1
|
Roy S, Ghosh A, Majie A, Karmakar V, Das S, Dinda SC, Bose A, Gorain B. Terpenoids as potential phytoconstituent in the treatment of diabetes: From preclinical to clinical advancement. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 129:155638. [PMID: 38728916 DOI: 10.1016/j.phymed.2024.155638] [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: 12/17/2023] [Revised: 03/21/2024] [Accepted: 04/13/2024] [Indexed: 05/12/2024]
Abstract
BACKGROUND Diabetes mellitus, a hyperglycemic condition associated with multitudinous organ dysfunction, is a hallmark of the metabolic disorder. This life-threatening condition affects millions of individuals globally, harming them financially, physically and psychologically in the course of therapy. PURPOSES The course therapy for illnesses has undergone ground-breaking transformations due to recent technical advances and insights. Alternatively, the administration of hyperglycemia-reducing agents results in several complications, including severe cardiovascular disease, kidney failure, hepatic problems, and several dermatological conditions. Consideration of alternate diabetic therapy having minimal side effects or no adverse reactions has been driven by such problems. STUDY DESIGN An extensive literature study was conducted in authoritative scientific databases such as PubMed, Scopus, and Web of Science to identify the studies elucidating the bioactivities of terpenoids in diabetic conditions. METHODS Keywords including 'terpenoids', 'monoterpenes', 'diterpenes', 'sesquiterpenes', 'diabetes', 'diabetes mellitus', 'clinical trials', 'preclinical studies', and 'increased blood glucose' were used to identify the relevant research articles. The exclusion criteria, such as English language, duplication, open access, abstract only, and studies not involving preclinical and clinical research, were set. Based on these criteria, 937 relevant articles were selected for further evaluation. RESULTS Triterpenes can serve as therapeutic agents for diabetic retinopathy, peripheral neuropathy, and kidney dysfunction by inhibiting several pathways linked to hyperglycemia and its complications. Therefore, it is essential to draw special attention to these compounds' therapeutic effectiveness and provide scientific professionals with novel data. CONCLUSION This study addressed recent progress in research focussing on mechanisms of terpenoid, its by-products, physiological actions, and therapeutic applications, particularly in diabetic and associated disorders.
Collapse
Affiliation(s)
- Sukanta Roy
- School of Pharmacy, The Neotia University, Diamond Harbour Rd, Sarisha, West Bengal, India
| | - Arya Ghosh
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, India
| | - Ankit Majie
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, India
| | - Varnita Karmakar
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, India
| | - Sourav Das
- School of Pharmacy, The Neotia University, Diamond Harbour Rd, Sarisha, West Bengal, India
| | - Subas Chandra Dinda
- School of Pharmacy, The Neotia University, Diamond Harbour Rd, Sarisha, West Bengal, India
| | - Anirbandeep Bose
- School of Medical Science, Adamas University, Barbaria, Jagannathpur, Kolkata, India.
| | - Bapi Gorain
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, India.
| |
Collapse
|
2
|
Oruc M, Gedik ME, Uner M, Ulug E, Unal RN, Gunaydin G, Dogrul AB. Effectiveness of metformin for the reversal of cold-ischemia-induced damage in hepatosteatosis. Clin Res Hepatol Gastroenterol 2024; 48:102314. [PMID: 38467276 DOI: 10.1016/j.clinre.2024.102314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 02/12/2024] [Accepted: 03/06/2024] [Indexed: 03/13/2024]
Abstract
BACKGROUND Primary dysfunction and rejection are more common in donor liver tissues with steatosis. AMP-activated protein kinase (AMPK) assumes organ-protective functions during ischemia. Metformin was used for the activation of AMPK in hepatocytes. The aim of this study is to investigate the effectiveness of metformin administration for the reversal of cold-ischemia-induced damage in hepatosteatosis. MATERIAL AND METHODS Seven-week-old C7BL56 male-mice (n = 109) were separated into four groups depending on diet type and metformin use. A specific diet model was followed for 10 weeks to induce hepatosteatosis. A group of the animals was administered with metformin for the last four weeks via oral gavage. After resection, the liver tissues were perfused and kept for 0-6-12-24 h in the UW solution. Histopathological examinations were performed, and Western blot was utilized to analyze p-AMPK and AMPK expression levels. RESULTS Hepatosteatosis decreased significantly with metformin. The steatotic liver group had more prominent pericentral inflammation, necrosis as well as showing a decreased and more delayed AMPK response than the non-fat group. All these alterations could be corrected using metformin. CONCLUSION Metformin can increase the resistance of livers with hepatosteatosis to cold-ischemia-induced damage, which in turn may pave the way for successful transplantation of fatty living-donor livers.
