1
|
Ezenabor EH, Adeyemi AA, Adeyemi OS. Gut Microbiota and Metabolic Syndrome: Relationships and Opportunities for New Therapeutic Strategies. SCIENTIFICA 2024; 2024:4222083. [PMID: 39041052 PMCID: PMC11262881 DOI: 10.1155/2024/4222083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 05/10/2024] [Accepted: 07/04/2024] [Indexed: 07/24/2024]
Abstract
Since its discovery, numerous studies have shown the role of the microbiota in well-being and disease. The gut microbiota represents an essential factor that plays a multidirectional role that affects not just the gut but also other parts of the body, including the brain, endocrine system, humoral system, immune system, and metabolic pathways, as well as host-microbiome interactions. Through a comprehensive analysis of existing literature using the desktop research methodology, this review elucidates the mechanisms by which gut microbiota dysbiosis contributes to metabolic dysfunction, including obesity, dyslipidaemia, hypertension, atherosclerosis, hyperuricemia, and hyperglycaemia. Furthermore, it examines the bidirectional communication pathways between gut microbiota and host metabolism, highlighting the role of microbial-derived metabolites, immune modulation, and gut barrier integrity in shaping metabolic homeostasis. Importantly, the review identifies promising therapeutic strategies targeting the gut microbiota as potential interventions for metabolic syndrome, including probiotics, prebiotics, symbiotics, dietary modifications, and faecal microbiota transplantation. By delineating the bidirectional interactions between gut microbiota and metabolic syndrome, the review not only advances our understanding of disease pathophysiology but also underscores the potential for innovative microbiota-based interventions to mitigate the global burden of metabolic syndrome and its associated complications.
Collapse
Affiliation(s)
- Emmanuel Henry Ezenabor
- Department of BiochemistryMedicinal Biochemistry, Nanomedicine & Toxicology LaboratoryBowen University, Iwo 232102, Osun State, Nigeria
| | - Aishat Abimbola Adeyemi
- Department of BiochemistryMedicinal Biochemistry, Nanomedicine & Toxicology LaboratoryBowen University, Iwo 232102, Osun State, Nigeria
| | - Oluyomi Stephen Adeyemi
- Department of BiochemistryMedicinal Biochemistry, Nanomedicine & Toxicology LaboratoryBowen University, Iwo 232102, Osun State, Nigeria
| |
Collapse
|
2
|
Bhat A, Carranza FR, Tuckowski AM, Leiser SF. Flavin-containing monooxygenase (FMO): Beyond xenobiotics. Bioessays 2024; 46:e2400029. [PMID: 38713170 PMCID: PMC11447872 DOI: 10.1002/bies.202400029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 05/08/2024]
Abstract
Flavin-containing monooxygenases (FMOs), traditionally known for detoxifying xenobiotics, are now recognized for their involvement in endogenous metabolism. We recently discovered that an isoform of FMO, fmo-2 in Caenorhabditis elegans, alters endogenous metabolism to impact longevity and stress tolerance. Increased expression of fmo-2 in C. elegans modifies the flux through the key pathway known as One Carbon Metabolism (OCM). This modified flux results in a decrease in the ratio of S-adenosyl-methionine (SAM) to S-adenosyl-homocysteine (SAH), consequently diminishing methylation capacity. Here we discuss how FMO-2-mediated formate production during tryptophan metabolism may serve as a trigger for changing the flux in OCM. We suggest formate bridges tryptophan and OCM, altering metabolic flux away from methylation during fmo-2 overexpression. Additionally, we highlight how these metabolic results intersect with the mTOR and AMPK pathways, in addition to mitochondrial metabolism. In conclusion, the goal of this essay is to bring attention to the central role of FMO enzymes but lack of understanding of their mechanisms. We justify a call for a deeper understanding of FMO enzyme's role in metabolic rewiring through tryptophan/formate or other yet unidentified substrates. Additionally, we emphasize the identification of novel drugs and microbes to induce FMO activity and extend lifespan.
