1
|
She W, Su J, Ma W, Ma G, Li J, Zhang H, Qiu C, Li X. Natural products protect against spinal cord injury by inhibiting ferroptosis: a literature review. Front Pharmacol 2025; 16:1557133. [PMID: 40248093 PMCID: PMC12003294 DOI: 10.3389/fphar.2025.1557133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 03/10/2025] [Indexed: 04/19/2025] Open
Abstract
Spinal cord injury (SCI) is a severe traumatic condition that frequently results in various neurological disabilities, including significant sensory, motor, and autonomic dysfunctions. Ferroptosis, a recently identified non-apoptotic form of cell death, is characterized by the accumulation of reactive oxygen species (ROS), intracellular iron overload, and lipid peroxidation, ultimately culminating in cell death. Recent studies have demonstrated that ferroptosis plays a critical role in the pathophysiology of SCI, contributing significantly to neural cell demise. Three key cellular enzymatic antioxidants such as glutathione peroxidase 4 (GPX4), ferroptosis suppressor protein 1 (FSP1), and dihydroorotate dehydrogenase (DHODH), have been elucidated as crucial components in the defense against ferroptosis. Natural products, which are bioactive compounds mostly derived from plants, have garnered considerable attention for their potential therapeutic effects. Numerous studies have reported that several natural products can effectively mitigate neural cell death and alleviate SCI symptoms. This review summarizes fifteen natural products containing (-)-Epigallocatechin-3-gallate (EGCG), Proanthocyanidin, Carnosic acid, Astragaloside IV, Trehalose, 8-gingerol, Quercetin, Resveratrol, Albiflorin, Alpha-tocopherol, Celastrol, Hispolon, Dendrobium Nobile Polysaccharide, Silibinin, and Tetramethylpyrazine that have shown promise in treating SCI by inhibiting ferroptosis. Additionally, this review provides an overview of the mechanisms involved in these studies and proposes several perspectives to guide future research directions.
Collapse
Affiliation(s)
- Wei She
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Department of Orthopaedic Surgery, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Junxiao Su
- Department of Orthopaedic Surgery, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Wenji Ma
- Department of Orthopaedic Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Guohai Ma
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Jianfu Li
- Department of Orthopaedic Surgery, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Hui Zhang
- Department of Orthopaedic Surgery, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Cheng Qiu
- Department of Orthopaedic Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Xingyong Li
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Department of Orthopaedic Surgery, Gansu Provincial Hospital, Lanzhou, Gansu, China
| |
Collapse
|
2
|
Hao J, Ye Y, Zhang G, Shen H, Li J, Chen G. Mechanisms of nitric oxide in spinal cord injury. Med Gas Res 2024; 14:192-200. [PMID: 39073327 DOI: 10.4103/mgr.medgasres-d-23-00006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 12/04/2023] [Indexed: 07/30/2024] Open
Abstract
Spinal cord injury (SCI) is a primary lesion of the spinal cord that results from external forces or diseases, accompanied by a cascade of secondary events. Nitric oxide, an endogenous gas that functions as a signaling molecule in the human body, plays a crucial role in vasodilation of smooth muscles, regulation of blood flow and pressure, and inflammatory response. This article provides a comprehensive overview of the involvement of nitric oxide in SCI and highlights recent advances in basic research on pharmacological agents that inhibit nitric oxide elevation after SCI, offering valuable insights for future therapeutic interventions targeting SCI.
Collapse
Affiliation(s)
- Jiahui Hao
- Brain and Nerve Research Laboratory, Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | | | | | | | | | | |
Collapse
|
3
|
Sahranavard M, Hosseinjani H, Emadzadeh M, Jamialahmadi T, Sahebkar A. Effect of trehalose on mortality and disease severity in ICU-admitted patients: Protocol for a triple-blind, randomized, placebo-controlled clinical trial. Contemp Clin Trials Commun 2024; 40:101324. [PMID: 39021672 PMCID: PMC11252791 DOI: 10.1016/j.conctc.2024.101324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 05/04/2024] [Accepted: 06/13/2024] [Indexed: 07/20/2024] Open
Abstract
Background Improvement in organ failure in intensive care unit (ICU) patients is accompanied by lower mortality rate. A disaccharide, trehalose is a candidate to improve organ failure and survival by autophagy induction and enhancing oxidative stress defense. The aim of this study is to assess the effectiveness of trehalose in improving clinical outcome and reducing mortality in ICU patients. Methods a triple-blind, randomized, placebo-controlled, two arm, parallel-group, superiority clinical trial will enroll 200 ICU-admitted patients at Imam Reza hospital, Mashhad, Iran. The patients will be randomly allocated to receive either a 100 ml solution of 15 % trehalose or normal saline intravenously. Primary outcomes include ICU mortality and 60-day mortality, while secondary outcomes focus on blood parameters on day 5 and length of hospital/ICU stay. Conclusion Trehalose has demonstrated beneficial effects in diverse patients; however, no study has evaluated its effect in all ICU-admitted patients. Consequently, this study provides an opportunity to investigate whether trehalose's anti-inflammatory effects, mediated by inducing autophagy and enhancing oxidative stress defense, can play a role in reducing mortality and improving clinical outcomes in the critically ill patients. If successful, trehalose could offer a potential therapeutic approach in the ICU setting.
Collapse
Affiliation(s)
- Mehrdad Sahranavard
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hesamoddin Hosseinjani
- Department of Clinical Pharmacy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Emadzadeh
- Clinical Research Development Unit, Ghaem Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Tannaz Jamialahmadi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
4
|
Darvishzadeh Mahani F, Raji-Amirhasani A, Khaksari M, Mousavi MS, Bashiri H, Hajializadeh Z, Alavi SS. Caloric and time restriction diets improve acute kidney injury in experimental menopausal rats: role of silent information regulator 2 homolog 1 and transforming growth factor beta 1. Mol Biol Rep 2024; 51:812. [PMID: 39007943 DOI: 10.1007/s11033-024-09716-x] [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: 03/28/2024] [Accepted: 06/10/2024] [Indexed: 07/16/2024]
Abstract
BACKGROUND Estrogen has a protective impact on acute kidney injury (AKI); moreover, reducing the daily intake of calories impedes developing diseases. The present study aimed to determine the effects of calorie restriction (CR) and time restriction (TR) diets on the expression of silent information regulator 2 homolog 1 (SIRT1), transforming growth factor beta 1 (TGF-β1), and other indicators in the presence and absence of ovaries in AKI female rats. METHODS The female rats were divided into two groups, ovariectomized (OVX) and sham, and were placed on CR and TR diets for eight weeks; afterward, AKI was induced by injecting glycerol, and kidney injury indicators and biochemical parameters were measured before and after AKI. RESULTS After AKI, the levels of urine albumin excretion rate, urea, and creatinine in serum, and TGF-β1 increased, while creatinine clearance and SIRT1 decreased in kidney tissue. CR improved kidney indicators and caused a reduction in TGF-β1 and an increase in SIRT1 in ovary-intact rats. Moreover, CR prevented total antioxidant capacity (TAC) decrease and malondialdehyde (MDA) increase resulting from AKI. Before AKI, an increase in body weight, fasting blood sugar (FBS), low-density lipoprotein (LDL), triglyceride (TG), and total cholesterol (TC), and a decrease in high-density lipoprotein (HDL) were observed in OVX rats compared to sham rats, but CR prevented these changes. The effects of TR were similar to those of CR in all indicators except for TGF-β1, SIRT1, urea, creatinine, and albumin. CONCLUSION The present study indicated that CR is more effective than TR in preventing AKI, probably by increasing SIRT1 and decreasing TGF-β1 in ovary-intact animals.
