1
|
Li S, Han H, Yang K, Li X, Ma L, Yang Z, Zhao YX. Emerging role of metabolic reprogramming in the immune microenvironment and immunotherapy of thyroid cancer. Int Immunopharmacol 2025; 144:113702. [PMID: 39602959 DOI: 10.1016/j.intimp.2024.113702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/07/2024] [Accepted: 11/20/2024] [Indexed: 11/29/2024]
Abstract
The metabolic reprogramming of cancer cells is a hallmark of many malignancies. To meet the energy acquisition needs of tumor cells for rapid proliferation, tumor cells reprogram their nutrient metabolism, which is caused by the abnormal expression of transcription factors and signaling molecules related to energy metabolic pathways as well as the upregulation and downregulation of abnormal metabolic enzymes, receptors, and mediators. Thyroid cancer (TC) is the most common endocrine tumor, and immunotherapy has become the mainstream choice for clinical benefit after the failure of surgical, endocrine, and radioiodine therapies. TC change the tumor microenvironment (TME) through nutrient competition and metabolites, causing metabolic reprogramming of immune cells, profoundly changing immune cell function, and promoting immune evasion of tumor cells. A deeper understanding of how metabolic reprogramming alters the TME and controls immune cell fate and function will help improve the effectiveness of TC immunotherapy and patient outcomes. This paper aims to elucidate the metabolic communication that occurs between immune cells around TC and discusses how metabolic reprogramming in TC affects the immune microenvironment and the effectiveness of anti-cancer immunotherapy. Finally, targeting key metabolic checkpoints during metabolic reprogramming, combined with immunotherapy, is a promising strategy.
Collapse
Affiliation(s)
- Shouhua Li
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China.
| | - Hengtong Han
- The Seventh Department of General Surgery, Department of Thyroid Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China.
| | - Kaili Yang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China.
| | - Xiaoxiao Li
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China.
| | - Libin Ma
- The Seventh Department of General Surgery, Department of Thyroid Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China.
| | - Ze Yang
- The Seventh Department of General Surgery, Department of Thyroid Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China.
| | - Yong-Xun Zhao
- The Seventh Department of General Surgery, Department of Thyroid Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
2
|
Luangphiphat W, Prombutara P, Muangsillapasart V, Sukitpunyaroj D, Eeckhout E, Taweechotipatr M. Exploring of gut microbiota features in dyslipidemia and chronic coronary syndrome patients undergoing coronary angiography. Front Microbiol 2024; 15:1384146. [PMID: 38646625 PMCID: PMC11026706 DOI: 10.3389/fmicb.2024.1384146] [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: 02/12/2024] [Accepted: 03/25/2024] [Indexed: 04/23/2024] Open
Abstract
Chronic coronary syndrome (CCS) has a high mortality rate, and dyslipidemia is a major risk factor. Atherosclerosis, a cause of CCS, is influenced by gut microbiota dysbiosis and its metabolites. The objective of this study was to study the diversity and composition of gut microbiota and related clinical parameters among CCS patients undergoing coronary angiography and dyslipidemia patients in comparison to healthy volunteers in Thailand. CCS patients had more risk factors and higher inflammatory markers, high-sensitivity C-reactive protein (hs-CRP) than others. The alpha diversity was lower in dyslipidemia and CCS patients than in the healthy group. A significant difference in the composition of gut microbiota was observed among the three groups. The relative abundance of Proteobacteria, Fusobacteria, Enterobacteriaceae, Prevotella, and Streptococcus was significantly increased while Roseburia, Ruminococcus, and Faecalibacterium were lower in CCS patients. In CCS patients, Lachnospiraceae, Peptostreptococcaceae, and Pediococcus were positively correlated with hs-CRP. In dyslipidemia patients, Megasphaera was strongly positively correlated with triglyceride (TG) level and negatively correlated with high-density lipoprotein cholesterol (HDL-C). The modification of gut microbiota was associated with changes in clinical parameters involved in the development of coronary artery disease (CAD) in CCS patients.