Collapse
Affiliation(s)
- Ajay Bhat
- Molecular & Integrative Physiology Department, University of Michigan, Ann Arbor, Michigan, USA
| | - Faith R Carranza
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, Michigan, USA
| | - Angela M Tuckowski
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, Michigan, USA
| | - Scott F Leiser
- Molecular & Integrative Physiology Department, University of Michigan, Ann Arbor, Michigan, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
3
|
Xing H, Xu P, Ma Y, Li T, Zhang Y, Ding X, Liu L, Keerman M, Niu Q. TFEB ameliorates DEHP-induced neurotoxicity by activating GAL3/TRIM16 axis dependent lysophagy and alleviating lysosomal dysfunction. ENVIRONMENTAL TOXICOLOGY 2024; 39:3779-3789. [PMID: 38488668 DOI: 10.1002/tox.24221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 02/17/2024] [Accepted: 03/04/2024] [Indexed: 06/12/2024]
Abstract
Di-(2-ethylhexyl) phthalate (DEHP) is a commonly used plasticizer with known neurotoxic effects. However, the specific mechanism underlying this neurotoxicity remains unclear. This study aimed to investigate the role of lysosomal function and lysophagy in DEHP-induced neurotoxicity, with a particular focus on the regulatory role of Transcription factor EB (TFEB). To achieve this, we utilized in vitro models of DEHP-exposed SH-SY5Y cells and HT22 cells. Our findings revealed that DEHP exposure led to lysosomal damage and dysfunction. Moreover, we observed impaired autophagic degradation, characterized by elevated levels of LC3II and p62. DEHP treatment downregulated the expression of TFEB, GAL3, and TRIM16, while upregulating the expression of PARP. This led to the inhibition of GAL3/TRIM16 axis dependent lysophagy and ultimately excessive apoptosis in neuronal cells. Importantly, TFEB overexpression alleviated lysosomal dysfunction, activated lysophagy, and mitigated DEHP-induced apoptosis. Overall, our results suggest that DEHP induces not only lysosomal dysfunction, but also inhibits lysophagy through the suppression of GAL3/TRIM16 axis. Consequently, impaired clearance of damaged lysosomes occurs, culminating in neuronal apoptosis. Taken together, our findings highlight the critical role of TFEB in regulating lysophagy and lysosomal function. Furthermore, TFEB may serve as a potential therapeutic target for mitigating DEHP-induced neuronal toxicity.
Collapse
Affiliation(s)
- Hengrui Xing
- Department of Preventive Medicine, School of Medicine, Shihezi University, Shihezi, People's Republic of China
- Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, Shihezi, People's Republic of China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi, People's Republic of China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases (First Affiliated Hospital, School of Medicine, Shihezi University), Shihezi, People's Republic of China
| | - Panpan Xu
- Department of Preventive Medicine, School of Medicine, Shihezi University, Shihezi, People's Republic of China
- Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, Shihezi, People's Republic of China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi, People's Republic of China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases (First Affiliated Hospital, School of Medicine, Shihezi University), Shihezi, People's Republic of China
| | - Yue Ma
- Department of Preventive Medicine, School of Medicine, Shihezi University, Shihezi, People's Republic of China
- Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, Shihezi, People's Republic of China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi, People's Republic of China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases (First Affiliated Hospital, School of Medicine, Shihezi University), Shihezi, People's Republic of China
| | - Tingting Li
- Department of Preventive Medicine, School of Medicine, Shihezi University, Shihezi, People's Republic of China
- Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, Shihezi, People's Republic of China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi, People's Republic of China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases (First Affiliated Hospital, School of Medicine, Shihezi University), Shihezi, People's Republic of China
| | - Yue Zhang
- Department of Preventive Medicine, School of Medicine, Shihezi University, Shihezi, People's Republic of China
- Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, Shihezi, People's Republic of China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi, People's Republic of China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases (First Affiliated Hospital, School of Medicine, Shihezi University), Shihezi, People's Republic of China
| | - Xueman Ding
- Department of Preventive Medicine, School of Medicine, Shihezi University, Shihezi, People's Republic of China
- Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, Shihezi, People's Republic of China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi, People's Republic of China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases (First Affiliated Hospital, School of Medicine, Shihezi University), Shihezi, People's Republic of China
| | - Li Liu
- Department of Preventive Medicine, School of Medicine, Shihezi University, Shihezi, People's Republic of China
- Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, Shihezi, People's Republic of China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi, People's Republic of China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases (First Affiliated Hospital, School of Medicine, Shihezi University), Shihezi, People's Republic of China
| | - Mulatibieke Keerman
- Department of Preventive Medicine, School of Medicine, Shihezi University, Shihezi, People's Republic of China
- Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, Shihezi, People's Republic of China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi, People's Republic of China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases (First Affiliated Hospital, School of Medicine, Shihezi University), Shihezi, People's Republic of China
| | - Qiang Niu
- Department of Preventive Medicine, School of Medicine, Shihezi University, Shihezi, People's Republic of China
- Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, Shihezi, People's Republic of China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi, People's Republic of China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases (First Affiliated Hospital, School of Medicine, Shihezi University), Shihezi, People's Republic of China
| |
Collapse
|
4
|
Chen Y, Zhang J, Li F. Inhibitory role of remifentanil in hepatic ischemia-reperfusion injury through activation of Fmol/Parkin signaling pathway: A study based on network pharmacology analysis and high-throughput sequencing. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155300. [PMID: 38518639 DOI: 10.1016/j.phymed.2023.155300] [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: 05/05/2023] [Revised: 11/13/2023] [Accepted: 12/17/2023] [Indexed: 03/24/2024]
Abstract
BACKGROUND This study was conducted to elucidate the critical molecular pathways underlying the protective effects of remifentanil against hepatic ischemia-reperfusion injury in rats. Our approach integrated network pharmacology analysis with high-throughput sequencing to achieve a comprehensive understanding of the mechanisms involved. STUDY DESIGN/METHODS The study utilized GSE24430 gene expression data from GEO to investigate remifentanil's impact on Hepatic Ischemia-Reperfusion Injury in rats. Weighted Correlation Network Analysis (WGCNA) was employed to pinpoint crucial genes and identify modules of co-expressed genes. Differential analysis with the "Limma" package revealed genes differentially expressed in IRI vs. control groups. PubChem and PharmMapper provided target genes affected by remifentanil. Protein-protein interaction networks were constructed via GeneCards and STRING. Functional analysis pinpointed core genes involved in remifentanil's IRI alleviation. IRI rat models were established, and hepatic injury indicators, liver structure via H&E staining, autophagosome counts via electron microscopy, and gene/protein expression via RT-qPCR and Western blot were assessed. High-throughput sequencing analyzed molecular pathways affected by varying remifentanil doses in IRI rats. RESULTS In the study, we discovered four primary co-expression modules associated with hepatic IRI, and the grey module exhibited the highest correlation with hepatic IRI.A total of sixty-eight genes that were differentially expressed were found to have a connection with hepatic IRI.Network pharmacology analysis found that remifentanil may alleviate hepatic IRI through Fmol.found that the Fmol/Parkin signaling pathway may alleviate hepatic IRI via Additionally, the database autophagy. The established hepatic IRI rat models further confirmed the above findings. CONCLUSION Our study established that remifentanil triggers the Fmol/Parkin signaling cascade, amplifying the expression levels of Fmol and Parkin. This process culminates in the activation of autophagy within hepatic cells, ultimately alleviating hepatic ischemia-reperfusion injury (IRI).
Collapse
Affiliation(s)
- Yisi Chen
- Department of Anesthesiology, Huai'an First People's Hospital, Huai'an 223300, China.
| | - Jun Zhang
- Department of Anesthesiology, Huai'an First People's Hospital, Huai'an 223300, China
| | - Fayin Li
- Department of Anesthesiology, Huai'an First People's Hospital, Huai'an 223300, China
| |
Collapse
|
5
|
Ganouna-Cohen G, Marcouiller F, Blachot-Minassian B, Demarest M, Beauparlant CJ, Droit A, Belaidi E, Bairam A, Joseph V. Loss of testosterone induces postprandial insulin resistance and increases the expression of the hepatic antioxidant flavin-containing monooxygenases in mice exposed to intermittent hypoxia. Acta Physiol (Oxf) 2024; 240:e14089. [PMID: 38230898 DOI: 10.1111/apha.14089] [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: 06/25/2023] [Revised: 11/29/2023] [Accepted: 01/01/2024] [Indexed: 01/18/2024]
Abstract
AIM We tested the hypothesis that low testosterone alters the effects of intermittent hypoxia (IH) on glucose homeostasis, hepatic oxidative stress, and transcriptomic profile in male mice. METHODS We used sham-operated or orchiectomized (ORX) mice exposed to normoxia (Nx) or IH for 2 weeks. We performed fasting insulin and glucose tolerance tests and assessed fasting and postprandial insulin resistance with the HOMA-IR. The activity of hepatic prooxidant (NADPH oxidase-NOX), antioxidant enzymes (superoxide dismutase, catalase, and glutathione peroxidase-SOD, Cat, GPx), lipid peroxidation (MDA concentration), and the total concentration of glutathione (GSH) were measured under postprandial conditions. mRNA sequencing and pathway enrichment analyses were used to identify hepatic genes underlying the interactions between IH and testosterone. RESULTS In Sham mice, IH improves fasting insulin sensitivity and glucose tolerance, while there are no effects of IH in ORX mice. In ORX mice, IH induces postprandial hyperinsulinemia, insulin resistance, and a prooxidant profile of enzyme activity (low SOD activity) without altering hepatic MDA and GSH content. ORX and IH altered the expression of genes involved in oxidoreductase activities, cytochromes-dependent pathways, and glutathione metabolism. Among the genes upregulated in ORX-IH mice, the flavin-containing monooxygenases (FMO) are particularly relevant since these are potent hepatic antioxidants that could help prevent overt oxidative stress in ORX-IH mice. CONCLUSION Low levels of testosterone in male mice exposed to IH induce post-prandial hyperinsulinemia and insulin resistance and determine the mechanisms by which the liver handles IH-induced oxidative stress.
