1
|
Chae YR, Lee HB, Lee YR, Yoo G, Lee E, Park M, Choi SY, Park HY. Ameliorating effects of Orostachys japonica against high-fat diet-induced obesity and gut dysbiosis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 333:118443. [PMID: 38909828 DOI: 10.1016/j.jep.2024.118443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 06/03/2024] [Accepted: 06/05/2024] [Indexed: 06/25/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Orostachys japonica (rock pine) has been used as a folk remedy to treat inflammation, hepatitis, and cancer in East Asia. AIM OF THE STUDY The aim of this study was to investigate the effect of rock pine extract (RPE) on high-fat diet-induced obesity in mice and to examine its effects on gut dysbiosis. MATERIALS AND METHODS The characteristic compound of RPE, kaempferol-3-O-rutinoside, was quantified using high-performance liquid chromatography. The prebiotic potential of RPE was evaluated by assessing the prebiotic activity score obtained using four prebiotic strains and high-fat (HF)-induced obesity C57BL/6 mice model. Analysis included examining the lipid metabolism and inflammatory proteins and evaluating the changes in gut permeability and metabolites to elucidate the potential signaling pathways involved. RESULTS In vitro, RPE enhanced the proliferation of beneficial probiotic strains, including Lactiplantibacillus and Bifidobacterium. HF-induced model showed that the administration of 100 mg/kg/day of RPE for 8 weeks significantly (p < 0.05) reduced the body weight, serum lipid levels, and insulin resistance, which were associated with notable changes in lipid metabolism and inflammation-related markers. CONCLUSIONS Our results demonstrate that rock pine consumption could mitigate obesity and metabolic endotoxemia in HF-fed mice through enhancing intestinal environment.
Collapse
Affiliation(s)
- Yu-Rim Chae
- Food Functionality Research Division, Korea Food Research Institute, Wanju, 55365, Republic of Korea.
| | - Hye-Bin Lee
- Food Functionality Research Division, Korea Food Research Institute, Wanju, 55365, Republic of Korea.
| | - Yu Ra Lee
- Food Functionality Research Division, Korea Food Research Institute, Wanju, 55365, Republic of Korea.
| | - Guijae Yoo
- Food Functionality Research Division, Korea Food Research Institute, Wanju, 55365, Republic of Korea.
| | - Eunjung Lee
- Food Functionality Research Division, Korea Food Research Institute, Wanju, 55365, Republic of Korea; Department of Food Biotechnology, Korea National University of Science and Technology, Daejeon, 34113, Republic of Korea.
| | - Miri Park
- Food Functionality Research Division, Korea Food Research Institute, Wanju, 55365, Republic of Korea.
| | - Sang Yoon Choi
- Food Functionality Research Division, Korea Food Research Institute, Wanju, 55365, Republic of Korea.
| | - Ho-Young Park
- Food Functionality Research Division, Korea Food Research Institute, Wanju, 55365, Republic of Korea; Department of Food Biotechnology, Korea National University of Science and Technology, Daejeon, 34113, Republic of Korea.
| |
Collapse
|
2
|
Wang K, Dang X, Wang Y, Yang Q, Zhang T, Yang P, Yuan L, Xu R, Dang Y, Nan Y. Qianggu concentrate: unlocking bone protection power via antioxidative SIRT1/NRF2/HO-1 pathways in type 2 diabetic osteoporosis. Front Pharmacol 2024; 15:1426767. [PMID: 39175549 PMCID: PMC11338786 DOI: 10.3389/fphar.2024.1426767] [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: 05/02/2024] [Accepted: 07/26/2024] [Indexed: 08/24/2024] Open
Abstract
Background Qianggu Concentrate (QGHJ), a traditional Chinese medicine, is extensively used to treat Type 2 Diabetic Osteoporosis (T2DOP). Despite its widespread use, research on its therapeutic mechanisms within T2DOP is notably scarce. Objective To explore QGHJ's osteoprotection in T2DOP rats and BMSCs, focusing on the antioxidant activation of SIRT1/NRF2/HO-1 and NRF2 nuclear migration. Methods QGHJ constituent analysis was performed using UPLC-HRMS. Safety, bone-health efficacy, and glucose metabolic effects in T2DOP rats were evaluated via general condition assessments, biomarker profiling, micro-CT, biomechanics, staining methods, and ELISA, supplemented by RT-qPCR and Western blot. BMSCs' responses to QGHJ under oxidative stress, including viability, apoptosis, and osteogenic differentiation, were determined using CCK-8, flow cytometry, ALP/ARS staining, and molecular techniques. The modulation of the SIRT1/NRF2/HO-1 pathway by QGHJ was explored through oxidative stress biomarkers, immunofluorescence, and Western blot assays. Results UPLC-HRMS identified flavonoids, monoterpenes, and isoflavones as QGHJ's key compounds. In vivo, QGHJ proved safe and effective for T2DOP rats, enhancing bone mineral density, microenvironment, and biomechanical properties without impairing vital organs. It modulated bone markers PINP, TRACP 5b, RUNX2 and PPARγ, favoring bone anabolism and reduced catabolism, thus optimizing bone integrity. QGHJ also regulated glycemia and mitigated insulin resistance. In vitro, it preserved BMSCs' viability amidst oxidative stress, curbed apoptosis, and fostered osteogenesis with regulated RUNX2/PPARγ expression. Mechanistic insights revealed QGHJ activated the SIRT1/NRF2/HO-1 pathway, augmented NRF2 nuclear translocation, and enhanced the antioxidative response, promoting bone health under stress. Conclusion In T2DOP rat and BMSCs oxidative stress models, QGHJ's bone protection is anchored in its antioxidative mechanisms via the SIRT1/NRF2/HO-1 pathway activation and NRF2 nuclear translocation.