Collapse
Affiliation(s)
- Wongsakorn Luangphiphat
- Innovative Anatomy Program, Faculty of Medicine, Srinakharinwirot University, Bangkok, Thailand
- Division of Cardiology, Department of Medicine, Chulabhorn Hospital, Chulabhorn Royal Academy, Bangkok, Thailand
- Princess Srisavangavadhana College of Medicine, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Pinidphon Prombutara
- Omics Sciences and Bioinformatics Center, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
- Mod Gut Co., Ltd., Bangkok, Thailand
| | - Viroj Muangsillapasart
- Division of Cardiology, Department of Medicine, Chulabhorn Hospital, Chulabhorn Royal Academy, Bangkok, Thailand
- Princess Srisavangavadhana College of Medicine, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Damrong Sukitpunyaroj
- Division of Cardiology, Department of Medicine, Chulabhorn Hospital, Chulabhorn Royal Academy, Bangkok, Thailand
- Princess Srisavangavadhana College of Medicine, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Eric Eeckhout
- Service of Cardiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Malai Taweechotipatr
- Center of Excellence in Probiotics, Srinakharinwirot University, Bangkok, Thailand
- Department of Microbiology, Faculty of Medicine, Srinakharinwirot University, Bangkok, Thailand
- Clinical Research Center, Faculty of Medicine, Srinakharinwirot University, Ongkharak, Thailand
| |
Collapse
|
3
|
Wahabi S, Rtibi K, Atouani A, Sebai H. Anti-Obesity Actions of Two Separated Aqueous Extracts From Arbutus ( Arbutus unedo) and Hawthorn ( Crataegus monogyna) Fruits Against High-Fat Diet in Rats via Potent Antioxidant Target. Dose Response 2023; 21:15593258231179904. [PMID: 37275393 PMCID: PMC10236257 DOI: 10.1177/15593258231179904] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 05/17/2023] [Indexed: 06/07/2023] Open
Abstract
Arbutus unedo and Crataegus monogyna are widely distributed throughout the Mediterranean basin and commonly used in folk medicine against a wide range of diseases. Therefore, the present study has been designed to evaluate the anti-obesity potential of two aqueous extracts of the fruits of A. unedo (AUAE) and C. monogyna (CMAE). Male Wistar rats were supplied with a standard diet (SD), high-fat diet (HFD), HFD with the two separated extracts at the same dose (300 mg/kg, BW, p. o.), or HFD with atorvastatin-(ATOR) (2.1 mg/kg, BW, p. o.) for 12 weeks. Lipid profile and the liver and kidney linked-markers were assessed. Besides, obesity-related disorders' biomarkers were measured. AUAE, CMAE, and ATOR were observed to reduce significantly total body and organ weights following HFD-induced obese rat models. Likewise, epididymal and abdominal adipose tissue weights were noticeably decreased in HFD rats treated with both extracts and ATOR. Added to that, biochemical and metabolic changes were normalized by significant attenuation of lipid peroxidation accompanied with an increase of thiol-group concentrations and antioxidant status. More importantly, a modulation in trace element levels was revealed when compared with HFD group. Altogether, current study concluded that AUAE and CMAE could be potential candidates for the prevention and treatment of obesity and related disturbs induced by HFD.
Collapse
Affiliation(s)
- Soumaya Wahabi
- Laboratory of Functional Physiology
and Valorization of Bio-Ressources-Higher Institute of Biotechnology of Beja, University of Jendouba, Beja, Tunisia
| | - Kais Rtibi
- Laboratory of Functional Physiology
and Valorization of Bio-Ressources-Higher Institute of Biotechnology of Beja, University of Jendouba, Beja, Tunisia
| | - Amal Atouani
- Clinical Biology Laboratory, Beja
Regional Hospital, Beja, Tunisia
| | - Hichem Sebai
- Laboratory of Functional Physiology
and Valorization of Bio-Ressources-Higher Institute of Biotechnology of Beja, University of Jendouba, Beja, Tunisia
- University of Jendouba, Jendouba, Tunisia
| |
Collapse
|
4
|
Roessler J, Leistner DM, Landmesser U, Haghikia A. Modulatory role of gut microbiota in cholesterol and glucose metabolism: Potential implications for atherosclerotic cardiovascular disease Atherosclerosis. Atherosclerosis 2022; 359:1-12. [PMID: 36126379 DOI: 10.1016/j.atherosclerosis.2022.08.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 08/12/2022] [Accepted: 08/31/2022] [Indexed: 11/02/2022]
Abstract
Accumulating evidence suggests an important role of gut microbiota in physiological processes of host metabolism as well as cardiometabolic disease. Recent advances in metagenomic and metabolomic research have led to discoveries of novel pathways in which intestinal microbial metabolism of dietary nutrients is linked to metabolic profiles and cardiovascular disease risk. A number of metaorganismal circuits have been identified by microbiota transplantation studies and experimental models using germ-free rodents. Many of these pathways involve gut microbiota-related bioactive metabolites that impact host metabolism, in particular lipid and glucose homeostasis, partly via specific host receptors. In this review, we summarize the current knowledge of how the gut microbiome can impact cardiometabolic phenotypes and provide an overview of recent advances of gut microbiome research. Finally, the potential of modulating intestinal microbiota composition and/or targeting microbiota-related pathways for novel preventive and therapeutic strategies in cardiometabolic and cardiovascular diseases will be discussed.
