2) decreased, and PGE2 synthases cPGES, mPGES-1, and mPGES-2 were also reduced. At the same time, the PGE2 degradation enzyme 15-PGDH was also reduced. Changes in these enzymes ultimately result in the declination of PGE2. Besides, the expression of PGE2 receptors EP3 and EP4 is also inhibited, indicating that the function they mediate is also impaired. In conclusion, after cerebral ischemia reperfusion injury, the inhibition of ferroptosis inactivates the COX-2/PGE2 pathway.
Collapse
Affiliation(s)
- Yunfei Xu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.,Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, 410008, Hunan, China.,Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, 410008, Hunan, China.,China-Africa Research Center of Infectious Diseases, Central South University, Changsha, China
| | - Ying Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China. .,Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, 410008, Hunan, China. .,Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, 410008, Hunan, China. .,China-Africa Research Center of Infectious Diseases, Central South University, Changsha, China.
| | - Kexin Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.,Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, 410008, Hunan, China.,Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, 410008, Hunan, China.,China-Africa Research Center of Infectious Diseases, Central South University, Changsha, China
| | - Dun Yuan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Shun Yang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Lin Zhou
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.,Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, 410008, Hunan, China.,Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, 410008, Hunan, China.,China-Africa Research Center of Infectious Diseases, Central South University, Changsha, China
| | - Yao Zhao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.,Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, 410008, Hunan, China.,Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, 410008, Hunan, China.,China-Africa Research Center of Infectious Diseases, Central South University, Changsha, China
| | - Shuying Miao
- China-Africa Research Center of Infectious Diseases, Central South University, Changsha, China.,Department of Pathology, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, 210008, Jiangsu, China
| | - Caihong Lv
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.,Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, 410008, Hunan, China.,Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, 410008, Hunan, China.,China-Africa Research Center of Infectious Diseases, Central South University, Changsha, China
| | - Jie Zhao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China. .,Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, 410008, Hunan, China. .,China-Africa Research Center of Infectious Diseases, Central South University, Changsha, China.
| |
Collapse
|
6
|
Kuo YM, Lee YH. Epoxyeicosatrienoic acids and soluble epoxide hydrolase in physiology and diseases of the central nervous system. CHINESE J PHYSIOL 2022; 65:1-11. [PMID: 35229747 DOI: 10.4103/cjp.cjp_80_21] [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] [Indexed: 12/14/2022] Open
Abstract
Epoxyeicosatrienoic acids (EETs) are fatty acid signaling molecules synthesized by cytochrome P450 epoxygenases from arachidonic acid. The biological activity of EETs is terminated when being metabolized by soluble epoxide hydrolase (sEH), a process that serves as a key regulator of tissue EETs levels. EETs act through several signaling pathways to mediate various beneficial effects, including anti-inflammation, anti-apoptosis, and anti-oxidation with relieve of endoplasmic reticulum stress, thereby sEH has become a potential therapeutic target in cardiovascular disease and cancer therapy. Enzymes for EET biosynthesis and metabolism are both widely detected in both neuron and glial cells in the central nervous system (CNS). Recent studies discovered that astrocyte-derived EETs not only mediate neurovascular coupling and neuronal excitability by maintaining glutamate homeostasis but also glia-dependent neuroprotection. Genetic ablation as well as pharmacologic inhibition of sEH has greatly helped to elucidate the physiologic actions of EETs, and maintaining or elevating brain EETs level has been demonstrated beneficial effects in CNS disease models. Here, we review the literature regarding the studies on the bioactivity of EETs and their metabolic enzyme sEH with special attention paid to their action mechanisms in the CNS, including their modulation of neuronal activity, attenuation of neuroinflammation, regulation of cerebral blood flow, and improvement of neuronal and glial cells survival. We further reviewed the recent advance on the potential application of sEH inhibition for treating cerebrovascular disease, epilepsy, and pain disorder.