Collapse
Affiliation(s)
- Gauthier Ganouna-Cohen
- Département de Pédiatrie, Faculté de Médecine, Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, Quebec, Canada
| | - François Marcouiller
- Département de Pédiatrie, Faculté de Médecine, Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, Quebec, Canada
| | - Britanny Blachot-Minassian
- Département de Pédiatrie, Faculté de Médecine, Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, Quebec, Canada
- HP2, INSERM, U1300, Université Grenoble Alpes, Grenoble, France
| | - Maud Demarest
- Département de Pédiatrie, Faculté de Médecine, Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, Quebec, Canada
| | - Charles Joly Beauparlant
- Département de Médecine Moléculaire, Faculté de Médecine, Centre de Recherche du Centre Hospitalo-Universitaire de Québec, Québec, Quebec, Canada
| | - Arnaud Droit
- Département de Médecine Moléculaire, Faculté de Médecine, Centre de Recherche du Centre Hospitalo-Universitaire de Québec, Québec, Quebec, Canada
| | - Elise Belaidi
- HP2, INSERM, U1300, Université Grenoble Alpes, Grenoble, France
- UMR5305-LBTI, CNRS, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Aida Bairam
- Département de Pédiatrie, Faculté de Médecine, Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, Quebec, Canada
| | - Vincent Joseph
- Département de Pédiatrie, Faculté de Médecine, Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, Quebec, Canada
| |
Collapse
|
6
|
Dunaevskaya SS, Sergeeva EY, Titova NM, Fefelova YA, Deulina VV. [Role of superoxide dismutase in acute pancreatitis: from antioxidant protection to gene regulation]. Khirurgiia (Mosk) 2024:112-117. [PMID: 38634592 DOI: 10.17116/hirurgia2024041112] [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] [Indexed: 04/19/2024]
Abstract
We present modern data on the role of superoxide dismutase (SOD) in antioxidant protection and gene regulation in acute pancreatitis. Antioxidant enzymes are essential in pathogenesis of numerous diseases. SOD is one of the key enzymes of antioxidant system. In this review, we analyzed activity of this enzyme depending on various factors, mechanisms and role in physiological and pathological processes, in particular, acute pancreatitis. SOD is significantly less active in patients with severe acute pancreatitis accompanied by renal failure, severe circulatory disorders and high mortality. There are some SOD gene polymorphisms, in particular, acute destructive pancreatitis R213G, contributing to acute inflammation. Thus, SOD is not only one of the key antioxidant enzymes, but also potential transcription factor regulating activity of signaling pathways. These aspects can underlie new therapies for diseases.