Collapse
Affiliation(s)
- Mustafa Oruc
- Department of General Surgery, Faculty Of Medicine, School of Medicine, Hacettepe University, Floor B, 06230, Ankara, Altindag 06230, Turkey
| | - Mustafa Emre Gedik
- Department of Basic Oncology, Hacettepe University Cancer Institute, Ankara 06230, Turkey
| | - Meral Uner
- Department of Pathology, Hacettepe University School of Medicine, Ankara 06230, Turkey
| | - Elif Ulug
- Department of Nutrition and Dietetics, Hacettepe University, Ankara 06230, Turkey
| | - Reyhan Nergiz Unal
- Department of Nutrition and Dietetics, Hacettepe University, Ankara 06230, Turkey
| | - Gurcan Gunaydin
- Department of Basic Oncology, Hacettepe University Cancer Institute, Ankara 06230, Turkey
| | - Ahmet Bulent Dogrul
- Department of General Surgery, Faculty Of Medicine, School of Medicine, Hacettepe University, Floor B, 06230, Ankara, Altindag 06230, Turkey.
| |
Collapse
|
3
|
Kim N, Jung S, Lee E, Jo EB, Yoon S, Jeong Y. Gryllus bimaculatus De Geer hydrolysates alleviate lipid accumulation, inflammation, and endoplasmic reticulum stress in palmitic acid-treated human hepatoma G2 cells. JOURNAL OF ETHNOPHARMACOLOGY 2022; 291:115117. [PMID: 35182670 DOI: 10.1016/j.jep.2022.115117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 02/05/2022] [Accepted: 02/14/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Nonalcoholic fatty liver disease (NAFLD) is one of the most common hepatic diseases closely intertwined with saturated fatty acids intake. Therefore, various studies are being conducted to find natural substances to prevent either the onset or progression of NAFLD. According to traditional medicinal literature, it has been reported that Gryllus bimaculatus De Geer (GB) has systemic detoxifying activity; however, the preventive effects of GB on NAFLD have not been elucidated to date. AIM OF STUDY To evaluate the potential of GB as a material for the mitigation of NAFLD, we investigated the effects of GB hydrolysates on the hepatic lipid accumulation, inflammation, and endoplasmic reticulum (ER) stress in human hepatoma G2 (Hep G2) cells treated with palmitic acid (PA). METHODS Steamed and dried GB was defatted, pulverized, and then lyophilized following hydrolyzation using Neutrase® (GB-N) or Flavourzyme® (GB-F). Hep G2 cells were incubated with GB-N or GB-F at various concentrations (0, 0.25, 0.5, and 1 mg/mL) for 24 h, and then PA was treated for another 24 h. RESULTS The GB-N and GB-F significantly prevented the PA-induced intracellular lipid accumulation in the human liver cells (p < 0.05). Moreover, the GB-N and GB-F increased the hepatic cellular viability against the PA-treatment (p < 0.05). In addition, the GB-N and GB-F significantly ameliorated the PA-inducible proinflammatory cytokines mRNA expression, such as tumor necrosis factor-α and interleukin-1β, compared to the PA-treated hepatic cells (p < 0.05). Furthermore, the GB-N and GB-F inhibited the PA-inducible lipogenic mRNA expression, such as fatty acid synthase, sterol regulatory element-binding protein 1c, and peroxisome proliferator-activated receptor-γ (p < 0.05). Moreover, the GB-N and GB-F alleviated the ER stress-related mRNA expression, such as glucose regulatory protein 78 and X-box binding protein increased in PA-treated cells (p < 0.05). CONCLUSIONS These results indicate that GB-N and GB-F could be used as materials to prevent the NAFLD onset or progression with alleviating hepatic lipid accumulation, inflammation, and ER stress.