Collapse
Affiliation(s)
- Johann Roessler
- Department of Cardiology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - David M Leistner
- Department of Cardiology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany; DZHK (German Center of Cardiovascular Research), Partner Site Berlin, Berlin, Germany; Berlin Institute of Health (BIH), Berlin, Germany
| | - Ulf Landmesser
- Department of Cardiology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany; DZHK (German Center of Cardiovascular Research), Partner Site Berlin, Berlin, Germany; Berlin Institute of Health (BIH), Berlin, Germany
| | - Arash Haghikia
- Department of Cardiology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany; DZHK (German Center of Cardiovascular Research), Partner Site Berlin, Berlin, Germany; Berlin Institute of Health (BIH), Berlin, Germany.
| |
Collapse
|
5
|
Gruzdeva OV, Dyleva YA, Belik EV, Sinitsky MY, Stasev AN, Kokov AN, Brel NK, Krivkina EO, Bychkova EE, Tarasov RS, Barbarash OL. Relationship between Epicardial and Coronary Adipose Tissue and the Expression of Adiponectin, Leptin, and Interleukin 6 in Patients with Coronary Artery Disease. J Pers Med 2022; 12:jpm12020129. [PMID: 35207618 PMCID: PMC8877574 DOI: 10.3390/jpm12020129] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/12/2022] [Accepted: 01/17/2022] [Indexed: 12/11/2022] Open
Abstract
Adipose tissue (AT) is an endocrine and paracrine organ that synthesizes biologically active adipocytokines, which affect inflammation, fibrosis, and atherogenesis. Epicardial and perivascular fat depots are of great interest to researchers, owing to their potential effects on the myocardium and blood vessels. The aim of the study was to assess the expression and secretion of adipocytokine genes in the AT of patients with coronary artery disease (CAD) and patients with aortic or mitral valve replacement. This study included 84 patients with CAD and 50 patients with aortic or mitral valve replacement. Adipocytes were isolated from subcutaneous, epicardial (EAT), and perivascular AT (PVAT), and were cultured for 24 h. EAT exhibited the lowest level of adiponectin gene expression and secretion, regardless of nosology, and high expression levels of the leptin gene and interleukin-6 (IL-6). However, EAT adipocytes in patients with CAD were characterized by more pronounced changes in comparison with the group with heart defects. High leptin and IL-6 levels resulted in increased pro-inflammatory activity, as observed in both EAT and PVAT adipocytes, especially in individuals with CAD. Therefore, our results revealed the pathogenetic significance of alterations in the adipokine and cytokine status of adipocytes of EAT and PVAT in patients with CAD.