Collapse
Affiliation(s)
- Kaili Wang
- College of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan, China
| | - Xiang Dang
- Department of Endocrinology, Yinchuan Hospital of Traditional Chinese Medicine, Affiliated with Ningxia Medical University, Yinchuan, China
| | - Yanyan Wang
- Department of Endocrinology, Yinchuan Hospital of Traditional Chinese Medicine, Affiliated with Ningxia Medical University, Yinchuan, China
| | - Qing Yang
- Department of Endocrinology, Yinchuan Hospital of Traditional Chinese Medicine, Affiliated with Ningxia Medical University, Yinchuan, China
| | - Tingting Zhang
- College of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan, China
| | - Peng Yang
- College of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan, China
| | - Ling Yuan
- College of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Rongming Xu
- College of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Yuqi Dang
- Department of Endocrinology, Yinchuan Hospital of Traditional Chinese Medicine, Affiliated with Ningxia Medical University, Yinchuan, China
| | - Yi Nan
- College of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan, China
| |
Collapse
|
3
|
Lee SH, Suh JH, Heo MJ, Choi JM, Yang Y, Jung HJ, Gao Z, Yu Y, Jung SY, Kolonin MG, Cox AR, Hartig SM, Eltzschig HK, Ju C, Moore DD, Kim KH. The Hepatokine Orosomucoid 2 Mediates Beneficial Metabolic Effects of Bile Acids. Diabetes 2024; 73:701-712. [PMID: 38320268 PMCID: PMC11043061 DOI: 10.2337/db23-0520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 01/28/2024] [Indexed: 02/08/2024]
Abstract
Bile acids (BAs) are pleiotropic regulators of metabolism. Elevated levels of hepatic and circulating BAs improve energy metabolism in peripheral organs, but the precise mechanisms underlying the metabolic benefits and harm still need to be fully understood. In the current study, we identified orosomucoid 2 (ORM2) as a liver-secreted hormone (i.e., hepatokine) induced by BAs and investigated its role in BA-induced metabolic improvements in mouse models of diet-induced obesity. Contrary to our expectation, under a high-fat diet (HFD), our Orm2 knockout (Orm2-KO) exhibited a lean phenotype compared with C57BL/6J control, partly due to the increased energy expenditure. However, when challenged with a HFD supplemented with cholic acid, Orm2-KO eliminated the antiobesity effect of BAs, indicating that ORM2 governs BA-induced metabolic improvements. Moreover, hepatic ORM2 overexpression partially replicated BA effects by enhancing insulin sensitivity. Mechanistically, ORM2 suppressed interferon-γ/STAT1 activities in inguinal white adipose tissue depots, forming the basis for anti-inflammatory effects of BAs and improving glucose homeostasis. In conclusion, our study provides new insights into the molecular mechanisms of BA-induced liver-adipose cross talk through ORM2 induction. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Sung Ho Lee
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Chonnam National University, Gwangju, Korea
| | - Ji Ho Suh
- Department of Anesthesiology, Critical Care and Pain Medicine and Center for Perioperative Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX
| | - Mi Jeong Heo
- Department of Anesthesiology, Critical Care and Pain Medicine and Center for Perioperative Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX
| | - Jong Min Choi
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX
| | - Yang Yang
- Department of Anesthesiology, Critical Care and Pain Medicine and Center for Perioperative Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX
| | - Hyun-Jung Jung
- Department of Anesthesiology, Critical Care and Pain Medicine and Center for Perioperative Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX
| | - Zhanguo Gao
- The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX
| | - Yongmei Yu
- The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX
| | - Sung Yun Jung
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX
| | - Mikhail G. Kolonin
- The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX
| | - Aaron R. Cox
- The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX
| | - Sean M. Hartig
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Holger K. Eltzschig
- Department of Anesthesiology, Critical Care and Pain Medicine and Center for Perioperative Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX
| | - Cynthia Ju
- Department of Anesthesiology, Critical Care and Pain Medicine and Center for Perioperative Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX
| | - David D. Moore
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA
| | - Kang Ho Kim
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
- Department of Anesthesiology, Critical Care and Pain Medicine and Center for Perioperative Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX
| |
Collapse
|
4
|
Benvenuti M, Piazza SD, Salis A, Cecchi G, Zotti M, Scarfì S, Damonte G. A novel method for the extraction and characterization of metabolites from Basidiomycota: Pleurotus ostreatus (Jacq.) P. Kumm., 1871 as a case study. SEPARATION SCIENCE PLUS 2023; 6. [DOI: 10.1002/sscp.202300116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 09/25/2023] [Indexed: 07/04/2024]
Abstract
AbstractIn recent years, the interest in the exploitation of fungal metabolites has grown considerably, given their application in numerous sectors involving human health. However, their identification and characterization by conventional analytical approaches is generally limited to single families of molecules per method of analysis. This constitutes a limiting factor of primary importance in the study of both the metabolic pattern of a single fungal sample and the discovery of its possible applications. In this work, a reverse‐phase high‐performance liquid chromatography coupled with mass spectrometry method for the profile determination of primary and secondary metabolites produced by the oyster‐mushroom Pleurotus ostreatus (Jacq.) P. Kumm., 1871, has been developed. By using a concomitant extraction in three different polarity‐decreasing solvents, namely methanol, ethanol, and acetonitrile, this method allowed the simultaneous analysis of all extracted metabolites belonging to the widest possible range of chemical families, giving an advantage for both qualitative and quantitative determination of known and unknown compounds. The method appears to be valuable and robust for the study of complex matrices like raw fungi extract such as those of Pleurotus ostreatus cultivated on different substrates and/or exposed to multiple stressors.
Collapse
Affiliation(s)
- Mirko Benvenuti
- Department of Experimental Medicine (DIMES) University of Genova Genova Italy
| | - Simone Di Piazza
- Department of Earth, Environment and Life Sciences (DISTAV) University of Genova Genova Italy
| | - Annalisa Salis
- Department of Experimental Medicine (DIMES) University of Genova Genova Italy
| | - Grazia Cecchi
- Department of Earth, Environment and Life Sciences (DISTAV) University of Genova Genova Italy
| | - Mirca Zotti
- Department of Earth, Environment and Life Sciences (DISTAV) University of Genova Genova Italy
| | - Sonia Scarfì
- Department of Earth, Environment and Life Sciences (DISTAV) University of Genova Genova Italy
| | - Gianluca Damonte
- Department of Experimental Medicine (DIMES) University of Genova Genova Italy
| |
Collapse
|
5
|
Chen X, Chen S, Ren Q, Niu S, Pan X, Yue L, Li Z, Zhu R, Jia Z, Chen X, Zhen R, Ban J. Metabolomics Provides Insights into Renoprotective Effects of Semaglutide in Obese Mice. Drug Des Devel Ther 2022; 16:3893-3913. [PMID: 36388084 PMCID: PMC9656502 DOI: 10.2147/dddt.s383537] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 10/29/2022] [Indexed: 08/09/2024] Open
Abstract
PURPOSE Semaglutide, a new long-acting glucagon-like peptide-1 analogue, has shown benefits for renal diseases, but its direct role on kidney metabolism under obesity remains unclear. The study aims to elucidate the protective effect and metabolic modulation mechanism of semaglutide on obesity-related kidney injury. METHODS Male C57BL/6J mice were divided into control and obesity groups. Mice in the obesity group had a high-fat diet and were treated with or without semaglutide (30nmol/kg/day). The study assayed blood biochemistry and then evaluated renal pathological injury through Periodic Acid-Schiff staining and electron microscopy. Metabolomics was utilized to analyze obesity-related metabolites in kidney samples. RESULTS Semaglutide significantly improved glucose homeostasis, insulin resistance, and kidney injury in obese mice. We successfully identified 377 altered metabolites (P<0.05). It was suggested that semaglutide directly improved oxidative stress and inflammation-related metabolites such as nicotinamide adenine dinucleotide (NAD+) and adenosine in the kidney of obese mice, which have not been documented in obesity-related kidney injury. Relevant enriched pathways were included phospholipids and lysophospholipids metabolism, purine metabolism, NAD+ metabolism, and insulin resistance-related metabolism. They could serve as potential targets for intervention of obesity-related kidney injury. CONCLUSION Our study revealed the metabolomics-based renoprotective mechanism of semaglutide in obese mice for the first time. The innovation lied in the identified metabolites such as NAD+ and adenosine targeted by semaglutide, which have not been documented in obesity-related kidney injury. Semaglutide may be a promising therapy for obesity-related kidney diseases.