Collapse
Affiliation(s)
- Fatemeh Darvishzadeh Mahani
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Alireza Raji-Amirhasani
- Endocrinology and Metabolism Research Center, Kerman University of Medical SciencesKerman, Kerman, Iran
- Department of Physiology and Pharmacology, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Khaksari
- Endocrinology and Metabolism Research Center, Kerman University of Medical SciencesKerman, Kerman, Iran.
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
- Physiology Research Center, Department of Physiology and Pharmacology, 22 Bahman Blvd, Kerman, Iran.
| | - Maryam Sadat Mousavi
- Clinical Research Development Unit, Shafa Hospital, Kerman University of Medical Sciences, Kerman, Iran
| | - Hamideh Bashiri
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Zahra Hajializadeh
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Samaneh Sadat Alavi
- Department of Physiology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
5
|
Shafiei B, Afgar A, Nematollahi MH, Shabani M, Nazari-Robati M. Effect of trehalose on miR-132 and SIRT1 in the hippocampus of aged rats. Neurosci Lett 2023; 813:137418. [PMID: 37549864 DOI: 10.1016/j.neulet.2023.137418] [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: 06/12/2023] [Revised: 08/01/2023] [Accepted: 08/02/2023] [Indexed: 08/09/2023]
Abstract
Aging causes substantial molecular to morphological changes in the brain. The brain cells are more susceptible towards oxidative damage due to impaired antioxidant defense system. Sirtuin1 (SIRT1) is a crucial cellular survival protein, which its gene has been identified as a direct target of microRNA 132 (miR-132). Trehalose contributes to preventing neuronal damage through several mechanisms. However, little is known about the interactive effects of aging and trehalose on the expression pattern of miR-132 and SIRT1 in the hippocampus. Male Wistar rats were divided into four groups. Two groups of aged (24 months) and young (4 months) rats were administered 2% trehalose solution for 30 days. Two other groups of aged and young rats received regular tap water. At the end of treatment, the levels of Sirt1 mRNA and its protein, malondialdehyde, protein carbonyl content, total antioxidant capacity, tumor necrosis factor α (TNF-α), as well as the expression of miR-132 were measured in the hippocampus. We found that trehalose treatment upregulated the expression of SIRT1 and miR-132. Moreover, administration of trehalose enhanced the level of total antioxidant activity whereas reduced the levels of lipid peroxidation, protein carbonyl content, and TNF-α. In conclusion, our data indicated that trehalose restored antioxidant status and alleviated inflammation in the hippocampus which was probably associated with the upregulation of SIRT1 and miR-132.
Collapse
Affiliation(s)
- Bentolhoda Shafiei
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran; Department of Clinical Biochemistry, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Ali Afgar
- Research Center for Hydatid Disease in Iran, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Hadi Nematollahi
- Department of Clinical Biochemistry, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Shabani
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Mahdieh Nazari-Robati
- Department of Clinical Biochemistry, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
6
|
Tang D, Quan C, Huang S, Wei F. Integrating LC-MS and HS-GC-MS for the metabolite characterization of the Chinese medicinal plant Platostoma palustre under different processing methods. Front Nutr 2023; 10:1181942. [PMID: 37275652 PMCID: PMC10235517 DOI: 10.3389/fnut.2023.1181942] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 04/18/2023] [Indexed: 06/07/2023] Open
Abstract
Platostoma palustre (or Mesona chinensis Benth) is an important medicinal and edible plant in China and Southeast Asian countries. To study the effects of different processing methods on the quality, nutrition, and flavor of P. palustre, we adopted the LC-MS and HS-GC-MS to compare the influences of tedding (S), sweating (M), and drying (H) on the metabolites and volatile substances of P. palustre. Biochemical determinations revealed that the M treatment could promote the accumulation of the contents of total sugar, soluble sugar, and total pectin compared with the H and S treatments but decrease the total flavonoid contents. LC-MS and HS-GC-MS uncovered 98 differential metabolites and 27 differential volatile substances among the three treatments, respectively. Overall, the M treatment facilitated the stabilization and improvement of the quality of polysaccharides and volatile substances, while the H treatment could promote the level of amino acids in P. palustre. The current study provided a theoretical reference for establishing standardized processing methods and sustaining the quality stability of P. palustre in future.
Collapse
Affiliation(s)
- Danfeng Tang
- Guangxi Key Laboratory of Medicinal Resources Protection and Genetic Improvement, Guangxi Botanical Garden of Medicinal Plants, Nanning, China
- National Traditional Chinese Medicine Inheritance and Innovation Center, Guangxi Botanical Garden of Medicinal Plants, Nanning, China
| | - Changqian Quan
- Guangxi Key Laboratory of Medicinal Resources Protection and Genetic Improvement, Guangxi Botanical Garden of Medicinal Plants, Nanning, China
- National Traditional Chinese Medicine Inheritance and Innovation Center, Guangxi Botanical Garden of Medicinal Plants, Nanning, China
| | - Suhua Huang
- College of Pharmacy, Guangxi Medical University, Nanning, China
| | - Fan Wei
- Guangxi Key Laboratory of Medicinal Resources Protection and Genetic Improvement, Guangxi Botanical Garden of Medicinal Plants, Nanning, China
- National Traditional Chinese Medicine Inheritance and Innovation Center, Guangxi Botanical Garden of Medicinal Plants, Nanning, China
| |
Collapse
|
7
|
Dong F, Yan W, Meng Q, Song X, Cheng B, Liu Y, Yao R. Ebselen alleviates white matter lesions and improves cognitive deficits by attenuating oxidative stress via Keap1/Nrf2 pathway in chronic cerebral hypoperfusion mice. Behav Brain Res 2023; 448:114444. [PMID: 37098387 DOI: 10.1016/j.bbr.2023.114444] [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: 12/01/2022] [Revised: 03/02/2023] [Accepted: 03/20/2023] [Indexed: 04/27/2023]
Abstract
Oxidative stress is crucial in cerebral white matter lesions (WMLs) induced by chronic cerebral hypoperfusion. Therefore, ameliorating oxidative damage is considered to be a beneficial strategy for the treatment of WMLs. Ebselen (EbSe), a small lipid organoselenium compound, its lipid peroxidation activity is mediated through the glutathione peroxidase-mimetic properties. This study aimed to investigate the role of EbSe in WMLs after bilateral common carotid artery stenosis (BCAS). The BCAS model can moderately reduce cerebral blood flow, and mimics white matter damage caused by chronic cerebral hypoperfusion or small vessel disease. Laser Speckle Contrast Imaging (LSCI) was used to monitor the cerebral blood flow of mice. The spatial learning and memory were tested by using the eight-arm maze. LFB staining was used to detect demyelination. The expression of MBP, GFAP and Iba1 was assayed by immunofluorescence. The demyelination was assessed by Transmission Electron Microscope (TEM). The activities of MDA, SOD and GSH-Px were detected by assay kits. The mRNA levels of SOD, GSH-Px and HO-1 was detected by realtime PCR. The activation of the Nrf2/ARE pathway and the expression of SOD, GSH-Px and HO-1was assessed by Western blot. EbSe ameliorated cognitive deficits and white matter lesions induced by bilateral common carotid artery stenosis (BCAS). The expression of GFAP and Iba1 was decreased in the corpus callosum of BCAS mice after EbSe treatment. Moreover, EbSe alleviated the level of MDA by elevating the expression and mRNA of SOD, GSH-Px and HO-1 in BCAS mice. Furthermore, EbSe promoted the dissociation of the Keap1/Nrf2 complex, resulting in the accumulation of Nrf2 in the nucleus. This study demonstrates a favorable effect of EbSe on cognitive impairment in a chronic cerebral hypoperfusion model, and the improvement of EbSe's antioxidant property is mediated by Keap1/Nrf2 pathway.
