1
|
Vinjavarapu LA, Yadava S, Dontiboina HR, Chakravarthi G, Kakarla R. Neuroprotective effects of Indole 3-carbinol against Scopolamine-Induced cognitive and memory impairment in rats: modulation of oxidative stress, inflammatory and cholinergic pathways. Metab Brain Dis 2025; 40:154. [PMID: 40100291 DOI: 10.1007/s11011-025-01577-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 03/11/2025] [Indexed: 03/20/2025]
Abstract
Indole 3-carbinol (I3C), a natural compound found in cruciferous vegetables, has demonstrated neuroprotective effects by modulating oxidative stress, inflammation, and cholinergic pathways. This study aimed to evaluate the efficacy of I3C in preventing cognitive impairment induced by scopolamine in rats. Male Wistar rats were assigned to six groups: Control, Scopolamine (1 mg/kg), I3C (25 mg/kg), I3C (50 mg/kg), I3C (100 mg/kg), and Donepezil (5 mg/kg). Memory function was evaluated through behavioral assessments using the Y-maze and Novel Object Recognition (NOR) tests. Biochemical analyses were conducted to assess acetylcholinesterase (AChE) activity and oxidative stress markers, including malondialdehyde (MDA), superoxide dismutase (SOD), and catalase (CAT). ELISA were utilized to quantify oxidative stress regulators (NRF2 and HO-1), inflammatory cytokines (NF-kB, TNF-α, IL-6, and IL-10), and apoptosis-related markers (Cytochrome C, caspase 9, and caspase 3). Additionally, H&E and Nissl staining were performed to evaluate histopathological abnormalities. The findings revealed that I3C administration markedly enhanced cognitive performance in the Y-maze and NOR tests, which were attributed to decreased AChE activity and increased acetylcholine (ACh) levels. Furthermore, I3C significantly alleviated oxidative stress by upregulating antioxidant enzymes, including NRF2 and HO-1. Moreover, I3C mitigated inflammatory responses, as evidenced by elevated levels of IL-10 and reduced levels of NF-kB, TNF-α, and IL-6. These findings indicate that I3C exhibits neuroprotective effects by reducing oxidative stress, suppressing inflammation, and addressing abnormalities in the cholinergic pathway, highlighting its potential as a therapeutic approach for alleviating cognitive deficits.
Collapse
Affiliation(s)
- Laksmi Anusha Vinjavarapu
- KL College of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram, 522302, India
- SIMS College of Pharmacy, Guntur, Andhra Pradesh, 522001, India
| | - Srikanth Yadava
- KL College of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram, 522302, India
| | | | | | - Ramakrishna Kakarla
- KL College of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram, 522302, India.
| |
Collapse
|
2
|
Zhang H, Dong J, Zhang J, Chen H, Liu T, Gan R, Wen J, Li Y. Effects of borneol on apoptosis of hypoxia/reoxygenation H9c2 cells and myocardial ischemia-reperfusion injury rats. Acta Cir Bras 2025; 40:e402225. [PMID: 40105602 PMCID: PMC11908740 DOI: 10.1590/acb402225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 12/29/2024] [Indexed: 03/20/2025] Open
Abstract
PURPOSE To explore the protective effects of borneol in myocardial ischemia-reperfusion injury (MIRI) and the mechanism of apoptosis. METHODS Cell viability was detected by CCK-8. The total superoxide dismutase (T-SOD) and lactate dehydrogenase (LDH) leakage of cells were tested by biochemical assay kit. Detection of apoptosis was by flow cytometry. Serum levels of creatine kinase isoenzyme MB (CK-MB), LDH, and cardiac troponin I (cTnI) were detected by enzyme-linked immunosorbent assay. Myocardial infarction area and pathological changes were observed via 2,3,5-triphenyltetrazolium chloride (TTC) staining and hematoxylin and eosin staining. The expressions of apoptosis-related proteins in cells and myocardial tissues were detected by Western blot. RESULTS H9c2 cell viability was significantly increased by pretreatment with 16 and 32 μg/mL of borneol. Borneol pretreatment significantly increased the T-SOD levels and reduced LDH leakage and apoptosis. In MIRI rats, borneol pretreatment significantly reduced serum levels of CK-MB, LDH and cTnI, decreased myocardial infarction area, and improved myocardial injury in different degree. Western blot results showed that borneol pretreatment significantly reduced the expression of Bcl-2-associated X protein (Bax) and Cysteine-aspartate protease-3 (Caspase-3) in cells and myocardial tissues of rats. CONCLUSION Borneol can protect myocardial injury cells and mitigate MIRI by inhibiting cardiomyocyte apoptosis.
