1
|
Yu G, Wen W, Li Q, Chen H, Zhang S, Huang H, Zhang Q, Fu L. Heat-Processed Diet Rich in Advanced Glycation End Products Induced the Onset and Progression of NAFLD via Disrupting Gut Homeostasis and Hepatic Lipid Metabolism. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024. [PMID: 39635825 DOI: 10.1021/acs.jafc.4c08360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Epidemiologic studies have suggested an association between the consumption of dietary advanced glycation end products (dAGEs) and the incidence of nonalcoholic fatty liver disease (NAFLD). However, the precise mechanism by which dAGEs induce NAFLD development, particularly the pathogenic role of the gut-liver axis, remains poorly understood. In this study, by establishing a high-AGE diet (HAD)-fed C57BL/6 mouse model, we employed multiomics approaches combined with a series of biological analyses to investigate the effect of HAD on NAFLD in vivo. Our results showed that exposure to HAD led to fat accumulation, oxidative stress, inflammation, and fibrosis in the liver of mice. Transcriptome analysis further revealed that HAD exposure disrupted lipid metabolism and activated inflammation-related signaling pathways in the liver. Additionally, exposure to HAD induced perturbations in gut homeostasis, as evidenced by the compromised gut barrier function, reduced probiotic abundance, and increases in pathogenic bacterial proportions. Dysbiosis of gut homeostasis may further act as a trigger for the initiation and progression of NAFLD via the gut-liver axis. This study sheds light on the underlying mechanisms through which dAGEs contribute to the development of NAFLD and helps to understand the detrimental effects of food ultraprocessing products in modern diets. Future studies are needed to explore the in-depth mechanisms related to the gut-liver axis to consolidate our conclusions.
Collapse
Affiliation(s)
- Gang Yu
- School of Statistics and Mathematics and Collaborative Innovation Centre of Statistical Data, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, P. R. China
| | - Wenjiabao Wen
- School of Statistics and Mathematics and Collaborative Innovation Centre of Statistical Data, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, P. R. China
| | - Qianqian Li
- School of Statistics and Mathematics and Collaborative Innovation Centre of Statistical Data, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, P. R. China
| | - Hongbo Chen
- National Pre-packaged Food Quality Supervision and Inspection Center, Zhejiang Fangyuan Test Group Co., LTD., Hangzhou 310018, China
| | - Shuifeng Zhang
- National Pre-packaged Food Quality Supervision and Inspection Center, Zhejiang Fangyuan Test Group Co., LTD., Hangzhou 310018, China
| | - Hua Huang
- Quzhou Institute for Food and Drug Control, Quzhou 324000, China
| | - Qiaozhi Zhang
- School of Statistics and Mathematics and Collaborative Innovation Centre of Statistical Data, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, P. R. China
| | - Linglin Fu
- School of Statistics and Mathematics and Collaborative Innovation Centre of Statistical Data, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, P. R. China
| |
Collapse
|
2
|
Gangaiah D, Gu M, Zaparte A, Will O, Dolan LC, Goering A, Pillai J, Mane SP, Plata G, Helmes EB, Welsh DA, Mahajan AK. Effects of Limosilactobacillus reuteri strains PTA-126787 and PTA-126788 on intestinal barrier integrity and immune homeostasis in an alcohol-induced leaky gut model. Sci Rep 2024; 14:19584. [PMID: 39179898 PMCID: PMC11344072 DOI: 10.1038/s41598-024-70549-6] [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: 06/17/2024] [Accepted: 08/19/2024] [Indexed: 08/26/2024] Open
Abstract
Intestinal barrier is a first line of defense that prevents entry of various harmful substances from the lumen into the systemic environment. Impaired barrier function with consequent translocation of harmful substances into systemic circulation ("leaky gut") is a central theme in many gastrointestinal, autoimmune, mental, and metabolic diseases. Probiotics have emerged as a promising strategy to maintain intestinal integrity and address "leaky gut". Using in silico, in vitro and avian in vivo analyses, we previously showed that two novel L. reuteri strains, PTA-126787 (L. reuteri 3630) and PTA-126788 (L. reuteri 3632), isolated from broiler chickens possess favorable safety profiles. Consistent with a recent study, here we show that L. reuteri 3630 and 3632 are phylogenetically similar to human L. reuteri strains. Daily administration of high doses of L. reuteri 3630 and 3632 to Sprague Dawley rats for 28 days was found to be safe with no adverse effects. More importantly, administration of L. reuteri 3630 and 3632 significantly reduced markers associated with alcohol-induced leaky gut, by downregulating inflammatory cytokines and upregulating anti-inflammatory cytokines in an alcohol model of leaky gut in mice. While L. reuteri 3630 cells and supernatant showed no activation, L. reuteri 3632 cells but not supernatant showed activation of AhR, a key transcription factor that regulates gut and immune homeostasis. L. reuteri 3630 is creamish white in morphology typical of Lactobacillus species and L. reuteri 3632 displays a unique orange pigmentation, which was stable even after passaging for 480 generations. We identified a rare polyketide biosynthetic gene cluster in L. reuteri 3632 that likely encodes for the orange-pigmented secondary metabolite. Similar to L. reuteri 3632 cells, the purified orange metabolite activated AhR. All together, these data provide evidence on the phylogenetic relatedness, safety, efficacy, and one of the likely mechanisms of action of L. reuteri 3630 and 3632 for potential probiotic applications to address "leaky gut" and associated pathologies in humans.