Collapse
Affiliation(s)
- Fuxing Dong
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu Province, China; Public Experimental Research Center, Xuzhou Medical University, Xuzhou 221004, Jiangsu Province, China
| | - Weixing Yan
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu Province, China
| | - Qiqi Meng
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu Province, China
| | - Xueli Song
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu Province, China
| | - Bing Cheng
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu Province, China
| | - Yaping Liu
- Laboratory of National Experimental Teaching and Demonstration Center of Basic Medicine, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu Province, China
| | - Ruiqin Yao
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu Province, China.
| |
Collapse
|
8
|
Radbakhsh S, Mobini M, Gumpricht E, Banach M, Jamialahmadi T, Sahebkar A. The effect of intravenous trehalose administration in a patient with multiple sulfatase deficiency. Arch Med Sci 2023; 19:1564-1568. [PMID: 37732042 PMCID: PMC10507772 DOI: 10.5114/aoms/159711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 01/23/2023] [Indexed: 09/22/2023] Open
Abstract
Introduction We describe the case of a female child with multiple sulfatase deficiency (MSD) who received intravenous (IV) trehalose (15 g/week) for 3 months. Methods The efficacy of trehalose was evaluated by comparing serum biomarkers, a quality-of-life questionnaire (HRQoL), and imaging (brain MRI and ultrasonography of liver and spleen) at weeks 0 (W0) and 12 (W12). Results The improvement in quality of life assessments along with a slight decrease in the spleen dimensions were observed after 3-month intervention. Conclusions Future research with a larger MSD population and a longer-term follow-up is warranted to determine whether trehalose can improve MSD patient health and clinical outcomes.
Collapse
Affiliation(s)
- Shabnam Radbakhsh
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Moein Mobini
- Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Maciej Banach
- Cardiovascular Research Centre, University of Zielona Gora, Zielona Gora, Poland
| | - Tannaz Jamialahmadi
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
9
|
Forouzanfar F, Vakilzadeh MM, Mehri A, Pourbagher-Shahri AM, Ganjali S, Abbasifard M, Sahebkar A. Anti-arthritic and Antioxidant Effects of Trehalose in an Experimental Model of Arthritis. RECENT ADVANCES IN INFLAMMATION & ALLERGY DRUG DISCOVERY 2023; 17:145-151. [PMID: 37622696 DOI: 10.2174/2772270817666230825093141] [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: 03/03/2023] [Revised: 07/10/2023] [Accepted: 07/10/2023] [Indexed: 08/26/2023]
Abstract
BACKGROUND The purpose of the present study was to study the potential anti-arthritic and antioxidant effects of trehalose in an experimental model of complete Freund's adjuvant (CFA)-induced arthritis. METHODS Arthritis was induced via subcutaneous injection of CFA (0.1) into the right footpad of each rat. Trehalose (10 mg/kg per day) and indomethacin (5 mg/kg) as a reference drug were intraperitoneally injected into CFA-induced arthritic rats from days 0 to 21. Changes in paw volume, pain responses, arthritic score, and oxidative/antioxidative parameters were determined. RESULTS Trehalose administration could significantly decrease arthritis scores (p <0.01) and paw edema (p <0.001), and significantly increase the nociceptive threshold (p <0.05) in CFA-induced arthritic rats. Trehalose also significantly reduced the pro-oxidant-antioxidant balance values when compared to CFA treatment alone. In addition, no significant difference was found between the trehalose group and indomethacin as a positive control group. CONCLUSION The current study suggests that trehalose has a protective effect against arthritis, which may be mediated by antioxidative effects of this disaccharide.
Collapse
Affiliation(s)
- Fatemeh Forouzanfar
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Ali Mehri
- Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Shiva Ganjali
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT), School of Medicine, Deakin University, Geelong, Australia
| | - Mitra Abbasifard
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Department of Internal Medicine, Ali-Ibn Abi-Talib Hospital, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Medicine, The University of Western Australia, Perth, Australia
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
10
|
Cai J, Kong J, Ma S, Ban Y, Li J, Fan Z. Upregulation of TRPC6 inhibits astrocyte activation and proliferation after spinal cord injury in rats by suppressing AQP4 expression. Brain Res Bull 2022; 190:12-21. [PMID: 36115513 DOI: 10.1016/j.brainresbull.2022.09.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 09/10/2022] [Accepted: 09/13/2022] [Indexed: 11/21/2022]
Abstract
AIMS This work investigates the effects and mechanisms of inhibiting TRPC6 (a non-selective cation channel) downregulation on rat astrocyte activation and proliferation following spinal cord injury (SCI) by suppressing AQP4 expression. We used HYP9 (TRPC6-specific agonist) and TGN-020 (AQP4-specific inhibitor) to explore the relationship between TRPC6 and AQP4 and their probable protective effects on SCI. METHODS In a rat SCI model, we randomly assigned female Sprague-Dawley rats into the following four groups: Sham, SCI, SCI+HYP9, and SCI+TGN-020. Western blotting and immunofluorescence staining were used to determine protein expression among groups following SCI. TUNEL and immunofluorescence staining were used to identify changes in the rate of apoptosis and the fraction of surviving neurons after SCI. The Basso-Beattie-Bresnahan open-field locomotor scale was used to identify changes in motor function after SCI. In vitro astrocyte scratch model, we first used the CCK8 assay to test the effects of varying doses of HYP9 or TGN-020 on astrocytes and then split the astrocytes into four groups: Con, Scratch, Scratch+HYP9, and Scratch+TGN-020. Western blotting and immunofluorescence were used to identify changes in the expression of target proteins. RESULTS In vivo and in vitro models, SCI dramatically decreased TRPC6 while considerably upregulating AQP4, glial fibrillary acidic protein (GFAP), and proliferating cell nuclear antigen (PCNA) expression. However, HYP9 or TGN-020 significantly suppressed activation of astrocytes, promoted neurons survival in the anterior horn of the spinal cords, and benefited the recovery of motor function in the hind limbs of rats following SCI. Interestingly, TRPC6 agonists dramatically suppressed AQP4 overexpression, indicating that the probable mechanism of HYP9 benefiting alleviation of SCI may be connected to AQP4 inhibition and astrocyte activation and proliferation reduction. CONCLUSION we discovered for the first time that HYP9 inhibits astrocyte activation and proliferation by inhibiting AQP4 in SCI rats in vivo and in vitro models and that it preserves neuronal survival and functional recovery after SCI.