Collapse
Affiliation(s)
- S S Dunaevskaya
- Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, Russia
| | - E Yu Sergeeva
- Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, Russia
| | - N M Titova
- Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, Russia
| | - Yu A Fefelova
- Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, Russia
| | - V V Deulina
- Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, Russia
| |
Collapse
|
7
|
Takenoya F, Shibato J, Yamashita M, Kimura A, Hirako S, Chiba Y, Nonaka N, Shioda S, Rakwal R. Transcriptomic (DNA Microarray) and Metabolome (LC-TOF-MS) Analyses of the Liver in High-Fat Diet Mice after Intranasal Administration of GALP (Galanin-like Peptide). Int J Mol Sci 2023; 24:15825. [PMID: 37958806 PMCID: PMC10648535 DOI: 10.3390/ijms242115825] [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: 09/19/2023] [Revised: 10/20/2023] [Accepted: 10/28/2023] [Indexed: 11/15/2023] Open
Abstract
The aim of this research was to test the efficacy and potential clinical application of intranasal administration of galanin-like peptide (GALP) as an anti-obesity treatment under the hypothesis that GALP prevents obesity in mice fed a high-fat diet (HFD). Focusing on the mechanism of regulation of lipid metabolism in peripheral tissues via the autonomic nervous system, we confirmed that, compared with a control (saline), intranasally administered GALP prevented further body weight gain in diet-induced obesity (DIO) mice with continued access to an HFD. Using an omics-based approach, we identified several genes and metabolites in the liver tissue of DIO mice that were altered by the administration of intranasal GALP. We used whole-genome DNA microarray and metabolomics analyses to determine the anti-obesity effects of intranasal GALP in DIO mice fed an HFD. Transcriptomic profiling revealed the upregulation of flavin-containing dimethylaniline monooxygenase 3 (Fmo3), metallothionein 1 and 2 (Mt1 and Mt2, respectively), and the Aldh1a3, Defa3, and Defa20 genes. Analysis using the DAVID tool showed that intranasal GALP enhanced gene expression related to fatty acid elongation and unsaturated fatty acid synthesis and downregulated gene expression related to lipid and cholesterol synthesis, fat absorption, bile uptake, and excretion. Metabolite analysis revealed increased levels of coenzyme Q10 and oleoylethanolamide in the liver tissue, increased levels of deoxycholic acid (DCA) and taurocholic acid (TCA) in the bile acids, increased levels of taurochenodeoxycholic acid (TCDCA), and decreased levels of ursodeoxycholic acid (UDCA). In conclusion, intranasal GALP administration alleviated weight gain in obese mice fed an HFD via mechanisms involving antioxidant, anti-inflammatory, and fatty acid metabolism effects and genetic alterations. The gene expression data are publicly available at NCBI GSE243376.
Collapse
Affiliation(s)
- Fumiko Takenoya
- Department of Sport Sciences, Hoshi University School of Pharmacy and Pharmaceutical Sciences, Tokyo 142-8501, Japan; (F.T.); (M.Y.); (A.K.)
| | - Junko Shibato
- Department of Functional Morphology, Shonan University of Medical Sciences, Kanagawa 244-0806, Japan; (J.S.); (S.S.)
| | - Michio Yamashita
- Department of Sport Sciences, Hoshi University School of Pharmacy and Pharmaceutical Sciences, Tokyo 142-8501, Japan; (F.T.); (M.Y.); (A.K.)
| | - Ai Kimura
- Department of Sport Sciences, Hoshi University School of Pharmacy and Pharmaceutical Sciences, Tokyo 142-8501, Japan; (F.T.); (M.Y.); (A.K.)
| | - Satoshi Hirako
- Department of Health and Nutrition, University of Human Arts and Sciences, Saitama 339-8539, Japan;
| | - Yoshihiko Chiba
- Laboratory of Molecular Biology and Physiology, School of Pharmacy, Hoshi University, Tokyo 142-8501, Japan;
| | - Naoko Nonaka
- Department of Oral Anatomy and Developmental Biology, Showa University School of Dentistry, Tokyo 142-8555, Japan;
| | - Seiji Shioda
- Department of Functional Morphology, Shonan University of Medical Sciences, Kanagawa 244-0806, Japan; (J.S.); (S.S.)
| | - Randeep Rakwal
- Institute of Health and Sport Sciences, University of Tsukuba, Tsukuba 305-8574, Japan
| |
Collapse
|
8
|
Fu S, Tang X, Xu Y, Song X, Qian X, Hu Y, Zhang M. Analysis of the Potential Relationship between Aging and Pulmonary Fibrosis Based on Transcriptome. LIFE (BASEL, SWITZERLAND) 2022; 12:life12121961. [PMID: 36556326 PMCID: PMC9788318 DOI: 10.3390/life12121961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/19/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is an age-related interstitial lung disease with a high incidence in the elderly. Although many reports have shown that senescence can initiate pulmonary fibrosis, the relationship between aging and pulmonary fibrosis has not been explained systematically. In our study, young and old rats were intratracheally instilled with bleomycin (1 mg/kg), and the basic pathological indexes were determined using a commercial kit, hematoxylin, and eosin (H&E) and Masson's Trichrome staining, immunohistochemistry, immunohistofluorescence, and q-PCR. Then, the lung tissues of rats were sequenced by next-generation sequencing for transcriptome analysis. Bioinformatics was performed to analyze the possible differences in the mechanism of pulmonary fibrosis between aged and young rats. Finally, the related cytokines were determined by q-PCR and ELISA. The results indicate that pulmonary fibrosis in old rats is more serious than that in young rats under the same conditions. Additionally, transcriptomic and bioinformatics analysis with experimental validation indicate that the differences in pulmonary fibrosis between old and young rats are mainly related to the differential expression of cytokines, extracellular matrix (ECM), and other important signaling pathways. In conclusion, aging mainly affects pulmonary fibrosis through the ECM-receptor interaction, immune response, and chemokines.