Collapse
Affiliation(s)
| | - Min Gu
- Department of Medicine, LSU Health Sciences Center, New Orleans, LA, 70112, USA
| | - Aline Zaparte
- Department of Medicine, LSU Health Sciences Center, New Orleans, LA, 70112, USA
| | - Olaf Will
- Elanco Animal Health, Inc., Alfred-Nobel-Strasse 50, 40789, Monheim Am Rhein, Germany
| | - Laurie C Dolan
- GRAS Associates, 1180 Grand Park Avenue, North Bethesda, MD, 20852, USA
| | | | - Jason Pillai
- MicroMGx, Inc., 3440 S Dearborn St, Chicago, IL, 60616, USA
| | | | - German Plata
- BiomEdit, LLC, 2710 Innovation Way, Greenfield, IN, 46140, USA
| | - Emily B Helmes
- BiomEdit, LLC, 2710 Innovation Way, Greenfield, IN, 46140, USA
| | - David A Welsh
- Department of Medicine, LSU Health Sciences Center, New Orleans, LA, 70112, USA
| | | |
Collapse
|
3
|
Xu H, Zhou J, Ye F, Gao Y. Serum lipopolysaccharide associated with new-onset atrial fibrillation in patients with non-small-cell lung cancer a retrospective observational study. Front Surg 2024; 11:1404450. [PMID: 38783859 PMCID: PMC11112106 DOI: 10.3389/fsurg.2024.1404450] [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: 03/21/2024] [Accepted: 04/29/2024] [Indexed: 05/25/2024] Open
Abstract
Lipopolysaccharide (LPS) is related to atrial fibrillation (AF). But so far, the relationship between LPS and new-onset AF (NOAF) in patients with lung cancer is unrevealed. This study was to investigate the association between LPS and NOAF in patients after lung cancer surgery. This was a single-center retrospective clinical observational study. Patients diagnosed with non-small-cell lung cancer (NSCLC) were enrolled. All patients receiving lung cancer surgery and at least 24 h electrocardiogram (ECG) examination was recorded during the hospitalization. The incidence of NOAF in this study was 34/406 (8.4%). The univariate analysis showed that NOAF was associated with age, intraoperative blood transfusion (IBT), chronic obstructive pulmonary disorder (COPD), and LPS. After adjusting risk factors, it was found that age, IBT and LPS (OR, 1.031; 95% CI: 1.001-1.042; P = 0.002) were still risk factors for NOAF. The area under curve (AUC) value was 0.709 for the LPS. When the LPS was added to the conventional model, the Net reclassification index (NRI) and integrated discrimination index (IDI) were improved significantly. Elevated LPS is associated with an increased risk of NOAF in patients after lung cancer surgery. LPS contributed to the discrimination of the NOAF risk model and improved it markedly.