Collapse
Affiliation(s)
- Jiajun Cai
- Department of Orthopedics, The First Affiliated Hospital, Jinzhou Medical University, Jinzhou 121000, China
| | - Jundong Kong
- Department of Orthopedics, The First Affiliated Hospital, Jinzhou Medical University, Jinzhou 121000, China
| | - Song Ma
- Department of Orthopedics, The First Affiliated Hospital, Jinzhou Medical University, Jinzhou 121000, China
| | - Yaozu Ban
- Department of Orthopedics, The First Affiliated Hospital, Jinzhou Medical University, Jinzhou 121000, China
| | - Jian Li
- Department of Orthopedics, The First Affiliated Hospital, Jinzhou Medical University, Jinzhou 121000, China.
| | - Zhongkai Fan
- Department of Orthopedics, The First Affiliated Hospital, Jinzhou Medical University, Jinzhou 121000, China.
| |
Collapse
|
11
|
Trehalose-Carnosine Prevents the Effects of Spinal Cord Injury Through Regulating Acute Inflammation and Zinc(II) Ion Homeostasis. Cell Mol Neurobiol 2022; 43:1637-1659. [PMID: 36121569 PMCID: PMC10079760 DOI: 10.1007/s10571-022-01273-w] [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: 12/22/2021] [Accepted: 08/11/2022] [Indexed: 11/03/2022]
Abstract
Spinal cord injury (SCI) leads to long-term and permanent motor dysfunctions, and nervous system abnormalities. Injury to the spinal cord triggers a signaling cascade that results in activation of the inflammatory cascade, apoptosis, and Zn(II) ion homeostasis. Trehalose (Tre), a nonreducing disaccharide, and L-carnosine (Car), (β-alanyl-L-histidine), one of the endogenous histidine dipeptides have been recognized to suppress early inflammatory effects, oxidative stress and to possess neuroprotective effects. We report on the effects of the conjugation of Tre with Car (Tre-car) in reducing inflammation in in vitro and in vivo models. The in vitro study was performed using rat pheochromocytoma cells (PC12 cell line). After 24 h, Tre-car, Car, Tre, and Tre + Car mixture treatments, cells were collected and used to investigate Zn2+ homeostasis. The in vivo model of SCI was induced by extradural compression of the spinal cord at the T6-T8 levels. After treatments with Tre, Car and Tre-Car conjugate 1 and 6 h after SCI, spinal cord tissue was collected for analysis. In vitro results demonstrated the ionophore effect and chelating features of L-carnosine and its conjugate. In vivo, the Tre-car conjugate treatment counteracted the activation of the early inflammatory cascade, oxidative stress and apoptosis after SCI. The Tre-car conjugate stimulated neurotrophic factors release, and influenced Zn2+ homeostasis. We demonstrated that Tre-car, Tre and Car treatments improved tissue recovery after SCI. Tre-car decreased proinflammatory, oxidative stress mediators release, upregulated neurotrophic factors and restored Zn2+ homeostasis, suggesting that Tre-car may represent a promising therapeutic agent for counteracting the consequences of SCI.
Collapse
|
12
|
Ghorbani Dehbalaei M, Sahebkar A, Safarian M, Khadem-Rezaiyan M, Rezaee H, Naeini F, Norouzy A. Study protocol for a pilot randomised controlled trial evaluating the effectiveness of oral trehalose on inflammatory factors, oxidative stress, nutritional and clinical status in traumatic head injury patients receiving enteral nutrition. BMJ Open 2022; 12:e060605. [PMID: 36123055 PMCID: PMC9486343 DOI: 10.1136/bmjopen-2021-060605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
INTRODUCTION In traumatic brain injury (TBI) patients, inflammatory processes and oxidative stress have been linked to the development of neurodegenerative diseases, disability, increased rate of muscle catabolism, malnutrition, hospital stay and mortality. Previous in vitro and in vivo studies have shown that trehalose can decrease inflammatory and oxidative factors. Therefore, the present study was designed to evaluate the effect of oral trehalose consumption on this marker in critically ill TBI patients at intensive care unit (ICU). METHODS AND ANALYSIS This study is a pilot randomised, prospective and double-blind clinical trial. The study sample size is of 20 (10 patients in each group) TBI patients aged 18-65 years at ICU. Randomisation is performed by permuted block randomisation method. The allocation ratio is 1:1. An intervention group will receive 30 g of trehalose instead, as a part of the carbohydrate of daily bolus enteral feeding and the control group will receive standard isocaloric hospital bolus enteral feeding for 12 days. The inflammatory factors (C reactive protein, interleukin 6) and oxidative stress markers (glutathione, malondialdehyde, superoxide dismutase, pro-oxidant-antioxidant balance, total antioxidant capacity) will be measured at the baseline, at the 6th day, and at the end of the study (12th day). Sequential Organ Failure Assessment, Acute Physiology and Chronic Health Evaluation II, Nutrition Risk in the Critically ill scores, 28-day mortality, anthropometric assessments and the clinical and nutritional status will be measured. Each patient's nutritional needs will be calculated individually. The statistical analysis would be based on the intention to treat. ETHICS AND DISSEMINATION The vice-chancellor of the research centre of Mashhad University of Medical Sciences is sponsoring this study. IR.MUMS.MEDICAL.REC.1400.113. TRIAL REGISTRATION NUMBER Iranian Registry of Clinical Trials (IRCT) Id: IRCT20210508051223N1, Registration date: 26 July 2021.
Collapse
Affiliation(s)
- Moazzameh Ghorbani Dehbalaei
- Department of Clinical Nutrition, School of Nutritional Science, Mashhad University of Medical Sciences, Mashhad, Iran
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Safarian
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Khadem-Rezaiyan
- Resident of Community Medicine, Community Medicine Department, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamid Rezaee
- Department of Neurosurgery, Shahid Kamyab Hospital, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Naeini
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Abdolreza Norouzy
- Nutrition Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
13
|
Shafiei B, Shabani M, Afgar A, Rajizadeh MA, Nazari-Robati M. Trehalose Attenuates Learning and Memory Impairments in Aged Rats via Overexpression of miR-181c. Neurochem Res 2022; 47:3309-3317. [PMID: 35906351 DOI: 10.1007/s11064-022-03687-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 07/05/2022] [Accepted: 07/11/2022] [Indexed: 12/25/2022]
Abstract
MicroRNAs have been recognized as important regulators of the aging process. Trehalose, a natural disaccharide, displays protective effects against neuronal impairment through several mechanisms. However, little is known about the interactive effects of aging and trehalose on behavioral function and underlying miRNA expression patterns in the hippocampus of young and old rats. Male Wistar rats were divided into four groups. Two groups of aged (24 months) and young (4 months) rats were administered 2% trehalose solution for 30 days. Two other groups of aged and young rats received regular tap water. At the end of treatment, rats were assessed for cognitive behavior using the Morris water maze test. The expression level of miR-181c and mir-34c was also measured by qRT-PCR. We found that trehalose treatment reduced learning and memory impairment in old rats compared to control old animals (p < 0.05). In contrast, cognitive performance was not significantly improved in trehalose-treated young rats in comparison with young controls (p > 0.05). We also showed that the expression level of miR-181c was significantly increased in trehalose-treated rats (p < 0.01). However, analysis of miR-34c expression level indicated no significant difference between trehalose-treated old rats and non-treated old animals (p > 0.05). Our results indicated that trehalose treatment improved learning and memory function in aged rats by targeting miR-181c. Therefore, trehalose administration may provide a therapeutic strategy to ameliorate age-associated cognitive impairment.