Collapse
|
6
|
Wang Y, You S, Su S, Yeon A, Lo EM, Kim S, Mohler JL, Freeman MR, Kim HL. Cholesterol-Lowering Intervention Decreases mTOR Complex 2 Signaling and Enhances Antitumor Immunity. Clin Cancer Res 2022; 28:414-424. [PMID: 34728526 PMCID: PMC8776603 DOI: 10.1158/1078-0432.ccr-21-1535] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 08/08/2021] [Accepted: 10/28/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE There is a need for strategies to prevent prostate cancer. Cholesterol-lowering interventions are employed widely and safely to reduce risk of cardiovascular disease and has been proposed for chemoprevention. Using preclinical models and a window-of-opportunity clinical trial, we describe an adaptive antitumor immunity resulting from cholesterol lowering. EXPERIMENTAL DESIGN Statins do not reliably lower serum cholesterol in mice. Therefore, oral ezetimibe was administered to mice to lower serum cholesterol to clinically relevant levels and evaluated the final adaptive immune response. T-lymphocytes-specific mTORC2 knockout mice were used to evaluate mTOR signaling and antitumor immunity. Pretreatment and posttreatment prostate tumors and lymphocytes were examined from a window-of-opportunity clinical trial where men with prostate cancer were treated with 2 to 6 weeks of aggressive cholesterol-lowering intervention prior to radical prostatectomy. RESULTS Mice treated with oral ezetimibe exhibited enhanced antitumor immunity against syngeneic cancers in a CD8+ lymphocyte-dependent manner, produced immunity that was transferrable through lymphocytes, and had enhanced central CD8+ T-cell memory. In mice and in patients undergoing prostatectomy, lowering serum cholesterol inhibited mTORC2 signaling in lymphocytes and increased infiltration of CD8+ lymphocytes into prostate tumors. T-lymphocyte-specific mTORC2 knockout mice demonstrated enhanced CD8+ lymphocyte function and antitumor capacity. In patients, cholesterol-lowering intervention prior to prostatectomy decreased the proliferation of normal prostate and low-grade adenocarcinomas. CONCLUSIONS Lowering serum cholesterol decreased signaling through mTORC2 and enhanced antitumor CD8+ T-cell memory. We provide a rationale for large-scale clinical testing of cholesterol lowering strategies for prostate cancer chemoprevention.
Collapse
Affiliation(s)
- Yanping Wang
- Department of Surgery/Division of Urology, Cedars-Sinai Medical Center, Los Angeles, California
| | - Sungyong You
- Department of Surgery/Division of Urology, Cedars-Sinai Medical Center, Los Angeles, California
| | - Shengchen Su
- Department of Surgery/Division of Urology, Cedars-Sinai Medical Center, Los Angeles, California
| | - Austin Yeon
- Department of Surgery/Division of Urology, Cedars-Sinai Medical Center, Los Angeles, California
| | - Eric M Lo
- Department of Surgery/Division of Urology, Cedars-Sinai Medical Center, Los Angeles, California
| | - Sungjin Kim
- Biostatistics and Bioinformatics Core, Cedars-Sinai Medical Center, Los Angeles, California
| | - James L Mohler
- Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Michael R Freeman
- Department of Surgery/Division of Urology, Cedars-Sinai Medical Center, Los Angeles, California
| | - Hyung L Kim
- Department of Surgery/Division of Urology, Cedars-Sinai Medical Center, Los Angeles, California.
| |
Collapse
|
7
|
Gendaszewska-Darmach E, Garstka MA, Błażewska KM. Targeting Small GTPases and Their Prenylation in Diabetes Mellitus. J Med Chem 2021; 64:9677-9710. [PMID: 34236862 PMCID: PMC8389838 DOI: 10.1021/acs.jmedchem.1c00410] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
![]()
A fundamental role
of pancreatic β-cells to maintain proper
blood glucose level is controlled by the Ras superfamily of small
GTPases that undergo post-translational modifications, including prenylation.
This covalent attachment with either a farnesyl or a geranylgeranyl
group controls their localization, activity, and protein–protein
interactions. Small GTPases are critical in maintaining glucose homeostasis
acting in the pancreas and metabolically active tissues such as skeletal
muscles, liver, or adipocytes. Hyperglycemia-induced upregulation
of small GTPases suggests that inhibition of these pathways deserves
to be considered as a potential therapeutic approach in treating T2D.
This Perspective presents how inhibition of various points in the
mevalonate pathway might affect protein prenylation and functioning
of diabetes-affected tissues and contribute to chronic inflammation
involved in diabetes mellitus (T2D) development. We also demonstrate
the currently available molecular tools to decipher the mechanisms
linking the mevalonate pathway’s enzymes and GTPases with diabetes.