Collapse
Affiliation(s)
| | - Jie Zhou
- Department of Thoracic Surgery, Xuzhou Medical University Affiliated Hospital Sihong Branch, The First People's Hospital of Sihong County, Suqian, Jiangsu, China
| | | | | |
Collapse
|
4
|
Ren H, Wang Z, Li Y, Liu J. Association of lipopolysaccharide with new-onset atrial fibrillation in ST-segment elevation myocardial infarction. Heliyon 2024; 10:e27552. [PMID: 38496897 PMCID: PMC10944234 DOI: 10.1016/j.heliyon.2024.e27552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 03/01/2024] [Accepted: 03/01/2024] [Indexed: 03/19/2024] Open
Abstract
Background Lipopolysaccharide (LPS) is related to various cardiovascular diseases. However, the relationship between LPS and new-onset atrial fibrillation (NOAF) after ST-segment elevation myocardial infarction (STEMI) has yet to be elucidated. This study aimed to evaluate the impact of LPS on NOAF in STEMI patients. Methods This was a single-center retrospective observational study including 806 patients diagnosed with STEMI. LPS levels were determined using a commercial ELISA kit. NOAF was characterized by postadmission AF with the absence of any prior history of AF. Results A total of 806 participants were enrolled, with 752 individuals in the non-AF group (93.3%) and 54 individuals in the AF group (6.7%). Multivariable analysis showed that LPS (OR = 1.047; 95% CI: 1.029-1.065, P < 0.001) was an independent risk marker for NOAF. The analysis of the ROC demonstrated that LPS had an AUC of 0.717 in predicting NOAF. When LPS was added to the conventional model, the ability of the risk model to discriminate and reclassify NOAF was improved significantly (IDI 0.053, P = 0.001; NRI 0.510, P < 0.001). Conclusion Elevated LPS is associated with an increased risk of NOAF in STEMI patients. The integration of LPS can improve the ability to predict NOAF in STEMI patients.
Collapse
Affiliation(s)
- Honglong Ren
- Department of Gastroenterology, The First People's Hospital of Yuhang District, Hangzhou, 311100, Zhejiang, China
| | - Zhonghua Wang
- Department of Gastroenterology, The First People's Hospital of Yuhang District, Hangzhou, 311100, Zhejiang, China
| | - Yong Li
- Department of Cardiology, The First People's Hospital of Yuhang District, Hangzhou, 311100, Zhejiang, China
| | - Jinqi Liu
- Department of Cardiology, Huai'an Second People's Hospital, 223001, Jiangsu, China
| |
Collapse
|
5
|
Gao C, Koko MY, Hong W, Gankhuyag J, Hui M, Gantumur MA, Dong N. Protective Properties of Intestinal Alkaline Phosphatase Supplementation on the Intestinal Barrier: Interactions and Effects. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:27-45. [PMID: 37964463 DOI: 10.1021/acs.jafc.3c05119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2023]
Abstract
The intestinal barrier is critical for maintaining intestinal homeostasis, and its dysfunction is associated with various diseases. Recent findings have revealed the multifunctional role of intestinal alkaline phosphatase (IAP) in diverse biological processes, including gut health maintenance and function. This review summarizes the protective effects of IAP on intestinal barrier integrity, encompassing the physical, chemical, microbial, and immune barriers. We discuss the results and insights from in vitro, animal model, and clinical studies as well as the available evidence regarding the impact of diet on IAP activity and expression. IAP can also be used as an indicator to assess intestinal-barrier-related diseases. Further research into the mechanisms of action and long-term health effects of IAP in maintaining overall intestinal health is essential for its future use as a dietary supplement or functional component in medical foods.
Collapse
Affiliation(s)
- Chenzhe Gao
- Laboratory of Molecular Nutrition and Immunity, College of Animal Science and Technology, Northeast Agricultural University, Harbin, People's Republic of China 150030
- College of Food, Northeast Agricultural University, Harbin, People's Republic of China 150030
| | - Marwa Yagoub Koko
- College of Food, Northeast Agricultural University, Harbin, People's Republic of China 150030
| | - Weichen Hong
- Laboratory of Molecular Nutrition and Immunity, College of Animal Science and Technology, Northeast Agricultural University, Harbin, People's Republic of China 150030
| | - Javzan Gankhuyag
- College of Food, Northeast Agricultural University, Harbin, People's Republic of China 150030
| | - Mizhou Hui
- College of Food, Northeast Agricultural University, Harbin, People's Republic of China 150030
| | - Munkh-Amgalan Gantumur
- College of Food, Northeast Agricultural University, Harbin, People's Republic of China 150030
| | - Na Dong
- Laboratory of Molecular Nutrition and Immunity, College of Animal Science and Technology, Northeast Agricultural University, Harbin, People's Republic of China 150030
| |
Collapse
|
6
|
Gawali B, Sridharan V, Krager KJ, Boerma M, Pawar SA. TLR4-A Pertinent Player in Radiation-Induced Heart Disease? Genes (Basel) 2023; 14:genes14051002. [PMID: 37239362 DOI: 10.3390/genes14051002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/18/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
The heart is one of the organs that is sensitive to developing delayed adverse effects of ionizing radiation (IR) exposure. Radiation-induced heart disease (RIHD) occurs in cancer patients and cancer survivors, as a side effect of radiation therapy of the chest, with manifestation several years post-radiotherapy. Moreover, the continued threat of nuclear bombs or terrorist attacks puts deployed military service members at risk of exposure to total or partial body irradiation. Individuals who survive acute injury from IR will experience delayed adverse effects that include fibrosis and chronic dysfunction of organ systems such as the heart within months to years after radiation exposure. Toll-like receptor 4 (TLR4) is an innate immune receptor that is implicated in several cardiovascular diseases. Studies in preclinical models have established the role of TLR4 as a driver of inflammation and associated cardiac fibrosis and dysfunction using transgenic models. This review explores the relevance of the TLR4 signaling pathway in radiation-induced inflammation and oxidative stress in acute as well as late effects on the heart tissue and the potential for the development of TLR4 inhibitors as a therapeutic target to treat or alleviate RIHD.