Collapse
Affiliation(s)
- Yi-Min Kuo
- Department of Anesthesiology, Taipei Veterans General Hospital; Department of Anesthesiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yi-Hsuan Lee
- Department and Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University; Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
7
|
Yu G, Sun W, Wang W, Le C, Liang D, Shuai L. Overexpression of microRNA-202-3p in bone marrow mesenchymal stem cells improves cerebral ischemia-reperfusion injury by promoting angiogenesis and inhibiting inflammation. Aging (Albany NY) 2021; 13:11877-11888. [PMID: 33893248 PMCID: PMC8109138 DOI: 10.18632/aging.202889] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 02/27/2021] [Indexed: 12/18/2022]
Abstract
BACKGROUND Cerebral ischemia-reperfusion injury (CIRI) can cause brain tissue inflammation, neuronal degeneration, and apoptosis. There is increasing evidence that microRNAs (miRNA) exert neuroprotective effects by regulating the inflammatory process during cerebral ischemia-reperfusion injury. Additionally, it is increasingly acknowledged that neuroinflammation is regulated by Toll-like receptor 4 (TLR4). However, it is unclear whether miRNA can exert its neuroprotective effects by regulating TLR4-mediated inflammation. METHODS The effects of BMSCs over-expressing miR-202-3p on CIRI, angiogenesis in midbrain tissue, and the release of inflammatory factors (IFs) in the serum were measured using in vivo rat models. We also used SH-SY5Y cells to establish an ischemia-reperfusion in vitro cell model. The interaction between miR-202-3p and TLR4 was analyzed by overexpressing miR-202-3p and knocking down TLR4. Knockdown of TLR4 was performed using siRNA. RESULTS Overexpression of miR-202-3p in BMSCs could significantly improve brain function and reduce brain damage. Simultaneously, miR-202-3p could significantly promote angiogenesis, increase the expression of vWF and VEGF, and reduce the expression of IFs. When the expression of TLR4 was significantly reduced in SH-SY5Y cells, the expression of IFs increased. Therefore, miRNA-202-3p may interact with TLR4 to modulate inflammation. CONCLUSION Our data indicated that miR-202-3p potentially exerts its neuroprotective effects and protects against CIRI by regulating TLR4-mediated inflammation.
Collapse
Affiliation(s)
- Guohua Yu
- Department of Rehabilitation, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Weiming Sun
- Department of Rehabilitation, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Wansong Wang
- Department of Rehabilitation, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Changhao Le
- Department of Rehabilitation, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Dehuan Liang
- Department of Rehabilitation, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Lang Shuai
- Department of Rehabilitation, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| |
Collapse
|
8
|
Mukherjee S, Kumar G, Patnaik R. Withanolide a penetrates brain via intra-nasal administration and exerts neuroprotection in cerebral ischemia reperfusion injury in mice. Xenobiotica 2020; 50:957-966. [PMID: 31870211 DOI: 10.1080/00498254.2019.1709228] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Sumedha Mukherjee
- Electrophysiology Lab, School of Biomedical Engineering, Indian Institute of Technology (BHU), Varanasi, India
| | - Gaurav Kumar
- Electrophysiology Lab, School of Biomedical Engineering, Indian Institute of Technology (BHU), Varanasi, India
- School of Biological and Biomedical Sciences, Galgotias University, Gr. Noida, India
| | - Ranjana Patnaik
- Electrophysiology Lab, School of Biomedical Engineering, Indian Institute of Technology (BHU), Varanasi, India
- School of Biological and Biomedical Sciences, Galgotias University, Gr. Noida, India
| |
Collapse
|
9
|
Excessive arachidonic acid induced actin bunching remodeling and podocyte injury via a PKA-c-Abl dependent pathway. Exp Cell Res 2019; 388:111808. [PMID: 31891685 DOI: 10.1016/j.yexcr.2019.111808] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 12/17/2019] [Accepted: 12/27/2019] [Indexed: 11/22/2022]
Abstract
Recent studies have shown that serum secretory phospholipase A2 group IB (sPLA2-IB) is associated with proteinuric kidney diseases and plays a pivotal role in podocyte injury via its natural receptor. Arachidonic acid (AA), as a major metabolite of sPLA2-IB, regulates the actin bungling remodeling and contributes to the podocyte injury. However, the underlying mechanism of AA in the regulation of podocyte actin remodeling and human podocyte injury is unclear. Here, we reported that AA induced F-actin cytoskeletal ring formation and promoted protein kinase A (PKA), nephrin and c-Abl phosphorylation. Moreover, AA promoted c-Abl translocation from the nucleus to the cytoplasm and increased the recruitment of c-Abl to p-nephrin by the interaction between them. H89 (PKA inhibitor) provided protection against AA-induced F-actin bunching remodeling, down-regulated nephrin phosphorylation, and suppressed the c-Abl translocation and activation. STI571 (c-Abl inhibitor) also improved the AA associated F-actin bunching remodeling. In addition, H89 and STI571 both alleviated apoptosis and adhesion damage of podocyte. These results indicate that an excess of AA treatment is detrimental to the podocyte actin cytoskeleton and promotes podocyte injury due to the activation of PKA-c-Abl signaling.