Collapse
Affiliation(s)
- Edyta Gendaszewska-Darmach
- Institute of Molecular and Industrial Biotechnology, Faculty of Biotechnology and Food Sciences, Lodz University of Technology, Stefanowskiego Street 4/10, 90-924 Łódź, Poland
| | - Malgorzata A Garstka
- Core Research Laboratory, Department of Endocrinology, Department of Tumor and Immunology, Precision Medical Institute, Western China Science and Technology Innovation Port, School of Medicine, the Second Affiliated Hospital of Xi'an Jiaotong University, DaMingGong, Jian Qiang Road, Wei Yang district, Xi'an 710016, China
| | - Katarzyna M Błażewska
- Institute of Organic Chemistry, Faculty of Chemistry, Lodz University of Technology, Żeromskiego Street 116, 90-924 Łódź, Poland
| |
Collapse
|
8
|
Lozano-Cuenca J, Valencia-Hernández I, López-Canales OA, Flores-Herrera H, López-Mayorga RM, Castillo-Henkel EF, López-Canales JS. Possible mechanisms involved in the effect of the subchronic administration of rosuvastatin on endothelial function in rats with metabolic syndrome. ACTA ACUST UNITED AC 2020; 53:e9304. [PMID: 32049102 PMCID: PMC7011172 DOI: 10.1590/1414-431x20199304] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Accepted: 11/11/2019] [Indexed: 01/18/2023]
Abstract
Metabolic syndrome is a multifaceted condition associated with a greater risk of various disorders (e.g., diabetes and heart disease). In a rat model of metabolic syndrome, an acute in vitro application of rosuvastatin causes relaxation of aortic rings. Since the outcome of a subchronic rosuvastatin treatment is unknown, the present study explored its effect on acetylcholine-induced vasorelaxation of aortic rings from rats with metabolic syndrome. Animals were submitted to a 16-week treatment, including a standard diet, a cafeteria-style diet (CAF-diet), or a CAF-diet with daily rosuvastatin treatment (10 mg/kg). After confirming the development of metabolic syndrome in rats, aortic segments were extracted from these animals (those treated with rosuvastatin and untreated) and the acetylcholine-induced relaxant effect on the corresponding rings was evaluated. Concentration-response curves were constructed for this effect in the presence/absence of L-NAME, ODQ, KT 5823, 4-aminopyridine (4-AP), tetraethylammonium (TEA), apamin plus charybdotoxin, glibenclamide, indomethacin, clotrimazole, and cycloheximide pretreatment. Compared to rings from control rats, acetylcholine-induced vasorelaxation decreased in rings from animals with metabolic syndrome, and was maintained at a normal level in animals with metabolic syndrome plus rosuvastatin treatment. The effect of rosuvastatin was inhibited by L-NAME, ODQ, KT 5823, TEA, apamin plus charybdotoxin, but unaffected by 4-AP, glibenclamide, indomethacin, clotrimazole, or cycloheximide. In conclusion, the subchronic administration of rosuvastatin to rats with metabolic syndrome improved the acetylcholine-induced relaxant response, involving stimulation of the NO/cGMP/PKG/Ca2+-activated K+ channel pathway.