Collapse
Affiliation(s)
- Basveshwar Gawali
- Department of Radiation Oncology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Vijayalakshmi Sridharan
- Division of Radiation Health, College of Pharmacy, the University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Kimberly J Krager
- Division of Radiation Health, College of Pharmacy, the University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Marjan Boerma
- Division of Radiation Health, College of Pharmacy, the University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Snehalata A Pawar
- Department of Radiation Oncology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| |
Collapse
|
7
|
Lyu Y, Huo J, Jiang W, Yang W, Wang S, Zhang S, Cheng Y, Jiang Z, Shan Q. Empagliflozin ameliorates cardiac dysfunction in heart failure mice via regulating mitochondrial dynamics. Eur J Pharmacol 2023; 942:175531. [PMID: 36690056 DOI: 10.1016/j.ejphar.2023.175531] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 01/18/2023] [Accepted: 01/19/2023] [Indexed: 01/22/2023]
Abstract
Empagliflozin has cardioprotective effects in patients with heart failure (HF). However, the mechanism by which empagliflozin protects against HF remains controversial. Study aimed to evaluate the effect of empagliflozin on myocardial fibrosis and cardiac function in HF mice and its possible mechanism. C57BL/6 mice were induced with HF by ligation of the left anterior descending coronary artery. At 4 weeks postoperation, mice were randomly given normal saline or empagliflozin for 8 weeks. Echocardiography was used to assess cardiac function. Masson's staining, immunohistochemistry and Western blot analysis were used to detect the degree of myocardial fibrosis. Changes in mitochondria were detected by observing mitochondrial morphology, measuring mitochondrial dynamics-related proteins and analysing the levels of adenosine triphosphate (ATP), adenosine monophosphate (AMP) and adenosine diphosphate (ADP). The mitochondrial fission inhibitor, mdivi1, was used to detect the relationship between mitochondrial dysfunction and cardiac dysfunction in HF mice. HF led to myocardial fibrosis and cardiac dysfunction. However, treatment with empagliflozin reduced these effects. Empagliflozin inhibited mitochondrial fission and improved energy metabolic efficiency in HF mice by regulating the expression of mitochondrial dynamics-related proteins. Similarly, mdivi1 attenuated mitochondrial dysfunction and cardiac dysfunction by inhibiting mitochondrial fission in HF mice. Regulation of mitochondrial dynamics, especially inhibition of mitochondrial fission, may be a potential target for reducing cardiac damage in patients with HF. Empagliflozin improved myocardial fibrosis and cardiac dysfunction by modulating mitochondrial dynamics in HF mice. Thus, the cardiac protective effect of empagliflozin may be related to the normalization of mitochondria and the increase in ATP production.