Collapse
Affiliation(s)
- Xing Chen
- Department of Nephrology, Hebei General Hospital, Shijiazhuang, 050051, People’s Republic of China
| | - Shuchun Chen
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, 050051, People’s Republic of China
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, 050017, People’s Republic of China
| | - Qingjuan Ren
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, 050017, People’s Republic of China
| | - Shu Niu
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, 050017, People’s Republic of China
| | - Xiaoyu Pan
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, 050017, People’s Republic of China
| | - Lin Yue
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, 050017, People’s Republic of China
| | - Zelin Li
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, 050017, People’s Republic of China
| | - Ruiyi Zhu
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, 050017, People’s Republic of China
| | - Zhuoya Jia
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, 050017, People’s Republic of China
| | - Xiaoyi Chen
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, 050017, People’s Republic of China
| | - Ruoxi Zhen
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, 050017, People’s Republic of China
| | - Jiangli Ban
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, 050017, People’s Republic of China
| |
Collapse
|
6
|
Sakuma T, Nakamura M, Chiba T, Iwanaga T, Kan M, Kojima R, Ao J, Ma Y, Unozawa H, Fujita N, Kanayama K, Kanzaki H, Koroki K, Kobayashi K, Nakagawa R, Kanogawa N, Kiyono S, Kondo T, Saito T, Ogasawara S, Nakamoto S, Muroyama R, Kato J, Kishimoto T, Kato N. A diet-induced murine model for non-alcoholic fatty liver disease with obesity and insulin resistance that rapidly develops steatohepatitis and fibrosis. J Transl Med 2022; 102:1150-1157. [PMID: 35643859 DOI: 10.1038/s41374-022-00807-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 05/08/2022] [Accepted: 05/09/2022] [Indexed: 11/09/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) has become the leading cause of chronic liver disease worldwide. Patients with NAFLD often suffer steatohepatitis, which can progress to cirrhosis and hepatocellular carcinoma. The presence of visceral obesity or type 2 diabetes mellitus (T2DM) is a major risk factor and potential therapeutic target for NAFLD. The establishment of animal models with these metabolic comorbidities and with the rapid progression of the disease is needed for developing treatments for NAFLD but remains to be archived. In the present study, KK-Ay mice, widely used as T2DM models, or C57BL6 mice were fed a high-fat, high-fructose, and high-cholesterol diet supplemented with cholic acid (NAFLD diet). The KK-Ay mice fed a NAFLD diet exhibited remarkable obesity and insulin resistance. A prominent accumulation of triglycerides and cholesterol in the liver was observed at 4 weeks. These mice developed steatohepatitis at 4 weeks and fibrosis at 12 weeks. In contrast, C57BL6 mice fed a NAFLD diet remained lean, although they still developed steatohepatitis and fibrosis. In summary, we established a diet-induced murine NAFLD model with the rapid development of steatohepatitis and fibrosis, bearing obesity and insulin resistance. This model could be useful as preclinical models for drug development of NAFLD.
Collapse
Affiliation(s)
- Takafumi Sakuma
- Department of Gastroenterology, Chiba University, Graduate School of Medicine, Chiba, 260-8677, Japan
| | - Masato Nakamura
- Department of Gastroenterology, Chiba University, Graduate School of Medicine, Chiba, 260-8677, Japan.
| | - Tetsuhiro Chiba
- Department of Gastroenterology, Chiba University, Graduate School of Medicine, Chiba, 260-8677, Japan
| | - Terunao Iwanaga
- Department of Gastroenterology, Chiba University, Graduate School of Medicine, Chiba, 260-8677, Japan
| | - Motoyasu Kan
- Department of Gastroenterology, Chiba University, Graduate School of Medicine, Chiba, 260-8677, Japan
| | - Ryuta Kojima
- Department of Gastroenterology, Chiba University, Graduate School of Medicine, Chiba, 260-8677, Japan
| | - Junjie Ao
- Department of Gastroenterology, Chiba University, Graduate School of Medicine, Chiba, 260-8677, Japan
| | - Yaojia Ma
- Department of Gastroenterology, Chiba University, Graduate School of Medicine, Chiba, 260-8677, Japan
| | - Hidemi Unozawa
- Department of Gastroenterology, Chiba University, Graduate School of Medicine, Chiba, 260-8677, Japan
| | - Naoto Fujita
- Department of Gastroenterology, Chiba University, Graduate School of Medicine, Chiba, 260-8677, Japan
| | - Kengo Kanayama
- Department of Gastroenterology, Chiba University, Graduate School of Medicine, Chiba, 260-8677, Japan
| | - Hiroaki Kanzaki
- Department of Gastroenterology, Chiba University, Graduate School of Medicine, Chiba, 260-8677, Japan
| | - Keisuke Koroki
- Department of Gastroenterology, Chiba University, Graduate School of Medicine, Chiba, 260-8677, Japan
| | - Kazufumi Kobayashi
- Department of Gastroenterology, Chiba University, Graduate School of Medicine, Chiba, 260-8677, Japan.,Translational Research and Development Center, Chiba University Hospital, Chiba, 260-8677, Japan
| | - Ryo Nakagawa
- Department of Gastroenterology, Chiba University, Graduate School of Medicine, Chiba, 260-8677, Japan
| | - Naoya Kanogawa