Collapse
Affiliation(s)
- Bentolhoda Shafiei
- Department of Clinical Biochemistry, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Shabani
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Ali Afgar
- Research Center for Hydatid Disease in Iran, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Amin Rajizadeh
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
- Physiology Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Mahdieh Nazari-Robati
- Department of Clinical Biochemistry, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
14
|
Gong F, Ge T, Liu J, Xiao J, Wu X, Wang H, Zhu Y, Xia D, Hu B. Trehalose inhibits ferroptosis via NRF2/HO-1 pathway and promotes functional recovery in mice with spinal cord injury. Aging (Albany NY) 2022; 14:3216-3232. [PMID: 35400664 PMCID: PMC9037257 DOI: 10.18632/aging.204009] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 03/25/2022] [Indexed: 11/25/2022]
Abstract
Spinal cord injury (SCI) is the main cause of severe damage to the central nervous system and leads to irreversible tissue loss and neurological dysfunction. Ferroptosis is a cell death pattern, newly discovered in recent years. Ferroptosis is an oxidizing cell death induced by small molecules, and is an iron-dependent process caused by the imbalance between the generation and degradation of lipid reactive oxygen species (ROS) in cells. As an antioxidant, trehalose can effectively prevent lipid peroxidation. Studies have reported that trehalose can improve the prognosis of SCI. However, it is unclear whether these benefits are related to ferroptosis. In this study, we demonstrated for the first time that trehalose reduces the degeneration and iron accumulation of neurons by inhibiting the production of ROS and ferroptosis caused by lipid peroxides after SCI, thus promoting the survival of neurons and improving the recovery of motor function. More specifically, we found that trehalose inhibited the expansion of cavities in the nerve tissue of mice with SCI, inhibited neuron loss, and improved functional recovery. In terms of mechanism, our results indicate that the neuroprotective effect of trehalose is due to the activation of the NRF2/HO-1 pathway, which in turn inhibits ferroptosis and ferroptosis-related inflammation. Our findings provide important insights into the previously unknown role of trehalose in SCI, as well as new evidence supporting the hypothesis that suppression of ferroptosis plays a key neuroprotective role in SCI.
Collapse
Affiliation(s)
- Fangyi Gong
- Department of Orthopedics, Ningbo First Hospital, Ningbo, China
| | - Ting Ge
- Department of Orthopedics, Ningbo First Hospital, Ningbo, China
| | - Jing Liu
- Department of Emergency Medicine, Ningbo First Hospital, Ningbo, China
| | - Jin Xiao
- Department of Orthopedics, Ningbo First Hospital, Ningbo, China
| | - Xiaochuan Wu
- Department of Orthopedics, Ningbo First Hospital, Ningbo, China
| | - Hehui Wang
- Department of Orthopedics, Ningbo First Hospital, Ningbo, China
| | - Yingchun Zhu
- Department of Orthopedics, Ningbo First Hospital, Ningbo, China
| | - Dongdong Xia
- Department of Orthopedics, Ningbo First Hospital, Ningbo, China
| | - Baiwen Hu
- Department of Orthopedics, Ningbo First Hospital, Ningbo, China
| |
Collapse
|
15
|
Mobini M, Radbakhsh S, Kubaski F, Eshraghi P, Vakili S, Vakili R, Khalili M, Varesvazirian M, Jamialahmadi T, Alamdaran SA, Sayedi SJ, Rajabi O, Emami SA, Reiner Ž, Sebkar A. Impact of Intravenous Trehalose Administration in Patients with Niemann-Pick Disease Types A and B. J Clin Med 2022; 11:247. [PMID: 35011993 PMCID: PMC8745869 DOI: 10.3390/jcm11010247] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/21/2021] [Accepted: 01/01/2022] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND AND AIMS Niemann-Pick disease (NPD) types A (NPA) and B (NPB) are caused by deficiency of the acid sphingomyelinase enzyme, which is encoded by the SMPD1 gene, resulting in progressive pathogenic accumulation of lipids in tissues. Trehalose has been suggested as an autophagy inducer with therapeutic neuroprotective effects. We performed a single-arm, open-label pilot study to assess the potential efficacy of trehalose treatment in patients with NPA and NPB patients. METHODS Five patients with NPD type A and B were enrolled in an open-label, single-arm clinical trial. Trehalose was administrated intravenously (IV) (15 g/week) for three months. The efficacy of trehalose in the management of clinical symptoms was evaluated in patients by assessing the quality of life, serum biomarkers, and high-resolution computed tomography (HRCT) of the lungs at the baseline and end of the interventional trial (day 0 and week 12). RESULTS The mean of TNO-AZL Preschool children Quality of Life (TAPQOL) scores increased in all patients after intervention at W12 compared to the baseline W0, although the difference was not statistically significant. The serum levels of lyso-SM-509 and lyso-SM were decreased in three and four patients out of five, respectively, compared with baseline. Elevated ALT and AST levels were decreased in all patients after 12 weeks of treatment; however, changes were not statistically significant. Pro-oxidant antioxidant balance (PAB) was also decreased and glutathione peroxidase (GPX) activity was increased in serum of patients at the end of the study. Imaging studies of spleen and lung HRCT showed improvement of symptoms in two patients. CONCLUSIONS Positive trends in health-related quality of life (HRQoL), serum biomarkers, and organomegaly were observed after 3 months of treatment with trehalose in patients with NPA and NPB. Although not statistically significant, due to the small number of patients enrolled, these results are encouraging and should be further explored.
Collapse
Affiliation(s)
- Moein Mobini
- Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran;
| | - Shabnam Radbakhsh
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran;
- Department of Medical Biotechnology and Nanotechnology, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran
| | - Francyne Kubaski
- Department of Genetics, UFRGS, Porto Alegre 91501970, Brazil;
- Medical Genetics Service, HCPA, Porto Alegre 90035903, Brazil
- Biodiscovery Lab, HCPA, Porto Alegre 90035903, Brazil
| | - Peyman Eshraghi
- Department of Pediatric Diseases, Akbar Hospital, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad 9177897157, Iran;
| | - Saba Vakili
- Medical Genetic Research Center, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran; (S.V.); (R.V.)
| | - Rahim Vakili
- Medical Genetic Research Center, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran; (S.V.); (R.V.)