Collapse
Affiliation(s)
- Nayeon Kim
- Department of Food Science and Nutrition, Dankook University, Cheonan, Chungnam, 31116, South Korea; Research Center for Industrialization of Natural Nutraceuticals, Dankook University, Cheonan, Chungnam, 31116, South Korea; R&D, Hanmi Natural Nutrition Co., Ltd., Paju, Gyeonggi, 10808, South Korea.
| | - Sunyoon Jung
- Department of Food Science and Nutrition, Dankook University, Cheonan, Chungnam, 31116, South Korea; Research Center for Industrialization of Natural Nutraceuticals, Dankook University, Cheonan, Chungnam, 31116, South Korea.
| | - Eunjung Lee
- Department of Food Science and Nutrition, Dankook University, Cheonan, Chungnam, 31116, South Korea; Research Center for Industrialization of Natural Nutraceuticals, Dankook University, Cheonan, Chungnam, 31116, South Korea.
| | - Eun-Byeol Jo
- Department of Food Science and Nutrition, Dankook University, Cheonan, Chungnam, 31116, South Korea; Research Center for Industrialization of Natural Nutraceuticals, Dankook University, Cheonan, Chungnam, 31116, South Korea.
| | - Seongjun Yoon
- Department of Baking Science, Hyejeon College, Hongsung, Chungnam, 32244, South Korea.
| | - Yoonhwa Jeong
- Department of Food Science and Nutrition, Dankook University, Cheonan, Chungnam, 31116, South Korea; Research Center for Industrialization of Natural Nutraceuticals, Dankook University, Cheonan, Chungnam, 31116, South Korea.
| |
Collapse
|
4
|
Low-dose metformin targets the lysosomal AMPK pathway through PEN2. Nature 2022; 603:159-165. [PMID: 35197629 PMCID: PMC8891018 DOI: 10.1038/s41586-022-04431-8] [Citation(s) in RCA: 235] [Impact Index Per Article: 117.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 01/10/2022] [Indexed: 12/14/2022]
Abstract
Metformin, the most prescribed antidiabetic medicine, has shown other benefits such as anti-ageing and anticancer effects1-4. For clinical doses of metformin, AMP-activated protein kinase (AMPK) has a major role in its mechanism of action4,5; however, the direct molecular target of metformin remains unknown. Here we show that clinically relevant concentrations of metformin inhibit the lysosomal proton pump v-ATPase, which is a central node for AMPK activation following glucose starvation6. We synthesize a photoactive metformin probe and identify PEN2, a subunit of γ-secretase7, as a binding partner of metformin with a dissociation constant at micromolar levels. Metformin-bound PEN2 forms a complex with ATP6AP1, a subunit of the v-ATPase8, which leads to the inhibition of v-ATPase and the activation of AMPK without effects on cellular AMP levels. Knockout of PEN2 or re-introduction of a PEN2 mutant that does not bind ATP6AP1 blunts AMPK activation. In vivo, liver-specific knockout of Pen2 abolishes metformin-mediated reduction of hepatic fat content, whereas intestine-specific knockout of Pen2 impairs its glucose-lowering effects. Furthermore, knockdown of pen-2 in Caenorhabditis elegans abrogates metformin-induced extension of lifespan. Together, these findings reveal that metformin binds PEN2 and initiates a signalling route that intersects, through ATP6AP1, the lysosomal glucose-sensing pathway for AMPK activation. This ensures that metformin exerts its therapeutic benefits in patients without substantial adverse effects.
Collapse
|
5
|
Mandwie M, Karunia J, Niaz A, Keay KA, Musumeci G, Rennie C, McGrath K, Al-Badri G, Castorina A. Metformin Treatment Attenuates Brain Inflammation and Rescues PACAP/VIP Neuropeptide Alterations in Mice Fed a High-Fat Diet. Int J Mol Sci 2021; 22:ijms222413660. [PMID: 34948457 PMCID: PMC8706124 DOI: 10.3390/ijms222413660] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 12/28/2022] Open
Abstract
High-fat diet (HFD)-induced comorbid cognitive and behavioural impairments are thought to be the result of persistent low-grade neuroinflammation. Metformin, a first-line medication for the treatment of type-2 diabetes, seems to ameliorate these comorbidities, but the underlying mechanism(s) are not clear. Pituitary adenylate cyclase-activating peptide (PACAP) and vasoactive intestinal peptide (VIP) are neuroprotective peptides endowed with anti-inflammatory properties. Alterations to the PACAP/VIP system could be pivotal during the development of HFD-induced neuroinflammation. To unveil the pathogenic mechanisms underlying HFD-induced neuroinflammation and assess metformin’s therapeutic activities, (1) we determined if HFD-induced proinflammatory activity was present in vulnerable brain regions associated with the development of comorbid behaviors, (2) investigated if the PACAP/VIP system is altered by HFD, and (3) assessed if metformin rescues such diet-induced neurochemical alterations. C57BL/6J male mice were divided into two groups to receive either standard chow (SC) or HFD for 16 weeks. A further HFD group received metformin (HFD + M) (300 mg/kg BW daily for 5 weeks) via oral gavage. Body weight, fasting glucose, and insulin levels were measured. After 16 weeks, the proinflammatory profile, glial activation markers, and changes within the PI3K/AKT intracellular pathway and the PACAP/VIP system were evaluated by real-time qPCR and/or Western blot in the hypothalamus, hippocampus, prefrontal cortex, and amygdala. Our data showed that HFD causes widespread low-grade neuroinflammation and gliosis, with regional-specific differences across brain regions. HFD also diminished phospho-AKT(Ser473) expression and caused significant disruptions to the PACAP/VIP system. Treatment with metformin attenuated these neuroinflammatory signatures and reversed PI3K/AKT and PACAP/VIP alterations caused by HFD. Altogether, our findings demonstrate that metformin treatment rescues HFD-induced neuroinflammation in vulnerable brain regions, most likely by a mechanism involving the reinstatement of PACAP/VIP system homeostasis. Data also suggests that the PI3K/AKT pathway, at least in part, mediates some of metformin’s beneficial effects.