Collapse
Affiliation(s)
- Hui Zhang
- North Sichuan Medical College – Department of Pharmacy – Nanchong, Sichuan – China
| | - Junfang Dong
- North Sichuan Medical College – Department of Pharmacy – Nanchong, Sichuan – China
| | - Jianwu Zhang
- North Sichuan Medical College – Department of Pharmacy – Nanchong, Sichuan – China
| | - Hongxue Chen
- North Sichuan Medical College – Department of Pharmacy – Nanchong, Sichuan – China
| | - Ting Liu
- North Sichuan Medical College – Department of Pharmacy – Nanchong, Sichuan – China
| | - Ruixue Gan
- North Sichuan Medical College – Department of Pharmacy – Nanchong, Sichuan – China
| | - Jing Wen
- North Sichuan Medical College – Department of Pharmacy – Nanchong, Sichuan – China
| | - Yangyou Li
- North Sichuan Medical College – Animal Experimental Center – Nanchong, Sichuan – China
| |
Collapse
|
3
|
Tian Y, Cheng J, Yang Y, Wang H, Fu Y, Li X, Wang W, Ma S, Xu X, Lu F, Feng P, Han S, Chen H, Hou H, Hu Q, Wu C. A 90-Day Subchronic Exposure to Heated Tobacco Product Aerosol Caused Differences in Intestinal Inflammation and Microbiome Dysregulation in Rats. Nicotine Tob Res 2025; 27:438-446. [PMID: 39028556 DOI: 10.1093/ntr/ntae179] [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: 10/04/2023] [Revised: 06/17/2024] [Accepted: 07/14/2024] [Indexed: 07/20/2024]
Abstract
INTRODUCTION Smoking is one of the most important predisposing factors of intestinal inflammatory diseases. Heated tobacco product (HTP) is a novel tobacco category that is claimed to deliver reduced chemicals to humans those reported in combustible cigarette smoke (CS). However, the effect of HTP on the intestine is still unknown. AIMS AND METHODS Our study aims to explore the potential effects of HTP on intestine. In the framework of Organization for Economic Co-operation and Development guidelines 413 guidelines, Sprague-Dawley rats were exposed to HTP aerosol and CS for 13 weeks. The atmosphere was characterized and oxidative stress and inflammation of the intestine were investigated after exposure. Furthermore, the feces we performed with 16S sequencing and metabolomics analysis. RESULTS HTP aerosol and CS led to obvious intestinal damage evidenced by increased intestinal proinflammatory cytokines and oxidative stress in male and female rats After HTP and CS exposure, the abundance that obviously changed were Lactobacillus and Turiciacter in male rats and Lactobacillus and Prevotella in female rats. HTP mainly induces the metabolism of amino acids and fatty acyls such as short-chain fatty acids and tryptophan, while CS is involved in the main metabolism of bile acids, especially indole and derivatives. Although different metabolic pathways in the gut are mediated by HTP and CS, both inflammation and oxidative stress were ultimately induced. CONCLUSIONS HTP aerosol and CS-induced intestinal damage are mediated by different gut microbiota and metabolites, while both lead to inflammation and oxidative stress. IMPLICATIONS The concentration of various harmful components in heated tobacco product aerosol is reported lower than that of traditional cigarette smoke, however, its health risk impact on consumers remains to be studied. Our research findings indicate that heated tobacco products and cigarette smoke inhalation induced intestinal damage through different metabolic pathways mediated by the gut microbiome, indicating the health risk of heated tobacco products in the intestine.