Collapse
Affiliation(s)
- San Fu
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Xiaoyan Tang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yiming Xu
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Xianrui Song
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Xiuhui Qian
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yingying Hu
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Mian Zhang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Correspondence: ; Tel.: +86-25-8618-513
| |
Collapse
|
9
|
Yang Z, Stemmer PM, Petriello MC. Proteomics-Based Identification of Interaction Partners of the Xenobiotic Detoxification Enzyme FMO3 Reveals Involvement in Urea Cycle. TOXICS 2022; 10:60. [PMID: 35202247 PMCID: PMC8877285 DOI: 10.3390/toxics10020060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/18/2022] [Accepted: 01/25/2022] [Indexed: 02/06/2023]
Abstract
The hepatic xenobiotic metabolizing enzyme flavin-containing monooxygenase 3 (FMO3) has been implicated in the development of cardiometabolic disease primarily due to its enzymatic product trimethylamine-N oxide (TMAO), which has recently been shown to be associated with multiple chronic diseases, including kidney and coronary artery diseases. Although TMAO may have causative roles as a pro-inflammatory mediator, the possibility for roles in metabolic disease for FMO3, irrespective of TMAO formation, does exist. We hypothesized that FMO3 may interact with other proteins known to be involved in cardiometabolic diseases and that modulating the expression of FMO3 may impact on these interaction partners. Here, we combine a co-immunoprecipitation strategy coupled to unbiased proteomic workflow to report a novel protein:protein interaction network for FMO3. We identified 51 FMO3 protein interaction partners, and through gene ontology analysis, have identified urea cycle as an enriched pathway. Using mice deficient in FMO3 on two separate backgrounds, we validated and further investigated expressional and functional associations between FMO3 and the identified urea cycle genes. FMO3-deficient mice showed hepatic overexpression of carbamoylphosphate synthetase (CPS1), the rate-limiting gene of urea cycle, and increased hepatic urea levels, especially in mice of FVB (Friend leukemia virus B strain) background. Finally, overexpression of FMO3 in murine AML12 hepatocytes led to downregulation of CPS1. Although there is past literature linking TMAO to urea cycle, this is the first published work showing that FMO3 and CPS1 may directly interact, implicating a role for FMO3 in chronic kidney disease irrespective of TMAO formation.
Collapse
Affiliation(s)
- Zhao Yang
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI 48202, USA; (Z.Y.); (P.M.S.)
| | - Paul M. Stemmer
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI 48202, USA; (Z.Y.); (P.M.S.)
- Department of Pharmaceutical Sciences, College of Pharmacy, Wayne State University, Detroit, MI 48202, USA
| | - Michael C. Petriello
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI 48202, USA; (Z.Y.); (P.M.S.)
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI 48202, USA
| |
Collapse
|
10
|
Lei L, Zhao N, Zhang L, Chen J, Liu X, Piao S. Gut microbiota is a potential goalkeeper of dyslipidemia. Front Endocrinol (Lausanne) 2022; 13:950826. [PMID: 36176475 PMCID: PMC9513062 DOI: 10.3389/fendo.2022.950826] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 08/26/2022] [Indexed: 11/30/2022] Open
Abstract
Dyslipidemia, as a common metabolic disease, could cause atherosclerosis, coronary heart disease, stroke and other cardio-cerebrovascular diseases. It is mainly caused by the interaction of genetic and environmental factors and its incidence has increased for several years. A large number of studies have shown that gut microbiota disorder is related to the development of dyslipidemia closely. Especially its metabolites such as short-chain fatty acids, bile acids and trimethylamine N-oxide affect dyslipidemia by regulating cholesterol balance. In this paper, we systematically reviewed the literature and used knowledge graphs to analyze the research trends and characteristics of dyslipidemia mediated by gut microbiota, revealing that the interaction between diet and gut microbiota leads to dyslipidemia as one of the main factors. In addition, starting from the destruction of the dynamic balance between gut microbiota and host caused by dyslipidemia, we systematically summarize the molecular mechanism of gut microbiota regulating dyslipidemia and provide a theoretical basis for the treatment of dyslipidemia by targeting the gut microbiota.