| | - Manijeh Khalili
- Children and Adolescents Health Research Center, Research Institute of cellular and Molecular Science in Infectious Diseases, Zahedan University of Medical Science, Zahedan 9816743463, Iran;
| | - Majid Varesvazirian
- Shafa Hospital, Kerman University of Medical Sciences, Kerman 7618751151, Iran;
| | - Tannaz Jamialahmadi
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran;
| | - Seyed Ali Alamdaran
- Pediatric Radiology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran;
| | - Seyed Javad Sayedi
- Department of Pediatrics, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran;
| | - Omid Rajabi
- Department of Pharmaceutical and Food Control, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 9177948954, Iran;
| | - Seyed Ahmad Emami
- Department of Traditional Pharmacy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 9177948954, Iran;
| | - Željko Reiner
- Department of Internal Medicine, University Hospital Center Zagreb, University of Zagreb, Kišpatićeva 12, 1000 Zagreb, Croatia;
| | - Amirhossein Sebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 9177948954, Iran
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 9177948954, Iran
| |
Collapse
|
16
|
Masoudi A, Jorjani M, Alizadeh M, Mirzamohammadi S, Mohammadi M. Anti-inflammatory and antioxidant effects of astaxanthin following spinal cord injury in a rat animal model. Brain Res Bull 2021; 177:324-331. [PMID: 34688832 DOI: 10.1016/j.brainresbull.2021.10.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 10/11/2021] [Accepted: 10/19/2021] [Indexed: 10/20/2022]
Abstract
Spinal cord injury (SCI) is a severely debilitating problem leading to substantial decrease in the quality of life. After spinal cord injury, inflammation and oxidative stress plays a key role in initiating the secondary injury cascades leading to progressive tissue degradation and extreme functional deficits. Given that the primary mechanical injuries to spinal cord are rarely repaired, the pharmacological interventions may improve the neurological outcomes caused by secondary injury. Astaxanthin (AST) is considered as a xanthophyll carotenoid with potent antioxidant and anti-inflammatory properties, which has various pharmacological activities. In the present study, we aimed to firstly assess the protective effect of AST, and then to define the AST mechanism of action on a rat model of SCI. Based on the results of von Frey test, AST treatment significantly alleviated the SCI-induced neuropathic pain compared with the control groups (P < 0.05). The expression analysis by western blot shows reduced expression levels of COX-2, TNF-α, IL-1β, and IL-6 following AST treatment (P < 0.05). The activity of antioxidant enzymes was evaluated using ELISA. Therefore, ELISA experiments showed a significant reduction in the level of oxidative stress in SCI rat following AST treatment (P < 0.05). Furthermore, histopathological evaluations revealed that myelinated white matter and motor neuron number were significantly preserved after treatment with AST (P < 0.05). In conclusion, our study shows that AST could improve SCI through anti-inflammatory and antioxidant effects which leads to decreased tissue damage and mechanical pain after SCI.
Collapse
Affiliation(s)
- Alireza Masoudi
- Department of Pharmacology, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran.
| | - Masoumeh Jorjani
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Morteza Alizadeh
- Department of tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Solmaz Mirzamohammadi
- Department of Pharmacology, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Mola Mohammadi
- Physiology Departmen, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
17
|
Bahri F, Khaksari M, Movahedinia S, Shafiei B, Rajizadeh MA, Nazari-Robati M. Improving SIRT1 by trehalose supplementation reduces oxidative stress, inflammation, and histopathological scores in the kidney of aged rats. J Food Biochem 2021; 45:e13931. [PMID: 34494279 DOI: 10.1111/jfbc.13931] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 08/13/2021] [Accepted: 08/28/2021] [Indexed: 12/13/2022]
Abstract
The aging process leads to progressive loss of kidney function. Sirtuin1 (SIRT1) exerts renoprotective effects by conferring resistance to cellular stresses. Trehalose potentially displayed various beneficial effects to promote health span. In this study, we investigated the effects of trehalose on renal SIRT1 and kidney function in senescent rats. Trehalose (2% w/v) was administrated in drinking water for 1 month to male aged rats (24 months). Then, the level of SIRT1 mRNA and protein, malondialdehyde, total antioxidant capacity, tumor necrosis factor α as well as parameters related to the function and histology of the kidneys were evaluated. Trehalose supplementation increased the level of SIRT1, whereas alleviated the level of oxidative stress, inflammation, and histopathology scores in senescent tissues. However, trehalose administration did not alter kidney function indices in old rats. Collectively, these findings suggested that trehalose was an effective intervention to ameliorate some aspects of age-associated injury in the old kidneys. PRACTICAL APPLICATIONS: Aging is associated with impairment in renal structure and function. Trehalose is a natural disaccharide, which is widely distributed in many organisms. The consumption of trehalose as a dietary supplement is increasing worldwide. This study showed that trehalose administration to aged rats had renoprotective effects through reducing oxidative stress and inflammation, which was mediated by SIRT1. Our results provide useful information for individuals using this sugar as a supplement.
Collapse
Affiliation(s)
- Faegheh Bahri
- Department of Clinical Biochemistry, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Khaksari
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Sajjadeh Movahedinia
- Pathology and Stem Cell Research Center, Department of Pathology, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Bentolhoda Shafiei
- Department of Clinical Biochemistry, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Amin Rajizadeh
- Physiology Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Mahdieh Nazari-Robati
- Department of Clinical Biochemistry, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
18
|
Trehalose Augments Neuron Survival and Improves Recovery from Spinal Cord Injury via mTOR-Independent Activation of Autophagy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8898996. [PMID: 34336117 PMCID: PMC8289614 DOI: 10.1155/2021/8898996] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 06/08/2021] [Indexed: 01/11/2023]
Abstract
Spinal cord injury (SCI) is a major cause of irreversible nerve injury and leads to serious tissue loss and neurological dysfunction. Thorough investigation of cellular mechanisms, such as autophagy, is crucial for developing novel and effective therapeutics. We administered trehalose, an mTOR-independent autophagy agonist, in SCI rats suffering from moderate compression injury to elucidate the relationship between autophagy and SCI and evaluate trehalose's therapeutic potential. 60 rats were divided into 4 groups and were treated with either control vehicle, trehalose, chloroquine, or trehalose + chloroquine 2 weeks prior to administration of moderate spinal cord crush injury. 20 additional sham rats were treated with control vehicle. H&E staining, Nissl staining, western blot, and immunofluorescence studies were conducted to examine nerve morphology and quantify autophagy and mitochondrial-dependent apoptosis at various time points after surgery. Functional recovery was assessed over a period of 4 weeks after surgery. Trehalose promotes autophagosome recruitment via an mTOR-independent pathway, enhances autophagy flux in neurons, inhibits apoptosis via the intrinsic mitochondria-dependent pathway, reduces lesion cavity expansion, decreases neuron loss, and ultimately improves functional recovery following SCI (all p < 0.05). Furthermore, these effects were diminished upon administration of chloroquine, an autophagy flux inhibitor, indicating that trehalose's beneficial effects were due largely to activation of autophagy. This study presents new evidence that autophagy plays a critical neuroprotective and neuroregenerative role in SCI, and that mTOR-independent activation of autophagy with trehalose leads to improved outcomes. Thus, trehalose has great translational potential as a novel therapeutic agent after SCI.
Collapse
|
19
|
Han GH, Kim SJ, Ko WK, Lee D, Han IB, Sheen SH, Hong JB, Sohn S. Transplantation of tauroursodeoxycholic acid-inducing M2-phenotype macrophages promotes an anti-neuroinflammatory effect and functional recovery after spinal cord injury in rats. Cell Prolif 2021; 54:e13050. [PMID: 33960559 PMCID: PMC8168422 DOI: 10.1111/cpr.13050] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/25/2021] [Accepted: 04/13/2021] [Indexed: 12/14/2022] Open
Abstract
Objectives In this study, we study the transplantation of tauroursodeoxycholic acid (TUDCA)‐induced M2‐phenotype (M2) macrophages and their ability to promote anti‐neuroinflammatory effects and functional recovery in a spinal cord injury (SCI) model. Methods To this end, compared to the granulocyte‐macrophage colony‐stimulating factor (GM‐CSF), we evaluated whether TUDCA effectively differentiates bone marrow–derived macrophages (BMDMs) into M2 macrophages. Results The M2 expression markers in the TUDCA‐treated BMDM group were increased more than those in the GM‐CSF‐treated BMDM group. After the SCI and transplantation steps, pro‐inflammatory cytokine levels and the mitogen‐activated protein kinase (MAPK) pathway were significantly decreased in the TUDCA‐induced M2 group more than they were in the GM‐CSF‐induced M1 group and in the TUDCA group. Moreover, the TUDCA‐induced M2 group showed significantly enhanced tissue volumes and improved motor functions compared to the GM‐CSF‐induced M1 group and the TUDCA group. In addition, biotinylated dextran amine (BDA)–labelled corticospinal tract (CST) axons and neuronal nuclei marker (NeuN) levels were increased in the TUDCA‐induced M2 group more than those in the GM‐CSF‐induced M1 group and the TUDCA group. Conclusions This study demonstrates that the transplantation of TUDCA‐induced M2 macrophages promotes an anti‐neuroinflammatory effect and motor function recovery in SCI. Therefore, we suggest that the transplantation of TUDCA‐induced M2 macrophages represents a possible alternative cell therapy for SCI.