Collapse
|
10
|
PUFAs supplementation affects the renal expression of pannexin 1 and connexins in diabetic kidney of rats. Histochem Cell Biol 2019; 153:165-175. [PMID: 31858211 DOI: 10.1007/s00418-019-01838-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/14/2019] [Indexed: 12/26/2022]
Abstract
In diabetic nephropathy (DN), intercellular communication is disrupted. Connexins (Cx) have a crucial role in that process. Dietary ratios and supplementation with polyunsaturated fatty acids (PUFAs) can alleviate diabetic complications and cause alterations in Cx levels. Although pannexins (Panx) share similarities with members of the Cx family, their function in diabetic nephropathy has still not been fully determined. We studied the influence of PUFA supplementation on the immunoexpression of Px1 and Cx family members in diabetic kidneys of rats. Four groups of rats in experimental DM1 model were supplemented with different dietary n-6/n-3 ratios; ≈7 in control (C) and diabetic groups (STZ), ≈ 60 in the STZ + N6 group and ≈ 1 (containing 16% EPA and 19% DHA) in the STZ + N3 group. Immunoexpression of Cx40, Cx43, Cx45 and Panx1 was evaluated in the renal tissue of diabetic rats using immunohistochemistry. Diabetes significantly decreased the protein expression of Cx40 and Cx43 and increased Panx1 protein expression in the renal cortex (p < 0.05-p < 0.01). There was a significant impact of diet on Cx and Panx1 immunoexpression. Dietary supplementation with a high n-6/n-3 ratio downregulated the protein expression of Cx45 and Panx1 in diabetic rats (p < 0.05-p < 0.01), while Cx43 immunoexpression was increased in diabetic rats fed with high and low n-6/n-3 ratios (p < 0.01-p < 0.001). Hyperglycaemic conditions in DN interfere with cell-to-cell communication and disturb the connection between cells and their immediate environment due to variations in connexin and pannexin immunoexpression. These variations can be regulated by PUFA dietary intake, suggesting their beneficial effect and possible therapeutic option.
Collapse
|
11
|
Çakır M, Tekin S, Taşlıdere A, Çakan P, Düzova H, Gül CC. Protective effect of N-(p-amylcinnamoyl) anthranilic acid, phospholipase A 2 enzyme inhibitor, and transient receptor potential melastatin-2 channel blocker against renal ischemia-reperfusion injury. J Cell Biochem 2018; 120:3822-3832. [PMID: 30259992 DOI: 10.1002/jcb.27664] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 08/21/2018] [Indexed: 12/25/2022]
Abstract
The production of reactive oxygen species and inflammatory events are the underlying mechanisms of ischemia-reperfusion injury (IRI). It was determined that transient receptor potential melastatin-2 (TRPM2) channels and phospholipase A2 (PLA 2 ) enzymes were associated with inflammation and cell death. In this study, we investigated the effect of N-( p-amylcinnamoyl) anthranilic acid (ACA), a TRPM2 channel blocker, and PLA 2 enzyme inhibitor on renal IRI. A total of 36 male Sprague-Dawley rats were divided into four groups: control, ischemia-reperfusion (I/R), I/R + ACA 5 mg, I/R + ACA 25 mg. In I/R applied groups, the ischemia for 45 minutes and reperfusion for 24 hours were applied bilaterally to the kidneys. In the I/R group, serum levels of the blood urea nitrogen (BUN), creatinine, cystatin C (CysC), kidney injury molecule-1 (KIM-1), neutrophil gelatinase-associated lipocalin (NGAL), and interleukin-18 increased. On histopathological examination of renal tissue in the I/R group, the formation of glomerular and tubular damage was seen, and it was detected that there was an increase in the levels of malondialdehyde (MDA), caspase-3, total oxidant status (TOS), and oxidative stress index (OSI); and there was a decrease in total antioxidant capacity (TAC) and catalase enzyme activity. ACA administration reduced serum levels of BUN, creatinine, CysC, KIM-1, NGAL, interleukin-18. In the renal tissue, ACA administration reduced histopathological damage, levels of caspase-3, MDA, TOS, and OSI; and it increased the level of TAC and catalase enzyme activity. It has been shown with the histological and biochemical results in this study that ACA is protective against renal IRI.
Collapse
Affiliation(s)
- Murat Çakır
- Department of Physiology, Faculty of Medicine, University of Bozok, Yozgat, Turkey
| | - Suat Tekin
- Department of Physiology, Faculty of Medicine, University of Inonu, Malatya, Turkey
| | - Aslı Taşlıdere
- Department of Histology and Embryology, Faculty of Medicine, University of Inonu, Malatya, Turkey
| | - Pınar Çakan
- Department of Physiology, Faculty of Medicine, University of Inonu, Malatya, Turkey
| | - Halil Düzova
- Department of Physiology, Faculty of Medicine, University of Inonu, Malatya, Turkey
| | - Cemile Ceren Gül
- Department of Histology and Embryology, Faculty of Medicine, University of Inonu, Malatya, Turkey
| |
Collapse
|
12
|
Neuroprotective effects of epoxyeicosatrienoic acids. Prostaglandins Other Lipid Mediat 2018; 138:9-14. [DOI: 10.1016/j.prostaglandins.2018.07.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 06/19/2018] [Accepted: 07/17/2018] [Indexed: 11/22/2022]
|