Collapse
Affiliation(s)
- Yushan Tian
- Quality Satety and FCTC Research Team, China National Tobacco Quality Supervision and Test Center, Zhengzhou, China
- Risk Assessment and Awareness Team, Key Laboratory of Tobacco Biological Effects, Zhengzhou, China
- Research Division 1, Beijing Life Science Academy, Beijing, China
- Research Division 1, Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing, China
| | - Jiale Cheng
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yanan Yang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Hongjuan Wang
- Quality Satety and FCTC Research Team, China National Tobacco Quality Supervision and Test Center, Zhengzhou, China
- Risk Assessment and Awareness Team, Key Laboratory of Tobacco Biological Effects, Zhengzhou, China
- Research Division 1, Beijing Life Science Academy, Beijing, China
- Research Division 1, Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing, China
| | - Yaning Fu
- Quality Satety and FCTC Research Team, China National Tobacco Quality Supervision and Test Center, Zhengzhou, China
- Risk Assessment and Awareness Team, Key Laboratory of Tobacco Biological Effects, Zhengzhou, China
- Research Division 1, Beijing Life Science Academy, Beijing, China
- Research Division 1, Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing, China
| | - Xianmei Li
- Quality Satety and FCTC Research Team, China National Tobacco Quality Supervision and Test Center, Zhengzhou, China
- Risk Assessment and Awareness Team, Key Laboratory of Tobacco Biological Effects, Zhengzhou, China
- Research Division 1, Beijing Life Science Academy, Beijing, China
- Research Division 1, Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing, China
| | - Wenming Wang
- Quality Satety and FCTC Research Team, China National Tobacco Quality Supervision and Test Center, Zhengzhou, China
- Risk Assessment and Awareness Team, Key Laboratory of Tobacco Biological Effects, Zhengzhou, China
- Research Division 1, Beijing Life Science Academy, Beijing, China
- Research Division 1, Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing, China
| | - Shuhao Ma
- Quality Satety and FCTC Research Team, China National Tobacco Quality Supervision and Test Center, Zhengzhou, China
- Risk Assessment and Awareness Team, Key Laboratory of Tobacco Biological Effects, Zhengzhou, China
- Research Division 1, Beijing Life Science Academy, Beijing, China
- Research Division 1, Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing, China
| | - Xiaoxiao Xu
- Quality Satety and FCTC Research Team, China National Tobacco Quality Supervision and Test Center, Zhengzhou, China
- Risk Assessment and Awareness Team, Key Laboratory of Tobacco Biological Effects, Zhengzhou, China
- Research Division 1, Beijing Life Science Academy, Beijing, China
- Research Division 1, Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing, China
| | - Fengjun Lu
- Quality Satety and FCTC Research Team, China National Tobacco Quality Supervision and Test Center, Zhengzhou, China
- Risk Assessment and Awareness Team, Key Laboratory of Tobacco Biological Effects, Zhengzhou, China
- Research Division 1, Beijing Life Science Academy, Beijing, China
- Research Division 1, Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing, China
| | - Pengxia Feng
- Quality Satety and FCTC Research Team, China National Tobacco Quality Supervision and Test Center, Zhengzhou, China
- Risk Assessment and Awareness Team, Key Laboratory of Tobacco Biological Effects, Zhengzhou, China
- Research Division 1, Beijing Life Science Academy, Beijing, China
- Research Division 1, Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing, China
| | - Shulei Han
- Quality Satety and FCTC Research Team, China National Tobacco Quality Supervision and Test Center, Zhengzhou, China
- Risk Assessment and Awareness Team, Key Laboratory of Tobacco Biological Effects, Zhengzhou, China
- Research Division 1, Beijing Life Science Academy, Beijing, China
- Research Division 1, Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing, China
| | - Huan Chen
- Quality Satety and FCTC Research Team, China National Tobacco Quality Supervision and Test Center, Zhengzhou, China