Collapse
Affiliation(s)
- Gong Ho Han
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si, Korea.,Department of Biomedical Science, CHA University, Seongnam-si, Korea
| | - Seong Jun Kim
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si, Korea.,Department of Biomedical Science, CHA University, Seongnam-si, Korea
| | - Wan-Kyu Ko
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si, Korea.,Department of Biomedical Science, CHA University, Seongnam-si, Korea
| | - Daye Lee
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si, Korea.,Department of Biomedical Science, CHA University, Seongnam-si, Korea
| | - In-Bo Han
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si, Korea
| | - Seung Hun Sheen
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si, Korea
| | - Je Beom Hong
- Department of Neurosurgery, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Seil Sohn
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si, Korea
| |
Collapse
|
20
|
Inhibition of ERK1/2 phosphorylation attenuates spinal cord injury induced astrocyte activation and inflammation through negatively regulating aquaporin-4 in rats. Brain Res Bull 2021; 170:162-173. [PMID: 33592275 DOI: 10.1016/j.brainresbull.2021.02.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 01/31/2021] [Accepted: 02/08/2021] [Indexed: 01/09/2023]
Abstract
The extracellular signal-regulated kinase (ERK) pathway has been reported to play a pivotal role in mediating spinal cord injury (SCI) progression. The present study aimed to investigate the effects of phosphorylated ERK1/2 (p-ERK1/2) inhibition on SCI-induced astrocyte activation and inflammation and its possible mechanism in rats. Here, female Sprague-Dawley rats were randomly assigned to four groups: (1) Sham group, (2) SCI group, (3) TGN-020 group (aquaporin-4, AQP4, blocking agent), (4) PD98059 group (ERK blocking agent). A well SCI model was established by compressing the thoracic vertebra 10 level (weight 35 g, time 5 min) in rats. Western blotting and immunofluorescence staining were used to measure the expression of associated proteins after SCI. HE staining and Nissl staining were performed to detect the morphological changes of spinal cords and the number of surviving neurons following SCI, respectively. The Basso-Beattie-Bresnahan open-field rating scale was used to evaluate functional locomotor recovery following SCI in rats. Our results demonstrated that SCI significantly induced the upregulation of aquaporin-4, p-ERK1/2, glial fibrillary acidic protein, proliferating cell nuclear antigen, and proinflammatory cytokines (tumor necrosis factor-α, interleukin-6 and interleukin-1β). However, treatment with TGN-020 or PD98059 could effectively inhibit astrocyte proliferation and proinflammatory cytokine release, preserve the number of surviving ventral horn neurons, and subsequently improve the locomotor function of rats after SCI. Interestingly, the SCI-induced elevation of AQP4 expression was downregulated by p-ERK1/2 inhibition, suggesting that blocking ERK1/2 phosphorylation could attenuate astrocyte activation and inflammatory processes through negative regulation of AQP4. Therefore, p-ERK1/2 blockade may be employed as a therapeutic target for SCI.
Collapse
|
21
|
Zhang Y, DeBosch BJ. Microbial and metabolic impacts of trehalose and trehalose analogues. Gut Microbes 2020; 11:1475-1482. [PMID: 32329657 PMCID: PMC7524367 DOI: 10.1080/19490976.2020.1750273] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 03/21/2020] [Accepted: 03/25/2020] [Indexed: 02/03/2023] Open
Abstract
Trehalose is a disaccharide and fasting-mimetic that has been both canonized and vilified for its putative cardiometabolic and microbial effects. Trehalose analogues are currently under development to extend the key metabolic therapeutic actions of trehalose without adversely affecting host microbial communities. In the current study, we contrast the extent to which trehalose and its degradation-resistant analogue, lactotrehalose (LT), modulate microbial communities and host transcriptomic profiles. We demonstrate that trehalose and LT each exert adaptive metabolic and microbial effects that both overlap and diverge. We postulate that these effects depend both upon compound stability and bioavailability, and on stereospecific signal transduction. In context, the data suggest that trehalose is unlikely to be harmful, and yet it harbors unique effects that are not yet fully replicated by its analogues. These compounds are thus valuable probes to better define trehalose structure-function, and to offer as therapeutic metabolic agents.
Collapse
Affiliation(s)
- Yiming Zhang
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Brian J. DeBosch
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
- Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
22
|
Zhou K, Zheng Z, Li Y, Han W, Zhang J, Mao Y, Chen H, Zhang W, Liu M, Xie L, Zhang H, Xu H, Xiao J. TFE3, a potential therapeutic target for Spinal Cord Injury via augmenting autophagy flux and alleviating ER stress. Am J Cancer Res 2020; 10:9280-9302. [PMID: 32802192 PMCID: PMC7415792 DOI: 10.7150/thno.46566] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 06/20/2020] [Indexed: 12/11/2022] Open
Abstract
Background and Aim: Increasing evidence suggests that spinal cord injury (SCI)-induced defects in autophagic flux may contribute to an impaired ability for neurological repair following injury. Transcription factor E3 (TFE3) plays a crucial role in oxidative metabolism, lysosomal homeostasis, and autophagy induction. Here, we investigated the role of TFE3 in modulating autophagy following SCI and explored its impact on neurological recovery. Methods: Histological analysis via HE, Nissl and Mason staining, survival rate analysis, and behavioral testing via BMS and footprint analysis were used to determine functional recovery after SCI. Quantitative real-time polymerase chain reaction, Western blotting, immunofluorescence, TUNEL staining, enzyme-linked immunosorbent assays, and immunoprecipitation were applied to examine levels of autophagy flux, ER-stress-induced apoptosis, oxidative stress, and AMPK related signaling pathways. In vitro studies using PC12 cells were performed to discern the relationship between ROS accumulation and autophagy flux blockade. Results: Our results showed that in SCI, defects in autophagy flux contributes to ER stress, leading to neuronal death. Furthermore, SCI enhances the production of reactive oxygen species (ROS) that induce lysosomal dysfunction to impair autophagy flux. We also showed that TFE3 levels are inversely correlated with ROS levels, and increased TFE3 levels can lead to improved outcomes. Finally, we showed that activation of TFE3 after SCI is partly regulated by AMPK-mTOR and AMPK-SKP2-CARM1 signaling pathways. Conclusions: TFE3 is an important regulator in ROS-mediated autophagy dysfunction following SCI, and TFE3 may serve as a promising target for developing treatments for SCI.