Collapse
Affiliation(s)
- Mawj Mandwie
- Laboratory of Cellular and Molecular Neuroscience (LCMN), School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia; (M.M.); (J.K.); (A.N.); (G.A.-B.)
| | - Jocelyn Karunia
- Laboratory of Cellular and Molecular Neuroscience (LCMN), School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia; (M.M.); (J.K.); (A.N.); (G.A.-B.)
| | - Aram Niaz
- Laboratory of Cellular and Molecular Neuroscience (LCMN), School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia; (M.M.); (J.K.); (A.N.); (G.A.-B.)
| | - Kevin A. Keay
- Laboratory of Neural Structure and Function, School of Medical Science (Neuroscience), University of Sydney, Sydney, NSW 2006, Australia;
| | - Giuseppe Musumeci
- Department of Biomedical and Biotechnological Sciences, Anatomy, Histology and Movement Sciences Section, School of Medicine, University of Catania, 95125 Catania, Italy;
| | - Claire Rennie
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia; (C.R.); (K.M.)
| | - Kristine McGrath
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia; (C.R.); (K.M.)
| | - Ghaith Al-Badri
- Laboratory of Cellular and Molecular Neuroscience (LCMN), School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia; (M.M.); (J.K.); (A.N.); (G.A.-B.)
| | - Alessandro Castorina
- Laboratory of Cellular and Molecular Neuroscience (LCMN), School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia; (M.M.); (J.K.); (A.N.); (G.A.-B.)
- Laboratory of Neural Structure and Function, School of Medical Science (Neuroscience), University of Sydney, Sydney, NSW 2006, Australia;
- Correspondence:
| |
Collapse
|
6
|
Liu Y, Wen H, Wu X, Wu M, Liu L, Wang J, Huo G, Lyu J, Xie L, Dan M. The Bio-Persistence of Reversible Inflammatory, Histological Changes and Metabolic Profile Alterations in Rat Livers after Silver/Gold Nanorod Administration. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:2656. [PMID: 34685095 PMCID: PMC8538332 DOI: 10.3390/nano11102656] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/26/2021] [Accepted: 10/02/2021] [Indexed: 11/16/2022]
Abstract
As a widely applied nanomaterial, silver nanomaterials (AgNMs) have increased public concern about their potential adverse biological effects. However, there are few related researches on the long-term toxicity, especially on the reversibility of AgNMs in vivo. In the current study, this issue was tackled by exploring liver damage after an intravenous injection of silver nanorods with golden cores (Au@AgNRs) and its potential recovery in a relatively long term (8 w). After the administration of Au@AgNRs into rats, Ag was found to be rapidly cleared from blood within 10 min and mainly accumulated in liver as well as spleen until 8 w. All detected parameters almost displayed a two-stage response to Au@AgNRs administration, including biological markers, histological changes and metabolic variations. For the short-term (2 w) responses, some toxicological parameters (hematological changes, cytokines, liver damages etc.) significantly changed compared to control and AuNRs group. However, after a 6-week recovery, all abovementioned changes mostly returned to the normal levels in the Au@AgNRs group. These indicated that after a lengthy period, acute bioeffects elicited by AgNMs could be followed by the adaptive recovery, which will provide a novel and valuable toxicity mechanism of AgNMs for potential biomedical applications of AgNMs.