- Risk Assessment and Awareness Team, Key Laboratory of Tobacco Biological Effects, Zhengzhou, China
- Research Division 1, Beijing Life Science Academy, Beijing, China
- Research Division 1, Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing, China
| | - Hongwei Hou
- Quality Satety and FCTC Research Team, China National Tobacco Quality Supervision and Test Center, Zhengzhou, China
- Risk Assessment and Awareness Team, Key Laboratory of Tobacco Biological Effects, Zhengzhou, China
- Research Division 1, Beijing Life Science Academy, Beijing, China
- Research Division 1, Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing, China
| | - Qingyuan Hu
- Quality Satety and FCTC Research Team, China National Tobacco Quality Supervision and Test Center, Zhengzhou, China
- Risk Assessment and Awareness Team, Key Laboratory of Tobacco Biological Effects, Zhengzhou, China
- Research Division 1, Beijing Life Science Academy, Beijing, China
- Research Division 1, Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing, China
| | - Chongming Wu
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
4
|
Liu Y, Chen J, Xiong J, Hu JQ, Yang LY, Sun YX, Wei Y, Zhao Y, Li X, Zheng QH, Qi WC, Liang FR. Potential cardiac-derived exosomal miRNAs involved in cardiac healing and remodeling after myocardial ischemia-reperfusion injury. Sci Rep 2024; 14:24275. [PMID: 39414956 PMCID: PMC11484883 DOI: 10.1038/s41598-024-75517-8] [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: 01/09/2024] [Accepted: 10/07/2024] [Indexed: 10/18/2024] Open
Abstract
Migratory cells exist in the heart, such as immune cells, fibroblasts, endothelial cells, etc. During myocardium injury, such as ischemia-reperfusion (MIRI), cells migrate to the site of injury to perform repair functions. However, excessive aggregation of these cells may exacerbate damage to the structure and function of the heart, such as acute myocarditis and myocardial fibrosis. Myocardial injury releases exosomes, which are a type of vesicle with signal transduction function and the miRNA carried by exosomes can control cell migration function. Therefore, regulating this migratory cell population through cardiac-derived exosomal miRNA is crucial for protecting and maintaining cardiac function. Through whole transcriptome RNA sequencing, exosomal miRNA sequencing and single-cell dataset analysis, we (1) determined the potential molecular regulatory role of the lncRNA‒miRNA‒mRNA axis in MIRI, (2) screened four important exosomal miRNAs that could be released by cardiac tissue, and (3) screened seven genes related to cell locomotion that are regulated by four miRNAs, among which Tradd and Ephb6 may be specific for promoting migration of different cells of myocardial tissue in myocardial infarct. We generated a core miRNA‒mRNA network based on the functions of the target genes, which may be not only a target for cardiac repair but also a potential diagnostic marker for interactions between the heart and other tissues or organs. In conclusion, we elucidated the potential mechanism of MIRI in cardiac remodeling from the perspective of cell migration, and inhibition of cellular overmigration based on this network may provide new therapeutic targets for MIRI and to prevent MIRI from developing into other diseases.
Collapse
Affiliation(s)
- Yu Liu
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Jiao Chen
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Jian Xiong
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Jin-Qun Hu
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Li-Yuan Yang
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Yu-Xin Sun
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Ying Wei
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Yi Zhao
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Xiao Li
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Qian-Hua Zheng
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Wen-Chuan Qi
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China.
| | - Fan-Rong Liang
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China.