Collapse
Affiliation(s)
- YiTing Lyu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - JunYu Huo
- Department of Cardiology, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - WanYing Jiang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wen Yang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - ShengChan Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - ShiGeng Zhang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - YanDi Cheng
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - ZhiXin Jiang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - QiJun Shan
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
8
|
Fu H, Kong B, Zhu J, Huang H, Shuai W. Phenylacetylglutamine increases the susceptibility of ventricular arrhythmias in heart failure mice by exacerbated activation of the TLR4/AKT/mTOR signaling pathway. Int Immunopharmacol 2023; 116:109795. [PMID: 36736224 DOI: 10.1016/j.intimp.2023.109795] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/18/2023] [Accepted: 01/24/2023] [Indexed: 02/04/2023]
Abstract
BACKGROUND Intestinal microbial metabolites are a risk factor for cardiovascular diseases, and phenylacetylglutamine (PAGln) is a newly discovered intestinal metabolite in the latest study. In addition, elevated plasma PAGln concentration was associated with increased mortality and hospitalization rates in patients with heart failure (HF). However, the mechanism of PAGln leading to increased HF mortality is unclear. The present study was performed to investigate whether the PAGln deteriorated the susceptibility of ventricular arrhythmias (VAs) in the setting of HF. METHODS Thoracic aortic coarctation (TAC) was used to construct an animal model of HF in mice. Intraperitoneal injection of PAGln for 4 weeks intervened in HF mice. The concentration of PAGln was quantitatively determined by liquid chromatography-tandem mass spectrometry. Cardiac function was assessed by echocardiography; assessment of cardiac electrophysiological indexes was measured by electrocardiogram (ECG) and programmed electrical stimulation in isolated cardiac perfusion. Masson was stained for collagen deposition, and wheat germ agglutinin (WGA) was stained for the cross-sectional area of the myocytes. The qRT-PCR and Western Blotting were used to determine target gene expression in vivo and in vitro. RESULTS PAGln promoted the activation of cardiac inflammation and fibrosis and deteriorated cardiac function in HF mice. Moreover, PAGln extended APD90, shortened the ERP/APD90 and increased the incidence of VAs following HF in isolated heart perfusion. Mechanistically, PAGln significantly enhanced the activation of the TLR4/AKT/mTOR signaling pathway in vivo and in vitro. CONCLUSIONS PAGln increased the susceptibility of VAs in HF mice by activating the TLR4/AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Hui Fu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, China; Cardiovascular Research Institute of Wuhan University, Wuhan 430060, Hubei, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, Hubei, China
| | - Bin Kong
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, China; Cardiovascular Research Institute of Wuhan University, Wuhan 430060, Hubei, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, Hubei, China
| | - Jun Zhu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, China; Cardiovascular Research Institute of Wuhan University, Wuhan 430060, Hubei, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, Hubei, China
| | - He Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, China; Cardiovascular Research Institute of Wuhan University, Wuhan 430060, Hubei, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, Hubei, China.
| | - Wei Shuai
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, China; Cardiovascular Research Institute of Wuhan University, Wuhan 430060, Hubei, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, Hubei, China.
| |
Collapse
|
9
|
Ashraf H, Cossu D, Ruberto S, Noli M, Jasemi S, Simula ER, Sechi LA. Latent Potential of Multifunctional Selenium Nanoparticles in Neurological Diseases and Altered Gut Microbiota. MATERIALS (BASEL, SWITZERLAND) 2023; 16:699. [PMID: 36676436 PMCID: PMC9862321 DOI: 10.3390/ma16020699] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 12/31/2022] [Accepted: 01/06/2023] [Indexed: 06/17/2023]
Abstract
Neurological diseases remain a major concern due to the high world mortality rate and the absence of appropriate therapies to cross the blood-brain barrier (BBB). Therefore, the major focus is on the development of such strategies that not only enhance the efficacy of drugs but also increase their permeability in the BBB. Currently, nano-scale materials seem to be an appropriate approach to treating neurological diseases based on their drug-loading capacity, reduced toxicity, targeted delivery, and enhanced therapeutic effect. Selenium (Se) is an essential micronutrient and has been of remarkable interest owing to its essential role in the physiological activity of the nervous system, i.e., signal transmission, memory, coordination, and locomotor activity. A deficiency of Se leads to various neurological diseases such as Parkinson's disease, epilepsy, and Alzheimer's disease. Therefore, owing to the neuroprotective role of Se (selenium) nanoparticles (SeNPs) are of particular interest to treat neurological diseases. To date, many studies investigate the role of altered microbiota with neurological diseases; thus, the current review focused not only on the recent advancement in the field of nanotechnology, considering SeNPs to cure neurological diseases, but also on investigating the potential role of SeNPs in altered microbiota.