Collapse
Affiliation(s)
- Ying Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, NCNST-NIFDC Joint Laboratory for Measurement and Evaluation of Nanomaterials in Medical Applications, Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, No. 11 Beiyitiao Zhongguancun, Haidian District, Beijing 100190, China;
| | - Hairuo Wen
- National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, No. 8 Hongda Mid-Road, Beijing Economic and Technological Development Zone, Daxing District, Beijing 100176, China; (H.W.); (J.W.); (G.H.); or (J.L.)
| | - Xiaochun Wu
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, NCNST-NIFDC Joint Laboratory for Measurement and Evaluation of Nanomaterials in Medical Applications, Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, No. 11 Beiyitiao Zhongguancun, Haidian District, Beijing 100190, China; (X.W.); (M.W.); (L.L.)
| | - Meiyu Wu
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, NCNST-NIFDC Joint Laboratory for Measurement and Evaluation of Nanomaterials in Medical Applications, Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, No. 11 Beiyitiao Zhongguancun, Haidian District, Beijing 100190, China; (X.W.); (M.W.); (L.L.)
| | - Lin Liu
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, NCNST-NIFDC Joint Laboratory for Measurement and Evaluation of Nanomaterials in Medical Applications, Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, No. 11 Beiyitiao Zhongguancun, Haidian District, Beijing 100190, China; (X.W.); (M.W.); (L.L.)
| | - Jiahui Wang
- National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, No. 8 Hongda Mid-Road, Beijing Economic and Technological Development Zone, Daxing District, Beijing 100176, China; (H.W.); (J.W.); (G.H.); or (J.L.)
| | - Guitao Huo
- National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, No. 8 Hongda Mid-Road, Beijing Economic and Technological Development Zone, Daxing District, Beijing 100176, China; (H.W.); (J.W.); (G.H.); or (J.L.)
| | - Jianjun Lyu
- National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, No. 8 Hongda Mid-Road, Beijing Economic and Technological Development Zone, Daxing District, Beijing 100176, China; (H.W.); (J.W.); (G.H.); or (J.L.)
- Department of Pathology, InnoStar Bio-Tech Nantong Co., Ltd., Nantong 226133, China
| | - Liming Xie
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, NCNST-NIFDC Joint Laboratory for Measurement and Evaluation of Nanomaterials in Medical Applications, Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, No. 11 Beiyitiao Zhongguancun, Haidian District, Beijing 100190, China; (X.W.); (M.W.); (L.L.)
- School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mo Dan
- National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, No. 8 Hongda Mid-Road, Beijing Economic and Technological Development Zone, Daxing District, Beijing 100176, China; (H.W.); (J.W.); (G.H.); or (J.L.)
- The State Key Laboratory of New Pharmaceutical Preparations and Excipients, 226 Huanghe Road, Shijiazhuang 050035, China
| |
Collapse
|
7
|
Sharma M, Premkumar M, Kulkarni AV, Kumar P, Reddy DN, Rao NP. Drugs for Non-alcoholic Steatohepatitis (NASH): Quest for the Holy Grail. J Clin Transl Hepatol 2021; 9:40-50. [PMID: 33604254 PMCID: PMC7868704 DOI: 10.14218/jcth.2020.00055] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 10/29/2020] [Accepted: 11/12/2020] [Indexed: 12/12/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a global epidemic that is likely to become the most common cause of chronic liver disease in the next decade, worldwide. Though numerous drugs have been evaluated in clinical trials, most of them have returned inconclusive results and shown poorly-tolerated adverse effects. None of the drugs have been approved by the Food and Drug Administration for treating biopsy-proven non-alcoholic steatohepatitis (NASH). Vitamin E and pioglitazone have been extensively used in treatment of biopsy-proven nondiabetic NASH patients. Although some amelioration of inflammation has been seen, these drugs did not improve the fibrosis component of NASH. Therefore, dietary modification and weight reduction have remained the cornerstone of treatment of NASH; moreover, they have shown to improve histological activity as well as fibrosis. The search for an ideal drug or 'Holy Grail' within this landscape of possible agents continues, as weight reduction is achieved only in less than 10% of patients. In this current review, we summarize the drugs for NASH which are under investigation, and we provide a critical analysis of their up-to-date results and outcomes.