- Sichuan Clinical Medicine Research Center of Acupuncture-Moxibustion, Chengdu, 610075, China.
| |
Collapse
|
5
|
Russo MA, Puccetti M, Costantini C, Giovagnoli S, Ricci M, Garaci E, Romani L. Human and gut microbiota synergy in a metabolically active superorganism: a cardiovascular perspective. Front Cardiovasc Med 2024; 11:1411306. [PMID: 39465131 PMCID: PMC11502352 DOI: 10.3389/fcvm.2024.1411306] [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: 04/02/2024] [Accepted: 09/26/2024] [Indexed: 10/29/2024] Open
Abstract
Despite significant advances in diagnosis and treatment over recent decades, cardiovascular disease (CVD) remains one of the leading causes of morbidity and mortality in Western countries. This persistent burden is partly due to the incomplete understanding of fundamental pathogenic mechanisms, which limits the effectiveness of current therapeutic interventions. In this context, recent evidence highlights the pivotal role of immuno-inflammatory activation by the gut microbiome in influencing cardiovascular disorders, potentially opening new therapeutic avenues. Indeed, while atherosclerosis has been established as a chronic inflammatory disease of the arterial wall, accumulating data suggest that immune system regulation and anti-inflammatory pathways mediated by gut microbiota metabolites play a crucial role in a range of CVDs, including heart failure, pericardial disease, arrhythmias, and cardiomyopathies. Of particular interest is the emerging understanding of how tryptophan metabolism-by both host and microbiota-converges on the Aryl hydrocarbon Receptor (AhR), a key regulator of immune homeostasis. This review seeks to enhance our understanding of the role of the immune system and inflammation in CVD, with a focus on how gut microbiome-derived tryptophan metabolites, such as indoles and their derivatives, contribute to cardioimmunopathology. By exploring these mechanisms, we aim to facilitate the development of novel, microbiome-centered strategies for combating CVD.
Collapse
Affiliation(s)
| | - Matteo Puccetti
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Claudio Costantini
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Stefano Giovagnoli
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Maurizio Ricci
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Enrico Garaci
- San Raffaele Research Center, Sulmona, L’Aquila, Italy
| | - Luigina Romani
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
- San Raffaele Research Center, Sulmona, L’Aquila, Italy
| |
Collapse
|
6
|
Li Q, Ding J, Xia B, Liu K, Zheng K, Wu J, Huang C, Yuan X, You Q. L-theanine alleviates myocardial ischemia/reperfusion injury by suppressing oxidative stress and apoptosis through activation of the JAK2/STAT3 pathway in mice. Mol Med 2024; 30:98. [PMID: 38943069 PMCID: PMC11214244 DOI: 10.1186/s10020-024-00865-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 06/15/2024] [Indexed: 07/01/2024] Open
Abstract
BACKGROUND L-theanine is a unique non-protein amino acid in tea that is widely used as a safe food additive. We investigated the cardioprotective effects and mechanisms of L-theanine in myocardial ischemia-reperfusion injury (MIRI). METHODS The cardioprotective effects and mechanisms of L-theanine and the role of Janus Kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) signaling were investigated in MIRI mice using measures of cardiac function, oxidative stress, and apoptosis. RESULTS Administration of L-theanine (10 mg/kg, once daily) suppressed the MIRI-induced increase in infarct size and serum creatine kinase and lactate dehydrogenase levels, as well as MIRI-induced cardiac apoptosis, as evidenced by an increase in Bcl-2 expression and a decrease in Bax/caspase-3 expression. Administration of L-theanine also decreased the levels of parameters reflecting oxidative stress, such as dihydroethidium, malondialdehyde, and nitric oxide, and increased the levels of parameters reflecting anti-oxidation, such as total antioxidant capacity (T-AOC), glutathione (GSH), and superoxide dismutase (SOD) in ischemic heart tissue. Further analysis showed that L-theanine administration suppressed the MIRI-induced decrease of phospho-JAK2 and phospho-STAT3 in ischemic heart tissue. Inhibition of JAK2 by AG490 (5 mg/kg, once daily) abolished the cardioprotective effect of L-theanine, suggesting that the JAK2/STAT3 signaling pathway may play an essential role in mediating the anti-I/R effect of L-theanine. CONCLUSIONS L-theanine administration suppresses cellular apoptosis and oxidative stress in part via the JAK2/STAT3 signaling pathway, thereby attenuating MIRI-induced cardiac injury. L-theanine could be developed as a potential drug to alleviate cardiac damage in MIRI.