Collapse
Affiliation(s)
- Hajra Ashraf
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
| | - Davide Cossu
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
| | - Stefano Ruberto
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
| | - Marta Noli
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
| | - Seyedesomaye Jasemi
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
| | - Elena Rita Simula
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
| | - Leonardo A. Sechi
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
- Complex Structure of Microbiology and Virology, AOU Sassari, 07100 Sassari, Italy
| |
Collapse
|
10
|
Qiao L, Chen Y, Song X, Dou X, Xu C. Selenium Nanoparticles-Enriched Lactobacillus casei ATCC 393 Prevents Cognitive Dysfunction in Mice Through Modulating Microbiota-Gut-Brain Axis. Int J Nanomedicine 2022; 17:4807-4827. [PMID: 36246933 PMCID: PMC9562773 DOI: 10.2147/ijn.s374024] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 09/21/2022] [Indexed: 11/05/2022] Open
Abstract
Purpose The bidirectional communication between the gut and the central nervous system mediated by gut microbiota is closely related to the occurrence and development of neurodegenerative diseases, including Alzheimer's disease (AD). Selenium (Se) has been identified as playing a role against AD. Probiotics have beneficial effects on host brain function and behavior by modulating the microbiota-gut-brain axis. Herein, we evaluated the protective effects of Lactobacillus casei ATCC 393 (L. casei ATCC 393) and selenium nanoparticles-enriched L. casei ATCC 393 (L. casei ATCC 393-SeNPs) against D-galactose/aluminum chloride-induced AD model mice. Methods The Morris Water Maze (MWM) test was used to assess cognitive function of mice. The morphology and histopathological changes, antioxidant capacity and immune responses in the brain and ileum were evaluated. The alterations in intestinal permeability of the mice were determined using FITC-dextran. Gut microbiota composition was assessed using 16s rRNA sequencing. Results Thirteen weeks intervention with L. casei ATCC 393 or L. casei ATCC 393-SeNPs significantly improved cognitive dysfunction, and minimized amyloid beta (Aβ) aggregation, hyperphosphorylation of TAU protein, and prevented neuronal death by modulating Akt/cAMP-response element binding protein (CREB)/brain-derived neurotrophic factor (BDNF) signaling pathway. Moreover, compared with L. casei ATCC 393, L. casei ATCC 393-SeNPs further effectively mitigated intestinal barrier dysfunction by improving antioxidant capacity, regulating immune response, restoring gut microbiota balance, and increasing the level of short-chain fatty acids and neurotransmitters, thereby inhibiting the activation of microglia and protecting brain neurons from neurotoxicity such as oxidative stress and neuroinflammation. Conclusion These findings indicated that targeting the microbiota-gut-brain axis with L. casei ATCC 393-SeNPs may have therapeutic potential for the deficits of cognitive function in the AD model mice. Thus, we anticipate that L. casei ATCC 393-SeNPs may be a promising and safe Se nutritional supplement for use as a food additive to prevent the neurodegenerative disease.
Collapse
Affiliation(s)
- Lei Qiao
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an, People’s Republic of China
| | - Yue Chen
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an, People’s Republic of China
| | - Xiaofan Song
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an, People’s Republic of China
| | - Xina Dou
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an, People’s Republic of China
| | - Chunlan Xu
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an, People’s Republic of China,Correspondence: Chunlan Xu, The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an, People’s Republic of China, Tel +86 29-88460543, Fax +86 29-88460332, Email
| |
Collapse
|
11
|
Van den Eynde J, Jacquemyn X, Cloet N, Noé D, Gillijns H, Lox M, Gsell W, Himmelreich U, Luttun A, McCutcheon K, Janssens S, Oosterlinck W. Arteriovenous Fistulae in Chronic Kidney Disease and the Heart: Physiological, Histological, and Transcriptomic Characterization of a Novel Rat Model. J Am Heart Assoc 2022; 11:e027593. [PMID: 36205249 DOI: 10.1161/jaha.122.027593] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Arteriovenous fistulae (AVFs) are the gold standard for vascular access in those requiring hemodialysis but may put an extra hemodynamic stress on the cardiovascular system. The complex interactions between the heart, kidney, and AVFs remain incompletely understood. Methods and Results We characterized a novel rat model of five-sixths partial nephrectomy (NX) and AVFs. NX induced increases in urea, creatinine, and hippuric acid. The addition of an AVF (AVF+NX) further increased urea and a number of uremic toxins such as trimethylamine N-oxide and led to increases in cardiac index, left and right ventricular volumes, and right ventricular mass. Plasma levels of uremic toxins correlated well with ventricular morphology and function. Heart transcriptomes identified altered expression of 8 genes following NX and 894 genes following AVF+NX, whereas 290 and 1431 genes were altered in the kidney transcriptomes, respectively. Gene ontology and Kyoto Encyclopedia of Genes and Genomes analysis revealed gene expression changes related to cell division and immune activation in both organs, suppression of ribosomes and transcriptional activity in the heart, and altered renin-angiotensin signaling as well as chronodisruption in the kidney. All except the latter were worsened in AVF+NX compared with NX. Conclusions Inflammation and organ dysfunction in chronic kidney disease are exacerbated following AVF creation. Furthermore, our study provides important information for the discovery of novel biomarkers and therapeutic targets in the management of cardiorenal syndrome.