Collapse
Affiliation(s)
- Lirong Lei
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou, China
| | - Ning Zhao
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou, China
| | - Lei Zhang
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou, China
| | - Jiamei Chen
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou, China
| | - Xiaomin Liu
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou, China
| | - Shenghua Piao
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou, China
- *Correspondence: Shenghua Piao,
| |
Collapse
|
11
|
Saito K, Ito M, Chiba T, Jia H, Kato H. A Comparison of Gene Expression Profiles of Rat Tissues after Mild and Short-Term Calorie Restrictions. Nutrients 2021; 13:2277. [PMID: 34209243 PMCID: PMC8308279 DOI: 10.3390/nu13072277] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/22/2021] [Accepted: 06/24/2021] [Indexed: 12/14/2022] Open
Abstract
Many studies have shown the beneficial effects of calorie restriction (CR) on rodents' aging; however, the molecular mechanism explaining these beneficial effects is still not fully understood. Previously, we conducted transcriptomic analysis on rat liver with short-term and mild-to-moderate CR to elucidate its early response to such diet. Here, we expanded transcriptome analysis to muscle, adipose tissue, intestine, and brain and compared the gene expression profiles of these multiple organs and of our previous dataset. Several altered gene expressions were found, some of which known to be related to CR. Notably, the commonly regulated genes by CR include nicotinamide phosphoribosyltransferase and heat shock protein 90, which are involved in declining the aging process and thus potential therapeutic targets for aging-related diseases. The data obtained here provide information on early response markers and key mediators of the CR-induced delay in aging as well as on age-associated pathological changes in mammals.
Collapse
Affiliation(s)
- Kenji Saito
- Health Nutrition, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan; (K.S.); (M.I.); (H.J.)
| | - Maiko Ito
- Health Nutrition, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan; (K.S.); (M.I.); (H.J.)
- Biomedical Gerontology Laboratory, Faculty of Human Sciences, Waseda University, 2-579-1 Mikajima, Tokorozawa, Saitama 359-1164, Japan;
| | - Takuya Chiba
- Biomedical Gerontology Laboratory, Faculty of Human Sciences, Waseda University, 2-579-1 Mikajima, Tokorozawa, Saitama 359-1164, Japan;
| | - Huijuan Jia
- Health Nutrition, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan; (K.S.); (M.I.); (H.J.)
| | - Hisanori Kato
- Health Nutrition, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan; (K.S.); (M.I.); (H.J.)
| |
Collapse
|
12
|
Guo D, Cheng L, Shen Y, Li W, Li Q, Zhong Y, Miao Y. 6-Bromoindirubin-3'-oxime (6BIO) prevents myocardium from aging by inducing autophagy. Aging (Albany NY) 2020; 12:26047-26062. [PMID: 33401248 PMCID: PMC7803501 DOI: 10.18632/aging.202253] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 10/01/2020] [Indexed: 12/18/2022]
Abstract
6-Bromoindirubin-3’-oxime (6BIO) is a novel small molecule that exerts positive effects on several age-related alterations. However, the anti-aging effects of 6BIO on the aging heart remain unknown. Herein, we aim to investigate the effects of 6BIO on the myocardium and its underlying mechanism in vivo and vitro. Following 6BIO treatment, an increased p53 contents, a reduced p16 and β-gal levels, and attenuation of cardiac fibrosis were observed, suggesting 6BIO retarded aging of cardiomyocytes. As observed, 6BIO reduced p62 contents, elevated the levels of Beclin-1 and the ratio of LC3II/I, indicating the induction of autophagy, while the reduction of the accumulation of ROS indicated 6BIO alleviated oxidative stress. In addition, 6BIO treatment inhibited both GSK3β signaling and mTOR signaling. 6BIO might be a promising agent for preventing myocardium from aging.