Collapse
Affiliation(s)
- Qi Li
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong University, #20 Xishi Road, Nantong, 226001, Jiangsu, China
| | - Jiaqi Ding
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong University, #20 Xishi Road, Nantong, 226001, Jiangsu, China
| | - Boyu Xia
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong University, #20 Xishi Road, Nantong, 226001, Jiangsu, China
| | - Kun Liu
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong University, #20 Xishi Road, Nantong, 226001, Jiangsu, China
| | - Koulong Zheng
- Department of Cardiology, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Jingjing Wu
- Department of Cardiology, Suzhou Kowloon Hospital of Shanghai Jiaotong University School of Medicine, Suzhou, Jiangsu, China
| | - Chao Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Xiaomei Yuan
- Department of Cardiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China.
| | - Qingsheng You
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong University, #20 Xishi Road, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
7
|
Peng L, Zhu X, Wang C, Jiang Q, Yu S, Song G, Liu Q, Gong P. Indole-3-carbinol (I3C) reduces apoptosis and improves neurological function after cerebral ischemia-reperfusion injury by modulating microglia inflammation. Sci Rep 2024; 14:3145. [PMID: 38326384 PMCID: PMC10850550 DOI: 10.1038/s41598-024-53636-6] [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: 07/06/2023] [Accepted: 02/03/2024] [Indexed: 02/09/2024] Open
Abstract
Indole-3-carbinol(I3C) is a tumor chemopreventive substance that can be extracted from cruciferous vegetables. Indole-3-carbinol (I3C) has been shown to have antioxidant and anti-inflammatory effects. In this study, we investigated the cerebral protective effects of I3C in an in vivo rats model of middle cerebral artery occlusion (MCAO). 8-10 Week-Old male SD rat received I3C (150 mg/kg, once daily) for 3 days and underwent 3 h of middle cerebral artery occlusion (MCAO) followed by reperfusion. The results showed that I3C pretreatment (150 mg/kg, once daily) prevented CIRI-induced cerebral infarction in rats. I3C pretreatment also decreased the mRNA expression levels of several apoptotic proteins, including Bax, caspase-3 and caspase-9, by increasing the mRNA expression levels of the anti-apoptotic protein Bcl-2. Inhibited apoptosis in the brain cells of MCAO rats. In addition, we found that I3C pretreatment reduced neuronal loss, promoted neurological recovery after ischemia-reperfusion injury and increased seven-day survival in MCAO rats. I3C pretreatment also significantly reduced the expression of inducible nitric oxide synthase (INOS), interleukin-1β (IL-1β) and interleukin-6 (IL-6) mRNA in ischemic brain tissue; Increased expression of interleukin-4 (IL-4) and interleukin-10 (IL-10) mRNA. At the same time, I3C pretreatment significantly decreased the expression of the M1 microglial marker IBA1 after cerebral ischemia-reperfusion injury and increased the expression of these results in the M2 microglial marker CD206. I3C pretreatment also significantly decreased apoptosis and death of HAPI microglial cells after hypoxia induction, decreased interleukin-1β (IL-1β) and interleukin-6 (IL-6) mRNA The expression of interleukin-4 (IL-4) and interleukin-10 (IL-10) mRNAs was increased. These results suggest that I3C protects the brain from CIRI by regulating the anti-inflammatory and anti-apoptotic effects of microglia.