Collapse
Affiliation(s)
| | | | - Nicolas Cloet
- Department of Cardiovascular Sciences KU Leuven Leuven Belgium
| | - Dries Noé
- Department of Cardiovascular Sciences KU Leuven Leuven Belgium
| | - Hilde Gillijns
- Department of Cardiovascular Sciences KU Leuven Leuven Belgium
| | - Marleen Lox
- Department of Cardiovascular Sciences KU Leuven Leuven Belgium
| | - Willy Gsell
- MoSAIC, Biomedical MRI, Department of Imaging and Pathology KU Leuven Leuven Belgium
| | - Uwe Himmelreich
- MoSAIC, Biomedical MRI, Department of Imaging and Pathology KU Leuven Leuven Belgium
| | - Aernout Luttun
- Department of Cardiovascular Sciences KU Leuven Leuven Belgium.,Endothelial Cell Biology Unit, Center for Molecular and Vascular Biology KU Leuven Leuven Belgium
| | - Keir McCutcheon
- Department of Cardiology Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle University Newcastle United Kingdom
| | - Stefan Janssens
- Department of Cardiovascular Sciences KU Leuven Leuven Belgium.,Department of Cardiovascular Diseases University Hospitals Leuven Leuven Belgium
| | - Wouter Oosterlinck
- Department of Cardiovascular Sciences KU Leuven Leuven Belgium.,Department of Cardiovascular Diseases University Hospitals Leuven Leuven Belgium
| |
Collapse
|
12
|
Ju Z, Shen L, Zhou M, Luo J, Yu Z, Qu C, Lei R, Lei M, Huang R. Helicobacter pylori and Alzheimer's Disease-Related Metabolic Dysfunction: Activation of TLR4/Myd88 Inflammation Pathway from p53 Perspective and a Case Study of Low-Dose Radiation Intervention. ACS Chem Neurosci 2022; 13:1065-1081. [PMID: 35312296 DOI: 10.1021/acschemneuro.2c00082] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Gut dysbiosis is observed in Alzheimer's disease (AD) and is frequently associated with AD-induced metabolic dysfunction. However, the extent and specific underlying molecular mechanisms triggered by alterations of gut microbiota composition and function mediating AD-induced metabolic dysfunction in AD remain incompletely uncovered. Here, we indicate that Helicobacter pylori (H. pylori) is abundant in AD patients with relative metabolic dysfunction. Fecal microbiota transplantation from the AD patients promoted metabolic dysfunction in mice and increased gut permeability. H. pylori increased gut permeability through activation of the TLR4/Myd88 inflammation pathway in a p53-dependent manner, leading to metabolic dysfunction. Moreover, p53 deficiency reduced bile acid concentration, leading to an increased abundance of H. pylori colonization. Overall, these data identify H. pylori as a key promoter of AD-induced metabolic dysfunction.
Collapse
Affiliation(s)
- Zhao Ju
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan 410078, China
| | - Liangfang Shen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Meiling Zhou
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan 410078, China
| | - Jinhua Luo
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan 410078, China
| | - Zijian Yu
- The First Affiliated Hospital, University of South China, 69 Chuanshan Road, Hengyang, Hunan 421001, People’s Republic of China
| | - Can Qu
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan 410078, China
| | - Ridan Lei
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan 410078, China
| | - Mingjun Lei
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Ruixue Huang
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan 410078, China
| |
Collapse
|
13
|
Menichelli D, Carnevale R, Nocella C, Cammisotto V, Castellani V, Bartimoccia S, Frati G, Pignatelli P, Pastori D. Circulating Lipopolysaccharides and Impaired Antioxidant Status in Patients With Atrial Fibrillation. Data From the ATHERO-AF Study. Front Cardiovasc Med 2021; 8:779503. [PMID: 34869693 PMCID: PMC8635698 DOI: 10.3389/fcvm.2021.779503] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 10/26/2021] [Indexed: 01/06/2023] Open
Abstract
Objectives: Atrial fibrillation (AF) is characterized by an oxidative imbalance, which is associated with an increased risk of cardiovascular events (CVEs). It is unclear whether low grade endotoxemia may contribute to the impaired antioxidant status in AF patients. We investigated the relationship between circulating lipopolysaccharides (LPS) and antioxidant status in AF patients. Patients and Methods:Post-hoc analysis from the ongoing prospective observational cohort ATHERO-AF study including 907 patients. Antioxidant status was evaluated by the activity of glutathione peroxidase 3 (GPx3) and superoxide dismutase (SOD). Patients were divided into two groups to evaluate the risk of CVEs: (1) LPS below median and GPx3 above median (n = 254); (2) LPS above median and GPx3 below median (n = 