Collapse
Affiliation(s)
- J Lozano-Cuenca
- Department of Physiology and Cell Development, National Institute of Perinatology, Mexico City, Mexico
| | - I Valencia-Hernández
- Section of Postgraduate Studies and Investigation, Higher School of Medicine, National Polytechnic Institute, Mexico City, Mexico
| | - O A López-Canales
- Section of Postgraduate Studies and Investigation, Higher School of Medicine, National Polytechnic Institute, Mexico City, Mexico
| | - H Flores-Herrera
- Department of Immuno-Biochemistry, National Institute of Perinatology, Mexico City, Mexico
| | - R M López-Mayorga
- Section of Postgraduate Studies and Investigation, Higher School of Medicine, National Polytechnic Institute, Mexico City, Mexico
| | - E F Castillo-Henkel
- Section of Postgraduate Studies and Investigation, Higher School of Medicine, National Polytechnic Institute, Mexico City, Mexico
| | - J S López-Canales
- Department of Physiology and Cell Development, National Institute of Perinatology, Mexico City, Mexico.,Section of Postgraduate Studies and Investigation, Higher School of Medicine, National Polytechnic Institute, Mexico City, Mexico
| |
Collapse
|
9
|
Kim J, Lee H, An J, Song Y, Lee CK, Kim K, Kong H. Alterations in Gut Microbiota by Statin Therapy and Possible Intermediate Effects on Hyperglycemia and Hyperlipidemia. Front Microbiol 2019; 10:1947. [PMID: 31551944 PMCID: PMC6736992 DOI: 10.3389/fmicb.2019.01947] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Accepted: 08/08/2019] [Indexed: 12/17/2022] Open
Abstract
Dysbiosis of the gut microbiota is a contributing factor for obesity-related metabolic diseases such as hyperglycemia and hyperlipidemia. Pharmacotherapy for metabolic diseases involves the modulation of gut microbiota, which is suggested to be a potential therapeutic target. In this study, the modulation of gut microbiota by statins (cholesterol-lowering drugs: atorvastatin and rosuvastatin) was investigated in an aged mouse model of high-fat diet-induced obesity, and the association between gut microbiota and immune responses was described. Atorvastatin and rosuvastatin significantly increased the abundance of the genera Bacteroides, Butyricimonas, and Mucispirillum. Moreover, the abundance of these genera was correlated with the inflammatory response, including levels of IL-1β and TGFβ1 in the ileum. In addition, oral fecal microbiota transplantation with fecal material collected from rosuvastatin-treated mouse groups improved hyperglycemia. From these results, the effect of statins on metabolic improvements could be explained by altered gut microbiota. Our findings suggest that the modulation of gut microbiota by statins has an important role in the therapeutic actions of these drugs.
Collapse
Affiliation(s)
- Jiyeon Kim
- College of Pharmacy, Sahmyook University, Seoul, South Korea
| | - Heetae Lee
- College of Pharmacy, Sahmyook University, Seoul, South Korea
| | - Jinho An
- College of Pharmacy, Sahmyook University, Seoul, South Korea
| | - Youngcheon Song
- College of Pharmacy, Sahmyook University, Seoul, South Korea
| | - Chong-Kil Lee
- College of Pharmacy, Chungbuk National University, Cheongju, South Korea
| | - Kyungjae Kim
- College of Pharmacy, Sahmyook University, Seoul, South Korea
| | - Hyunseok Kong
- College of Animal Biotechnology and Resource, Sahmyook University, Seoul, South Korea
| |
Collapse
|
10
|
Sina Z, Nasrollahzadeh J, Shokraei S, Rismanchi M, Foroughi F. Black and red peppers attenuates plasma and lipopolysaccharide-induced splenocytes production of tumor necrosis factor-α in mice fed a high-fat, high-sucrose diet. J Funct Foods 2018. [DOI: 10.1016/j.jff.2018.07.043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
11
|
El-Shinnawy NA, Abd Elhalem SS, Haggag NZ, Badr G. Ameliorative role of camel whey protein and rosuvastatin on induced dyslipidemia in mice. Food Funct 2018; 9:1038-1047. [PMID: 29349446 DOI: 10.1039/c7fo01871a] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The incidence of obesity is rapidly increasing throughout the world. Dyslipidemia is a major risk factor for a number of chronic diseases, including diabetes and cardiovascular diseases. This work presents a novel approach to study the activity of camel whey protein (WP) with antioxidant and anti-inflammatory properties as a cheap dietary protein substance extracted from camel milk to produce satiety and help in building muscles. Mice model suffering from dyslipidemia as a result of feeding on high fat-cholesterol diet for 8 weeks were administrated with either camel WP and/or rosuvastatin for 4 weeks. Dyslipidemia revealed significant increase in anthropometrical measurements, levels of glucose, insulin, cholesterol, triglycerides, low-density lipoprotein, total leucocyte count, inflammatory cytokines and reactive oxygen species, accompanied by a significant elevation in activating transcription factor-3 and inducible nitric oxide synthase expressions. These alterations were correlated with a profound reduction in high-density lipoprotein, peroxisome proliferator-activated receptor alpha and adiponectin along with a decrease in liver and muscle mitochondrial proteins. Rosuvastatin treatment to mice suffering from dyslipidemia in combination with camel WP for 4 weeks ameliorated these parameters. Notably, animals treated with both camel WP and rosuvastatin exhibited a remarkable decrease in the incidence of dyslipidemia. In addition, camel WP succeeded to overcome the therapeutic drawback posed from rosuvastatin therapy alone with minimal side effects.