Collapse
Affiliation(s)
- Long Peng
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
- Department of Neurosurgery, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou, China
| | - Xingjia Zhu
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Chenxing Wang
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Qiaoji Jiang
- Department of Neurosurgery, Affiliated Yancheng Clinical College of Xuzhou Medical University, Yancheng, 224000, Jiangsu Province, China
| | - Shian Yu
- Department of Surgery, Infectious Disease Hospital Affiliated to Nanchang University (Nanchang Ninth Hospital), Nanchang, China
| | - Gaochao Song
- Department of Neurosurgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qianqian Liu
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China.
| | - Peipei Gong
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China.
| |
Collapse
|
8
|
Oktaviono YH, Lamara AD, Tri Saputra PB, Arnindita JN, Pasahari D, Saputra ME, Made Adnya Suasti N. The roles of trimethylamine-N-oxide in atherosclerosis and its potential therapeutic aspect: A literature review. BIOMOLECULES & BIOMEDICINE 2023; 23:936-948. [PMID: 37337893 PMCID: PMC10655873 DOI: 10.17305/bb.2023.8893] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 05/21/2023] [Accepted: 05/21/2023] [Indexed: 06/21/2023]
Abstract
Current research supports the evidence that the gut microbiome (GM), which consist of gut microbiota and their biologically active metabolites, is associated with atherosclerosis development. Trimethylamine-N-oxide (TMAO), a metabolite produced by the GM through trimethylamine (TMA) oxidation, significantly enhances the formation and vulnerability of atherosclerotic plaques. TMAO promotes inflammation and oxidative stress in endothelial cells, leading to vascular dysfunction and plaque formation. Dimethyl-1-butanol (DMB), iodomethylcholine (IMC) and fluoromethylcholine (FMC) have been recognized for their ability to reduce plasma TMAO by inhibiting trimethylamine lyase, a bacterial enzyme involved in the choline cleavage anaerobic process, thus reducing TMA formation. Conversely, indole-3-carbinol (I3C) and trigonelline inhibit TMA oxidation by inhibiting flavin-containing monooxygenase-3 (FMO3), resulting in reduced plasma TMAO. The combined use of inhibitors of choline trimethylamine lyase and flavin-containing monooxygenase-3 could provide novel therapeutic strategies for cardiovascular disease prevention by stabilizing existing atherosclerotic plaques. This review aims to present the current evidence of the roles of TMA/TMAO in atherosclerosis as well as its potential therapeutic prevention aspects.
Collapse
Affiliation(s)
- Yudi Her Oktaviono
- Department of Cardiology and Vascular Medicine, General Hospital Dr. Soetomo, Faculty of Medicine, Universitas Airlangga, Surabaya, East Java, Indonesia
| | - Ariikah Dyah Lamara
- Department of Cardiology and Vascular Medicine, General Hospital Dr. Soetomo, Faculty of Medicine, Universitas Airlangga, Surabaya, East Java, Indonesia
| | - Pandit Bagus Tri Saputra
- Department of Cardiology and Vascular Medicine, General Hospital Dr. Soetomo, Faculty of Medicine, Universitas Airlangga, Surabaya, East Java, Indonesia
| | | | - Diar Pasahari
- Faculty of Medicine, Universitas Airlangga, Surabaya, East Java, Indonesia
| | - Mahendra Eko Saputra
- Department of Cardiology and Vascular Medicine, General Hospital Dr. Soetomo, Faculty of Medicine, Universitas Airlangga, Surabaya, East Java, Indonesia
| | | |
Collapse
|
9
|
Centofanti F, Buono A, Verboni M, Tomino C, Lucarini S, Duranti A, Pandolfi PP, Novelli G. Synthetic Methodologies and Therapeutic Potential of Indole-3-Carbinol (I3C) and Its Derivatives. Pharmaceuticals (Basel) 2023; 16:240. [PMID: 37259386 PMCID: PMC9960368 DOI: 10.3390/ph16020240] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 01/31/2023] [Accepted: 02/03/2023] [Indexed: 01/01/2025] Open
Abstract
Indole-3-carbinol (I3C) is a natural product contained in vegetables belonging to the Brassicaceae family and has been studied in recent decades for its biological and pharmacological properties. Herein, we will analyze: (1) the biosynthetic processes and synthetic procedures through which I3C and its main derivatives have been obtained; (2) the characteristics that lead to believe that both I3C and its derivatives are responsible for several important activities-in particular, antitumor and antiviral, through insights concerning in vitro assays and in vivo tests; (3) the mechanisms of action of the most important compounds considered; (4) the potential social impact that the enhancement of the discussed molecules can have in the prevention and treatment of the pathologies' examined field-first of all, those related to respiratory tract disorders and cancer.