263). Results: The mean age was 73.5 ± 8.3 years, and 43.1% were women. Median LPS and GPx3 were 50.0 pg/ml [interquartile range (IQR) 15–108] and 20.0 U/ml (IQR 10.0–34.0), respectively. Patients of Groups 2 were older, with a higher prevalence of heart failure. LPS above the median was associated with reduced GPx3 [Odds Ratio for LPS 1.752, 95% Confidence Interval (CI) 1.344–2.285, p < 0.001] and SOD (OR 0.525, 95%CI 0.403–0.683) activity after adjustment for CHA2DS2VASc score. In a mean follow-up of 54.0 ± 36.8 months, 118 CVEs occurred, 42 in Group 1 and 76 in Group 2 (Log-Rank test p = 0.001). At multivariable Cox regression analysis, Group 2 was associated with a higher risk of CVEs [Hazard Ratio (HR) 1.644, 95%CI 1.117–2,421, p = 0.012], along with age ≥ 75 years (HR 2.035, 95%CI 1.394–2.972, p < 0.001), diabetes (HR 1.927, 95%CI 1.280–2.900, p = 0.002), and previous cerebrovascular disease (HR 1.895, 95%CI 1.251–2.870, p = 0.003) and previous cardiovascular disease (HR 1.708, 95%CI 1.149–2.538, p = 0.008). Conclusions: Our study indicates that circulating LPS may contribute to impaired antioxidant status in patients with AF. Patients with coincidentally high LPS and reduced GPx3 activity showed the highest risk of CVEs.
Collapse
Affiliation(s)
- Danilo Menichelli
- Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Roberto Carnevale
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy.,Department of Angio-Cardio-Neurology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, Località Camerelle, Pozzilli, Italy
| | - Cristina Nocella
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy.,Department of Angio-Cardio-Neurology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, Località Camerelle, Pozzilli, Italy
| | - Vittoria Cammisotto
- Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Valentina Castellani
- Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Simona Bartimoccia
- Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Giacomo Frati
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy.,Department of Angio-Cardio-Neurology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, Località Camerelle, Pozzilli, Italy
| | - Pasquale Pignatelli
- Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy.,Mediterranea Cardiocentro, Naples, Italy
| | - Daniele Pastori
- Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
14
|
Singh SB, Lin HC. Role of Intestinal Alkaline Phosphatase in Innate Immunity. Biomolecules 2021; 11:biom11121784. [PMID: 34944428 PMCID: PMC8698947 DOI: 10.3390/biom11121784] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 11/23/2021] [Accepted: 11/23/2021] [Indexed: 12/14/2022] Open
Abstract
Intestinal alkaline phosphatase (IAP) is a multi-functional protein that has been demonstrated to primarily protect the gut. The role of IAP in maintaining intestinal homeostasis is underscored by the observation that IAP expression is defective in many gastrointestinal-related disorders such as inflammatory bowel disease IBD, necrotizing enterocolitis, and metabolic syndrome and that exogenous IAP supplementation improves the outcomes associated with these disorders. Additionally, studies using transgenic IAP-knock out (IAP-KO) mouse models further support the importance of the defensive role of IAP in the intestine. Supplementation of exogenous IAP and cellular overexpression of IAP have also been used in vitro to dissect out the downstream mechanisms of this protein in mammalian cell lines. Some of the innate immune functions of IAP include lipopolysaccharide (LPS) detoxification, protection of gut barrier integrity, regulation of gut microbial communities and its anti-inflammatory roles. A novel function of IAP recently identified is the induction of autophagy. Due to its critical role in the gut physiology and its excellent safety profile, IAP has been used in phase 2a clinical trials for treating conditions such as sepsis-associated acute kidney injury. Many excellent reviews discuss the role of IAP in physiology and pathophysiology and here we extend these to include recent updates on this important host defense protein and discuss its role in innate immunity via its effects on bacteria as well as on host cells. We will also discuss the relationship between IAP and autophagy and how these two pathways may act in concert to protect the gut.
Collapse
Affiliation(s)
- Sudha B. Singh
- Biomedical Research Institute of New Mexico, Albuquerque, NM 87108, USA;
| | - Henry C. Lin
- Medicine Service, New Mexico VA Health Care System, Albuquerque, NM 87108, USA
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of New Mexico, Albuquerque, NM 87131, USA
- Correspondence:
| |
Collapse
|