Collapse
Affiliation(s)
- Donghao Guo
- Department of Geriatrics, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China.,Division of Cardiology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong
| | - Lizhen Cheng
- Department of Geriatrics, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Yun Shen
- Department of Geriatrics, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Wei Li
- Department of Geriatrics, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Qinjie Li
- Department of Geriatrics, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Yuan Zhong
- Department of Geriatrics, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Ya Miao
- Department of Geriatrics, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| |
Collapse
|
13
|
Abstract
We critically review potential involvement of trimethylamine N-oxide (TMAO) as a link between diet, the gut microbiota and CVD. Generated primarily from dietary choline and carnitine by gut bacteria and hepatic flavin-containing mono-oxygenase (FMO) activity, TMAO could promote cardiometabolic disease when chronically elevated. However, control of circulating TMAO is poorly understood, and diet, age, body mass, sex hormones, renal clearance, FMO3 expression and genetic background may explain as little as 25 % of TMAO variance. The basis of elevations with obesity, diabetes, atherosclerosis or CHD is similarly ill-defined, although gut microbiota profiles/remodelling appear critical. Elevated TMAO could promote CVD via inflammation, oxidative stress, scavenger receptor up-regulation, reverse cholesterol transport (RCT) inhibition, and cardiovascular dysfunction. However, concentrations influencing inflammation, scavenger receptors and RCT (≥100 µm) are only achieved in advanced heart failure or chronic kidney disease (CKD), and greatly exceed pathogenicity of <1-5 µm levels implied in some TMAO-CVD associations. There is also evidence that CVD risk is insensitive to TMAO variance beyond these levels in omnivores and vegetarians, and that major TMAO sources are cardioprotective. Assessing available evidence suggests that modest elevations in TMAO (≤10 µm) are a non-pathogenic consequence of diverse risk factors (ageing, obesity, dyslipidaemia, insulin resistance/diabetes, renal dysfunction), indirectly reflecting CVD risk without participating mechanistically. Nonetheless, TMAO may surpass a pathogenic threshold as a consequence of CVD/CKD, secondarily promoting disease progression. TMAO might thus reflect early CVD risk while providing a prognostic biomarker or secondary target in established disease, although mechanistic contributions to CVD await confirmation.
Collapse
|
14
|
Lin L, Li C, Zhang D, Yuan M, Chen CH, Li M. Synergic Effects of Berberine and Curcumin on Improving Cognitive Function in an Alzheimer's Disease Mouse Model. Neurochem Res 2020; 45:1130-1141. [PMID: 32080784 DOI: 10.1007/s11064-020-02992-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 02/07/2020] [Accepted: 02/17/2020] [Indexed: 12/17/2022]
Abstract
Alzheimer's disease (AD) is one of the most common neurodegenerative diseases, and no effective therapies have been found to prevent or cure AD to date. Berberine and curcumin are extracts from traditional Chinese herbs that have a long history of clinical benefits for AD. Here, using a transgenic AD mouse model, we found that the combined berberine and curcumin treatment had a much better effect on improving the cognitive function of mice than the single-drug treatment, suggesting synergic effects of the combined berberine and curcumin treatment. In addition, we found that the combined berberine and curcumin treatment had significant synergic effects on reducing soluble amyloid-β-peptide(1-42) production. Furthermore, the combination treatment also had remarkable synergic effects on decreasing inflammatory responses and oxidative stress in both the cortex and hippocampus of AD mice. We also found that the combination treatment performed much better than the single drugs in reducing the APP and BACE1 levels and increasing AMPKα phosphorylation and cell autophagy, which might be the underlying mechanism of the synergic effects. Taken together, the result of this study reveal the synergic effects and potential underlying mechanisms of the combined berberine and curcumin treatment in improving the symptoms of AD in mice. This study sheds light on a new strategy for exploring new phytotherapies for AD and also emphasizes that more research should focus on the synergic effects of herbal drugs in the future.
Collapse
Affiliation(s)
- Lin Lin
- Collaborative Innovation Center of Sichuan for Elderly Care and Health, Chengdu Medical College, Chengdu, 610500, Sichuan, China
| | - Cheng Li
- Department of Public Health, Chengdu Medical College, Chengdu, 610500, Sichuan, China
| | - Deyi Zhang
- Department of Anesthesiology, Mianyang People's Hospital, Mianyang, 621000, Sichuan, China
| | - Mingxiang Yuan
- Department of Gynaecology and Obstetrics, Mianyang People's Hospital, Mianyang, 621000, Sichuan, China
| | - Chun-Hai Chen
- Department of Occupational Health, Amy Medical University, Chongqing, 400038, China.
| | - Maoquan Li
- Affiliated Traditional Chinese Medicine Hospital of Chengdu Medical College, Chengdu, 610300, Sichuan, China. .,Chengdu Qingbaijiang District Traditional Chinese Medicine Hospital, Chengdu, 610300, Sichuan, China. .,Department of Public Health, Chengdu Medical College, Chengdu, 610500, Sichuan, China.
| |
Collapse
|