- Department of Gastroenterology, Chiba University, Graduate School of Medicine, Chiba, 260-8677, Japan
| | - Soichiro Kiyono
- Department of Gastroenterology, Chiba University, Graduate School of Medicine, Chiba, 260-8677, Japan
| | - Takayuki Kondo
- Department of Gastroenterology, Chiba University, Graduate School of Medicine, Chiba, 260-8677, Japan
| | - Tomoko Saito
- Department of Gastroenterology, Chiba University, Graduate School of Medicine, Chiba, 260-8677, Japan
| | - Sadahisa Ogasawara
- Department of Gastroenterology, Chiba University, Graduate School of Medicine, Chiba, 260-8677, Japan.,Translational Research and Development Center, Chiba University Hospital, Chiba, 260-8677, Japan
| | - Shingo Nakamoto
- Department of Gastroenterology, Chiba University, Graduate School of Medicine, Chiba, 260-8677, Japan
| | - Ryosuke Muroyama
- Department of Molecular Virology, Chiba University, Graduate School of Medicine, Chiba, 260-8677, Japan
| | - Jun Kato
- Department of Gastroenterology, Chiba University, Graduate School of Medicine, Chiba, 260-8677, Japan
| | - Takashi Kishimoto
- Department of Molecular Pathology, Chiba University, Graduate School of Medicine, Chiba, 260-8677, Japan
| | - Naoya Kato
- Department of Gastroenterology, Chiba University, Graduate School of Medicine, Chiba, 260-8677, Japan
| |
Collapse
|
7
|
Yang F, Zhu W, Edirisuriya P, Ai Q, Nie K, Ji X, Zhou K. Characterization of metabolites and biomarkers for the probiotic effects of Clostridium cochlearium on high-fat diet-induced obese C57BL/6 mice. Eur J Nutr 2022; 61:2217-2229. [DOI: 10.1007/s00394-022-02840-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 02/11/2022] [Indexed: 12/17/2022]
|
8
|
New Insights in the Control of Fat Homeostasis: The Role of Neurotensin. Int J Mol Sci 2022; 23:ijms23042209. [PMID: 35216326 PMCID: PMC8876516 DOI: 10.3390/ijms23042209] [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: 12/28/2021] [Revised: 02/07/2022] [Accepted: 02/15/2022] [Indexed: 12/10/2022] Open
Abstract
Neurotensin (NT) is a small peptide with pleiotropic functions, exerting its primary actions by controlling food intake and energy balance. The first evidence of an involvement of NT in metabolism came from studies on the central nervous system and brain circuits, where NT acts as a neurotransmitter, producing different effects in relation to the specific region involved. Moreover, newer interesting chapters on peripheral NT and metabolism have emerged since the first studies on the NT-mediated regulation of gut lipid absorption and fat homeostasis. Intriguingly, NT enhances fat absorption from the gut lumen in the presence of food with a high fat content, and this action may explain the strong association between high circulating levels of pro-NT, the NT stable precursor, and the increased incidence of metabolic disorders, cardiovascular diseases, and cancer observed in large population studies. This review aims to provide a synthetic overview of the main regulatory effects of NT on several biological pathways, particularly those involving energy balance, and will focus on new evidence on the role of NT in controlling fat homeostasis, thus influencing the risk of unfavorable cardio–metabolic outcomes and overall mortality in humans.
Collapse
|
9
|
Andrés S, Madsen O, Montero O, Martín A, Giráldez FJ. The Role of Feed Restriction on DNA Methylation, Feed Efficiency, Metabolome, Biochemical Profile, and Progesterone Patterns in the Female Filial Generation (F1) Obtained From Early Feed Restricted Ewes (F0). Front Physiol 2022; 12:779054. [PMID: 35024036 PMCID: PMC8745145 DOI: 10.3389/fphys.2021.779054] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 11/18/2021] [Indexed: 12/24/2022] Open
Abstract
Deficient management of replacement animals in the farm during early developmental windows may promote adverse programming effects on reproductive traits and subsequent transmission to the next generation. In this sense, DNA methylation profiles allow researchers to decode epigenetic regulation mechanisms in mammals and identify novel candidate genes correlated with phenotype differences in both dams and offspring. Therefore, improving knowledge in the field of epigenetics and intergenerational effects caused by prenatal and postnatal early nutritional events (e.g., feed restriction) is crucial for refining strategies dedicated to animal breeding. In this study, we determined differences in the global blood methylation patterns, biochemical profile, and metabolome of ewe lambs (F1) born from either early feed restricted dams (F0-RES) or fed ad libitum (F0-ADL). Our data show that functional categories such as those related to cellular processes, phosphorylation, nervous system, immunity response, or reproductive function were enriched significantly in the F1-RES lambs due to differences in the methylation of genes in these categories. These F1-RES lambs did not show differences in feed efficiency during the replacement period but presented higher levels of insulin and triglycerides and reduced concentration of progesterone, whereas the metabolome profile demonstrated variations in the bile acid composition when compared with the F1-ADL lambs. Taken together, all these results suggest that intergenerational effects caused by early feed restriction of dams (F0) may persist in the F1 female lambs with negative consequences on genes involved in cellular processes and reproductive traits.