Collapse
Affiliation(s)
- Nashwa Ahmed El-Shinnawy
- Zoology Department, Faculty of Women for Arts, Science and Education, Ain Shams University, 11757 Cairo, Egypt.
| | | | | | | |
Collapse
|
12
|
Yu Q, Wang F, Meng X, Gong Y, Wang Y, Xu C, Wang S. Short‑term use of atorvastatin affects glucose homeostasis and suppresses the expression of LDL receptors in the pancreas of mice. Mol Med Rep 2018; 18:2780-2788. [PMID: 30015940 PMCID: PMC6102652 DOI: 10.3892/mmr.2018.9239] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 05/03/2018] [Indexed: 01/30/2023] Open
Abstract
Low-density lipoprotein receptors (LDLRs) may serve a role in the diabetogenic effect of statins; however, the effects of statins on LDLR expression and its regulation in the pancreas and islets have yet to be determined. To exclude the long-term effects of treatment with atorvastatin, which allows mice to adapt, male C57BL/j and apolipoprotein E-deficient mice were acutely treated with oral atorvastatin for 6 weeks, and glucose homeostasis and LDLR expression in the pancreas and islets were examined. In the present study, it was observed that the short-term use of atorvastatin affected insulin sensitivity in normal mice and glucose tolerance in hyperlipidemic mice. Furthermore, it was identified that 6 weeks of treatment with atorvastatin suppressed LDLR expression in the pancreas and pancreatic islets in C57BL/j mice, and an increase in proprotein convertase subtilisin/kexin type 9 expression was additionally observed in the pancreas. However, 6 weeks of treatment with atorvastatin did not affect LDLR expression in the pancreas of hyperlipidemic mice. It may be concluded that the short-term use of atorvastatin disturbs glucose homeostasis and suppresses LDLR expression in the pancreas and pancreatic islets in C57BL/j mice, suggesting that the role of LDLR in the diabetogenic effect of statins requires further investigation.
Collapse
Affiliation(s)
- Qi Yu
- Institute of Material Medical, School of Pharmacy, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Fang Wang
- Shaanxi Key Laboratory of Ischemic Cardiovascular Diseases and Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, Shaanxi 710021, P.R. China
| | - Xiaodong Meng
- Shaanxi Key Laboratory of Ischemic Cardiovascular Diseases and Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, Shaanxi 710021, P.R. China
| | - Yiren Gong
- Shaanxi Key Laboratory of Ischemic Cardiovascular Diseases and Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, Shaanxi 710021, P.R. China
| | - Yanli Wang
- Shaanxi Key Laboratory of Ischemic Cardiovascular Diseases and Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, Shaanxi 710021, P.R. China
| | - Cangbao Xu
- Shaanxi Key Laboratory of Ischemic Cardiovascular Diseases and Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, Shaanxi 710021, P.R. China
| | - Siwang Wang
- Institute of Material Medical, School of Pharmacy, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| |
Collapse
|
13
|
Wang K, Bao L, Zhou N, Zhang J, Liao M, Zheng Z, Wang Y, Liu C, Wang J, Wang L, Wang W, Liu S, Liu H. Structural Modification of Natural Product Ganomycin I Leading to Discovery of a α-Glucosidase and HMG-CoA Reductase Dual Inhibitor Improving Obesity and Metabolic Dysfunction in Vivo. J Med Chem 2018; 61:3609-3625. [DOI: 10.1021/acs.jmedchem.8b00107] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Kai Wang
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Li Bao
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | | | - Jinjin Zhang
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Mingfang Liao
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Zhongyong Zheng
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Yujing Wang
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | | | | | - Lifeng Wang
- Beijing Kangyuan Pharmaceutical Co., Ltd., No. 3 Changliu Road, Changping District, Beijing 102200, P. R. China
| | | | - ShuangJiang Liu
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Hongwei Liu
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| |
Collapse
|