Collapse
|
23
|
Injectable Hydrogel Containing Tauroursodeoxycholic Acid for Anti-neuroinflammatory Therapy After Spinal Cord Injury in Rats. Mol Neurobiol 2020; 57:4007-4017. [PMID: 32647974 DOI: 10.1007/s12035-020-02010-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 07/01/2020] [Indexed: 12/12/2022]
Abstract
We investigate the anti-inflammatory effects of injectable hydrogel containing tauroursodeoxycholic acid (TUDCA) in a spinal cord injury (SCI) model. To this end, TUDCA-hydrogel (TC gel) is created by immersing the synthesized hydrogel in a TUDCA solution for 1 h. A mechanical SCI was imposed on rats, after which we injected the TC gel. After the SCI and injections, motor functions and lesions were significantly improved in the TC gel group compared with those in the saline group. The TC gel significantly decreased pro-inflammatory cytokine levels compared with the saline; TUDCA and glycol chitosan-oxidized hyaluronate were mixed at a ratio of 9:1 (CHA) gel independently. In addition, the TC gel significantly suppressed the phosphorylation of extracellular signal-regulated kinase (p-ERK) and c-Jun N-terminal kinase (p-JNK) in the mitogen-activated protein kinase (MAPK) pathway compared with the saline, TUDCA, and CHA gel independently. It also decreased tumor necrosis factor-α (TNF-α) and glial fibrillary acidic protein (GFAP), inflammatory marker, at the injured sites more than those in the saline, TUDCA, and CHA gel groups. In conclusion, the results of this study demonstrate the neuroinflammatory inhibition effects of TC gel in SCI and suggest that TC gel can be an alternative drug system for SCI cases.
Collapse
|
24
|
Nosaka N, Martinon D, Moreira D, Crother TR, Arditi M, Shimada K. Autophagy Protects Against Developing Increased Lung Permeability and Hypoxemia by Down Regulating Inflammasome Activity and IL-1β in LPS Plus Mechanical Ventilation-Induced Acute Lung Injury. Front Immunol 2020; 11:207. [PMID: 32117318 PMCID: PMC7033480 DOI: 10.3389/fimmu.2020.00207] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 01/27/2020] [Indexed: 01/04/2023] Open
Abstract
Targeting inflammasome activation to modulate interleukin (IL)-1β is a promising treatment strategy against acute respiratory distress syndrome and ventilator-induced lung injury (VILI). Autophagy is a key regulator of inflammasome activation in macrophages. Here, we investigated the role of autophagy in the development of acute lung injury (ALI) induced by lipopolysaccharide (LPS) and mechanical ventilation (MV). Two hours before starting MV, 0.2 mg/kg LPS was administered to mice intratracheally. Mice were then placed on high-volume MV (30 ml/kg with 3 cmH2O positive end-expiratory pressure for 2.5 h without additional oxygen application). Mice with myeloid-specific deletion of the autophagic protein ATG16L1 (Atg16l1fl/flLysMCre) suffered severe hypoxemia (adjusted p < 0.05) and increased lung permeability (p < 0.05, albumin level in bronchoalveolar lavage fluid) with significantly higher IL-1β release into alveolar space (p < 0.05). Induction of autophagy by fasting-induced starvation led to improved arterial oxygenation (adjusted p < 0.0001) and lung permeability (p < 0.05), as well as significantly suppressed IL-1β production (p < 0.01). Intratracheal treatment with anti-mouse IL-1β monoclonal antibody (mAb; 2.5 mg/kg) significantly improved arterial oxygenation (adjusted p < 0.01) as well as lung permeability (p < 0.05). On the other hand, deletion of IL-1α gene or use of anti-mouse IL-1α mAb (2.5 mg/kg) provided no significant protection, suggesting that the LPS and MV-induced ALI is primarily dependent on IL-1β, but independent of IL-1α. These observations suggest that autophagy has a protective role in controlling inflammasome activation and production of IL-1β, which plays a critical role in developing hypoxemia and increased lung permeability in LPS plus MV-induced acute lung injury.
Collapse
Affiliation(s)
- Nobuyuki Nosaka
- Division of Infectious Diseases and Immunology, Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Daisy Martinon
- Division of Infectious Diseases and Immunology, Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Debbie Moreira
- Division of Infectious Diseases and Immunology, Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Timothy R Crother
- Division of Infectious Diseases and Immunology, Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA, United States.,Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Moshe Arditi
- Division of Infectious Diseases and Immunology, Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA, United States.,Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Kenichi Shimada
- Division of Infectious Diseases and Immunology, Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA, United States.,Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| |
Collapse
|
25
|
Lu F, Sun X, Xu X, Jiang X. SILAC-based proteomic profiling of the suppression of TGF-β1-induced lung fibroblast-to-myofibroblast differentiation by trehalose. Toxicol Appl Pharmacol 2020; 391:114916. [PMID: 32035996 DOI: 10.1016/j.taap.2020.114916] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 02/05/2020] [Indexed: 12/16/2022]
Abstract
Fibroblast-to-myofibroblast differentiation is one of the most important characteristics of pulmonary fibrosis, and screening natural compounds targeting fibroblast differentiation is always a promising approach to discover drug candidates for treatment of pulmonary fibrosis. Trehalose reportedly has many potential medical applications, especially in treating neurodegeneration diseases. However, it remains unclear whether trehalose suppresses lung fibroblast differentiation. In this work, we found that trehalose decreased the expression levels of α-smooth muscle actin (α-SMA) following the induction of transforming growth factor β1 (TGF-β1) in pretreatment, co-treatment, and post-treatment groups. Trehalose also reduced the production of type I collagen, lung fibroblast-containing gel contractility and cell filament formation in TGF-β1-stimulated MRC-5 cells. Although trehalose is a known autophagy inducer, our results showed that its suppressive effect on fibroblast differentiation was not via trehalose-induced autophagy. And it did not affect canonical TGFβ/Smad2/3 pathway. By applying proteomic profiling technology, we demonstrated that the downregulation of β-catenin was involved in the trehalose-repressive action on fibroblast differentiation. The β-catenin agonist, SKL2001, reversed the suppressive effect of trehalose on fibroblast differentiation. Overall, these experiments demonstrated that trehalose suppressed fibroblast differentiation via the downregulation of β-catenin, but not through canonical autophagy and TGFβ/Smad2/3 pathway, which is not only a novel understanding of trehalose, but also quite helpful for in vivo research of trehalose on pulmonary fibrosis in future.
Collapse
Affiliation(s)
- Fanqing Lu
- College of Pharmaceutical Sciences, Soochow University, 199 Renai Road, Suzhou 215123, China
| | - Xionghua Sun
- College of Pharmaceutical Sciences, Soochow University, 199 Renai Road, Suzhou 215123, China
| | - Xiafang Xu
- College of Pharmaceutical Sciences, Soochow University, 199 Renai Road, Suzhou 215123, China
| | - Xiaogang Jiang
- College of Pharmaceutical Sciences, Soochow University, 199 Renai Road, Suzhou 215123, China.
| |
Collapse
|
26
|
Pawar R, Lakshmi PK, Kumar S, Dobriyal N, Sahi C. Ameliorative effect of Tinospora tuberculata in insulin resistance: Potential roles of oxidative stress resistance and heat shock protein 70 (Hsp 70) modulation. Pharmacogn Mag 2020. [DOI: 10.4103/pm.pm_366_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|