Collapse
Affiliation(s)
- Sonia Andrés
- Instituto de Ganadería de Montaña, CSIC-Universidad de León, León, Spain
| | - Ole Madsen
- Animal Breeding and Genomics, Wageningen University and Research, Wageningen, Netherlands
| | - Olimpio Montero
- Instituto de Biología y Genética Molecular, CSIC, Valladolid, Spain
| | - Alba Martín
- Instituto de Ganadería de Montaña, CSIC-Universidad de León, León, Spain
| | - F Javier Giráldez
- Instituto de Ganadería de Montaña, CSIC-Universidad de León, León, Spain
| |
Collapse
|
10
|
The Synbiotic Combination of Akkermansia muciniphila and Quercetin Ameliorates Early Obesity and NAFLD through Gut Microbiota Reshaping and Bile Acid Metabolism Modulation. Antioxidants (Basel) 2021; 10:antiox10122001. [PMID: 34943104 PMCID: PMC8698339 DOI: 10.3390/antiox10122001] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/10/2021] [Accepted: 12/13/2021] [Indexed: 12/11/2022] Open
Abstract
Gut microbiota plays a key role in obesity and non-alcoholic fatty liver disease (NAFLD), so synbiotics could be a therapeutic alternative. We aim to evaluate a nutritional intervention together with the administration of the bacteria Akkermansia muciniphila and the antioxidant quercetin in an in vivo model of early obesity and NAFLD. 21-day-old rats were fed with control or high-fat diet for six weeks. Then, all animals received control diet supplemented with/without quercetin and/or A. muciniphila for three weeks. Gut microbiota, NAFLD-related parameters, circulating bile acids (BAs) and liver gene expression were analyzed. The colonization with A. muciniphila was associated with less body fat, while synbiotic treatment caused a steatosis remission, linked to hepatic lipogenesis modulation. The synbiotic promoted higher abundance of Cyanobacteria and Oscillospira, and lower levels of Actinobacteria, Lactococcus, Lactobacillus and Roseburia. Moreover, it favored elevated unconjugated hydrophilic BAs plasma levels and enhanced hepatic expression of BA synthesis and transport genes. A. muciniphila correlated with circulating BAs and liver lipid and BA metabolism genes, suggesting a role of this bacterium in BA signaling. Beneficial effects of A. muciniphila and quercetin combination are driven by gut microbiota modulation, the shift in BAs and the gut-liver bile flow enhancement.
Collapse
|
11
|
Kim S, Choi S, Dutta M, Asubonteng JO, Polunas M, Goedken M, Gonzalez FJ, Cui JY, Gyamfi MA. Pregnane X receptor exacerbates nonalcoholic fatty liver disease accompanied by obesity- and inflammation-prone gut microbiome signature. Biochem Pharmacol 2021; 193:114698. [PMID: 34303710 PMCID: PMC9135326 DOI: 10.1016/j.bcp.2021.114698] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 06/18/2021] [Accepted: 06/21/2021] [Indexed: 02/06/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most prevalent chronic liver disease due to the current epidemics of obesity and diabetes. The pregnane X receptor (PXR) is a xenobiotic-sensing nuclear receptor known for trans-activating liver genes involved in drug metabolism and transport, and more recently implicated in energy metabolism. The gut microbiota can modulate the host xenobiotic biotransformation and contribute to the development of obesity. While the male sex confers a higher risk for NAFLD than women before menopause, the mechanism remains unknown. We hypothesized that the presence of PXR promotes obesity by modifying the gut-liver axis in a sex-specific manner. Male and female C57BL/6 (wild-type/WT) and PXR-knockout (PXR-KO) mice were fed control or high-fat diet (HFD) for 16-weeks. Serum parameters, liver histopathology, transcriptomic profiling, 16S-rDNA sequencing, and bile acid (BA) metabolomics were performed. PXR enhanced HFD-induced weight gain, hepatic steatosis and inflammation especially in males, accompanied by PXR-dependent up-regulation in hepatic genes involved in microbial response, inflammation, oxidative stress, and cancer; PXR-dependent increase in intestinal Firmicutes/Bacteroides ratio (hallmark of obesity) and the pro-inflammatory Lactobacillus, as well as a decrease in the anti-obese Allobaculum and the anti-inflammatory Bifidobacterum, with a PXR-dependent reduction of beneficial BAs in liver. The resistance to NAFLD in females may be explained by PXR-dependent decrease in pro-inflammatory bacteria (Ruminococcus gnavus and Peptococcaceae). In conclusion, PXR exacerbates hepatic steatosis and inflammation accompanied by obesity- and inflammation-prone gut microbiome signature, suggesting that gut microbiome may contribute to PXR-mediated exacerbation of NAFLD.