Collapse
Affiliation(s)
- Federica Centofanti
- Department of Biomedicine and Prevention, Tor Vergata University of Rome, 00133 Rome, Italy
| | - Alessandro Buono
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Michele Verboni
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Carlo Tomino
- Scientific Direction—IRCCS San Raffaele Rome, 00166 Rome, Italy
| | - Simone Lucarini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Andrea Duranti
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Pier Paolo Pandolfi
- William N. Pennington Cancer Institute, Renown Health, Nevada System of Higher Education, Reno, NV 89502, USA
| | - Giuseppe Novelli
- Department of Biomedicine and Prevention, Tor Vergata University of Rome, 00133 Rome, Italy
- Department of Pharmacology, School of Medicine, University of Nevada, Reno, NV 89557, USA
| |
Collapse
|
10
|
Sun LJ, Wang XY, Xia J, Xu YM, Liao YF, Qin YY, Ge XW, Zhao PW, Xu T, Zhu XL, Gao S, Xiao R, Liu XS, Zhou K. Xin-Ji-Er-Kang protects heart from ischemia-reperfusion injury by rebalancing lipid metabolism. Front Pharmacol 2022; 13:981766. [PMID: 36081937 PMCID: PMC9445194 DOI: 10.3389/fphar.2022.981766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 07/29/2022] [Indexed: 11/29/2022] Open
Abstract
Background and Purpose: We have previously reported a cardioprotective effect with Xin-Ji-Er-Kang (XJEK) treatment in mice with myocardial infarction (MI)-induced heart failure, but no report about its potential functions in myocardial ischemia-reperfusion (MIR) injury. Here we studied the therapeutic effects of XJEK on MIR injury and investigated the mechanisms involved. Experimental Approach: MIR model of Balb/c mice induced by left anterior descending coronary artery ligation for half an hour, followed by reperfusion, was utilized to study the potential therapeutic effects of XJEK on MIR-induced cardiac injury. Ultra-performance liquid chromatography tandem Orbitrap mass spectrometry platform was used for studying serum lipid metabolic signatures. Key Results: MIR caused cardiac dysfunctions, cardiac injury, myocardial fibrosis, and increased inflammation, and all the observed abnormalities caused by MIR were largely corrected by XJEK treatment. Mechanistically, XJEK exerts its cardioprotective effect in the context of MIR injury by suppressing MIR-induced inflammation and dysregulation of serum lipid metabolism. Conclusion and Implications: We have demonstrated for the first time that XJEK protects heart from MIR injury by restoring dysregulated lipidomics. Our data provide new evidence to support a therapeutic effect for XIEK on MIR-induced cardiac injury, and pave the way for exploring the therapeutic potential of XJEK in large animal study and early clinical trial.
Collapse
Affiliation(s)
- Li-Jun Sun
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Xiao-Yu Wang
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Jie Xia
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Yan-Mei Xu
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Yu-Feng Liao
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Yuan-Yuan Qin
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Xue-Wan Ge
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Pei-Wen Zhao
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Tong Xu
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Xiao-Ling Zhu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Shan Gao
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- *Correspondence: Shan Gao, ; Rui Xiao, ; Xue-Sheng Liu, ; Kai Zhou,
| | - Rui Xiao
- Ipswich Hospital, East Suffolk and North Essex NHS Foundation Trust, Ipswich, United Kingdom
- *Correspondence: Shan Gao, ; Rui Xiao, ; Xue-Sheng Liu, ; Kai Zhou,
| | - Xue-Sheng Liu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- *Correspondence: Shan Gao, ; Rui Xiao, ; Xue-Sheng Liu, ; Kai Zhou,
| | - Kai Zhou
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- *Correspondence: Shan Gao, ; Rui Xiao, ; Xue-Sheng Liu, ; Kai Zhou,
| |
Collapse
|