Collapse
Affiliation(s)
- Sarah Kim
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA
| | - Sora Choi
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, NC, USA
| | - Moumita Dutta
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA
| | - Jeffrey O Asubonteng
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, NC, USA
| | - Marianne Polunas
- Office of Research and Economic Development, Research Pathology Services, Rutgers University, Piscataway, NJ, USA
| | - Michael Goedken
- Office of Research and Economic Development, Research Pathology Services, Rutgers University, Piscataway, NJ, USA
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Julia Yue Cui
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA.
| | - Maxwell A Gyamfi
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, NC, USA.
| |
Collapse
|
12
|
Beneficial effects of a combination of Clostridium cochlearium and Lactobacillus acidophilus on body weight gain, insulin sensitivity, and gut microbiota in high-fat diet-induced obese mice. Nutrition 2021; 93:111439. [PMID: 34507264 DOI: 10.1016/j.nut.2021.111439] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/10/2021] [Accepted: 07/19/2021] [Indexed: 02/07/2023]
Abstract
OBJECTIVES Species Lactobacillus acidophilus and butyrate producer Clostridium cochlearium have been shown to have potential antiobesity effects. The aim of this study was to show that the combination of C. cochlearium and L. acidophilus (CC-LA) has beneficial effects on body weight control and glucose homeostasis in high-fat diet-induced obese (DIO) mice. METHODS In this study, thirty-six 6-wk-old male C57BL/6 mice were randomly assigned to three groups of 12 mice each. The experimental group (CC-LA) was administered with CC-LA mixture and fed ad libitum with a high-fat diet. High-fat diet (HF) control and low-fat diet (LF) control groups were treated with the same dose of sterile water as the CC-LA group. RESULTS After 17 wk of dietary intervention, the CC-LA group showed 17% less body weight gain than the HF group did (P < 0.01). The CC-LA group also showed significantly reduced incremental area under the curve of oral glucose tolerance test and homeostatic model assessment for insulin resistance compared with the HF group. The results from 16S rRNA sequencing analysis of gut microbiota showed that the CC-LA administration led to overall increased α-diversity indices, and a significant microbial separation from the HF group. The ratio of Firmicutes to Bacteroidetes (F/B) was reduced from 3.30 in the HF group to 1.94 in the CC-LA group. The relative abundances of certain obesity-related taxa were also decreased by CC-LA administration. CONCLUSION The present study provided evidence that the CC-LA combination reduced obesity and improved glucose metabolism in high-fat diet-treated DIO mice, potentially mediated by the modulation of gut microbiota.
Collapse
|
13
|
Li J, Song J, Yan B, Weiss HL, Weiss LT, Gao T, Evers BM. Neurotensin differentially regulates bile acid metabolism and intestinal FXR-bile acid transporter axis in response to nutrient abundance. FASEB J 2021; 35:e21371. [PMID: 33811694 DOI: 10.1096/fj.202001692r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 12/18/2020] [Accepted: 12/31/2020] [Indexed: 01/07/2023]
Abstract
Studies demonstrate a role for neurotensin (NT) in obesity and related comorbidities. Bile acid (BA) homeostasis alterations are associated with obesity. We determined the effect of NT on BA metabolism in obese and non-obese conditions. Plasma and fecal BA profiles were analyzed by LC-MS/MS in male and female NT+/+ and NT-/- mice fed low-fat (LFD) or high-fat diet (HFD) for 6 weeks (early stage of obesity) or greater than 20 weeks (late stage of obesity). The nuclear farnesoid X receptor (FXR) and BA transporter mRNA expression were assessed in ileum, mouse enteroids, and human cell lines. HFD decreased plasma primary and secondary BAs in NT+/+ mice; HFD-induced decrease of plasma BAs was improved in NT-deficient mice. In NT+/+ mice, HFD inhibited ileal FXR and BA transporter expression; HFD-decreased expression of FXR and BA transporters was prevented in NT-/- mice. Compared with LFD-fed NT+/+ mice, LFD-fed NT-/- mice had relatively lower levels of ileal FXR and BA transporter expression. Moreover, NT stimulates the expression of FXR and BA transporters in Caco-2 cells; however, stimulated expression of BA transporters was attenuated in NT-/- enteroids. Therefore, we demonstrate that HFD disrupts the BA metabolism and ileal FXR and BA transporter axis which are improved in the absence of NT, suggesting that NT contributes to HFD-induced disruption of BA metabolism and plays an inhibitory role in the regulation of ileal FXR and BA transporter signaling under obese conditions. Conversely, NT positively regulates the expression of ileal FXR and BA transporters under non-obese conditions. Therefore, NT plays a dual role in obese and non-obese conditions, suggesting possible therapeutic strategies for obesity control.
Collapse
Affiliation(s)
- Jing Li
- Department of Surgery, University of Kentucky, Lexington, KY, USA.,Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Jun Song
- Department of Surgery, University of Kentucky, Lexington, KY, USA.,Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Baoxiang Yan
- Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Heidi L Weiss
- Markey Cancer Center, University of Kentucky, Lexington, KY, USA.,Department of Internal Medicine, University of Kentucky, Lexington, KY, USA
| | - L Todd Weiss
- Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Tianyan Gao
- Markey Cancer Center, University of Kentucky, Lexington, KY, USA.,Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, USA
| | - B Mark Evers
- Department of Surgery, University of Kentucky, Lexington, KY, USA.,Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
14
|
Ipsen DH, Lykkesfeldt J, Tveden-Nyborg P. Animal Models of Fibrosis in Nonalcoholic Steatohepatitis: Do They Reflect Human Disease? Adv Nutr 2020; 11:1696-1711. [PMID: 33191435 PMCID: PMC7666900 DOI: 10.1093/advances/nmaa081] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 05/06/2020] [Accepted: 06/11/2020] [Indexed: 12/12/2022] Open
Abstract
Nonalcoholic steatohepatitis (NASH) is one of the most common chronic liver diseases in the world, yet no pharmacotherapies are available. The lack of translational animal models is a major barrier impeding elucidation of disease mechanisms and drug development. Multiple preclinical models of NASH have been proposed and can broadly be characterized as diet-induced, deficiency-induced, toxin-induced, genetically induced, or a combination of these. However, very few models develop advanced fibrosis while still reflecting human disease etiology or pathology, which is problematic since fibrosis stage is considered the best prognostic marker in patients and an important endpoint in clinical trials of NASH. While mice and rats predominate the NASH research, several other species have emerged as promising models. This review critically evaluates animal models of NASH, focusing on their ability to develop advanced fibrosis while maintaining their relevance to the human condition.
Collapse
Affiliation(s)
- David H Ipsen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C, Denmark
| | - Jens Lykkesfeldt
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C, Denmark
| | | |
Collapse
|
15
|
Liou CJ, Dai YW, Wang CL, Fang LW, Huang WC. Maslinic acid protects against obesity-induced nonalcoholic fatty liver disease in mice through regulation of the Sirt1/AMPK signaling pathway. FASEB J 2019; 33:11791-11803. [PMID: 31361524 DOI: 10.1096/fj.201900413rrr] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Maslinic acid is a pentacyclic triterpenoid that is distributed in the peel of olives. Previous studies found that maslinic acid inhibited inflammatory response and antioxidant effects. We investigated whether maslinic acid ameliorates nonalcoholic fatty liver disease in mice with high-fat-diet (HFD)-induced obesity and evaluated the regulation of lipogenesis in hepatocytes. Male C57BL/6 mice fed a normal diet or HFD (60% fat, w/w) were tested for 16 wk. After the fourth week, mice were injected intraperitoneally with maslinic acid for 12 wk. In another experiment, HepG2 cells were treated with oleic acid to induce lipid accumulation or maslinic acid to evaluate lipogenesis. Maslinic acid significantly reduced body weight compared with HFD-fed mice. Maslinic acid reduced liver weight and liver lipid accumulation and improved hepatocyte steatosis. Furthermore, serum glucose, leptin, and free fatty acid concentrations significantly reduced, but the serum adiponectin concentration was higher, in the maslinic acid group than in the HFD group. In liver tissue, maslinic acid suppressed transcription factors involved in lipogenesis and increased adipose triglyceride lipase. In vitro, maslinic acid decreased lipogenesis by activating AMPK. These findings suggest that maslinic acid acts against hepatic steatosis by regulating enzyme activity involved in lipogenesis, lipolysis, and fatty acid oxidation in the liver.-Liou, C.-J., Dai, Y.-W., Wang, C.-L., Fang, L.-W., Huang, W.-C. Maslinic acid protects against obesity-induced nonalcoholic fatty liver disease in mice through regulation of the Sirt1/AMPK signaling pathway.
Collapse
Affiliation(s)
- Chian-Jiun Liou
- Division of Basic Medical Sciences, Department of Nursing, Research Center for Chinese Herbal Medicine, Graduate Institute of Health Industry Technology, Chang Gung University of Science and Technology, Taoyuan City, Taiwan.,Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, Linkou, Taoyuan City, Taiwan
| | - Yi-Wen Dai
- Research Center for Food and Cosmetic Safety, Research Center for Chinese Herbal Medicine, Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan City, Taiwan
| | - Chia-Ling Wang
- Research Center for Food and Cosmetic Safety, Research Center for Chinese Herbal Medicine, Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan City, Taiwan
| | - Li-Wen Fang
- Department of Nutrition, I-Shou University, Kaohsiung City, Taiwan
| | - Wen-Chung Huang
- Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, Linkou, Taoyuan City, Taiwan.,Research Center for Food and Cosmetic Safety, Research Center for Chinese Herbal Medicine, Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan City, Taiwan
| |
Collapse
|