Collapse
Affiliation(s)
- Mithun Sharma
- Department of Hepatology, Asian Institute of Gastroenterology, Hyderabad, India
| | | | - Anand V Kulkarni
- Department of Hepatology, Asian Institute of Gastroenterology, Hyderabad, India
- Correspondence to: Dr. Anand V Kulkarni, Department of Hepatology and Liver Transplantation, Asian Institute of Gastroenterology, Hyderabad, India. Tel: +91-40-42444222, E-mail:
| | - Pramod Kumar
- Department of Hepatology, Asian Institute of Gastroenterology, Hyderabad, India
| | - D Nageshwar Reddy
- Department of Gastroenterology, Asian Institute of Gastroenterology, Hyderabad, India
| | - Nagaraja Padaki Rao
- Department of Hepatology, Asian Institute of Gastroenterology, Hyderabad, India
| |
Collapse
|
8
|
Budhwar S, Chakraborty M, Sethi K, Chatterjee A. Antidiabetic properties of rice and wheat bran-A review. J Food Biochem 2020; 44:e13424. [PMID: 32761956 DOI: 10.1111/jfbc.13424] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 07/16/2020] [Accepted: 07/16/2020] [Indexed: 01/09/2023]
Abstract
In the present study, the increasing demand for the by-products of rice and wheat, especially their bran part obtained by milling has been discussed along with their properties in controlling diabetes. It is composed of macronutrients and micronutrients, including fibers, and trace elements and different phytochemicals. Previously, they were being used as animal fodder or for other compost matter. Contrarily, it can be utilized for humankind to save world hunger and to cater to the need for extra food demand and eradication of malnourishment, particularly in the developing countries. The bran part can act as a defense against different chronic diseases, particularly diabetes, which accounts for 3.2 million deaths worldwide every year. Keeping this in view, the current review discusses the nutritional composition, biological, and therapeutic properties of rice and wheat bran. PRACTICAL APPLICATIONS: Properly processed agricultural wastes can yield resourceful and economical by-products. The bran part of rice and wheat is such an agricultural byproduct which is cheap and easily available. They contain vast amount of beneficial biochemical constituents. Properly processed bran part can be utilized for preparation of various value-added food products which can save the world hunger, extra food demand, and malnourishment and will be a boon for the developing countries. It can be also useful in combating several chronic diseases including diabetes through dietary intake.
Collapse
Affiliation(s)
- Savita Budhwar
- Department of Nutrition Biology, School of Interdisciplinary and Applied Life Sciences, Central University of Haryana, Mahendergarh, India
| | - Manali Chakraborty
- Department of Nutrition Biology, School of Interdisciplinary and Applied Life Sciences, Central University of Haryana, Mahendergarh, India
| | - Kashika Sethi
- Department of Nutrition Biology, School of Interdisciplinary and Applied Life Sciences, Central University of Haryana, Mahendergarh, India
| | - Arnab Chatterjee
- Department of Nutrition Biology, School of Interdisciplinary and Applied Life Sciences, Central University of Haryana, Mahendergarh, India
| |
Collapse
|
9
|
Woo M, Noh JS. Regulatory Effects of Skate Skin-Derived Collagen Peptides with Different Molecular Weights on Lipid Metabolism in the Liver and Adipose Tissue. Biomedicines 2020; 8:biomedicines8070187. [PMID: 32630126 PMCID: PMC7400643 DOI: 10.3390/biomedicines8070187] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/27/2020] [Accepted: 06/29/2020] [Indexed: 11/16/2022] Open
Abstract
This study investigated the effects of skate skin collagen peptide (SSCP) with different molecular weights (MWs) on the lipid metabolism in the liver and adipose tissue. Male db/db mice were orally administered with water (control group) or low SSCP (LCP group) or high SSCP (HCP group) MW for 8 weeks whereas male m/m mice were used for comparison (normal group) (n = 10 each group). Compared to the control group, the LCP and HCP groups had lower adipose tissue mass, plasma and hepatic lipid concentrations, and plasma leptin levels (p < 0.05). Protein expression levels of lipogenesis-related protein were reduced in both liver and adipose tissues of SSCP-fed groups whereas those for lipolysis were elevated (p < 0.05). In particular, the LCP had the higher effects relative to the HCP. The above results were supported by histological analysis, revealing that SSCP administration decreased the size of adipose droplets and suppressed hepatic lipid accumulation. Our results showed that SSCP has potential antiobesity properties through the improvement of lipid metabolism in the liver and adipose tissue; in particular, the lower MW of collagen peptide had the greater effects.
Collapse
Affiliation(s)
- Minji Woo
- Busan Innovation Institute of Industry, Science & Technology Planning (BISTEP), Busan 48058, Korea;
- Department of Food Science and Nutrition and Kimchi Research Institute, Pusan National University, Busan 46241, Korea
| | - Jeong Sook Noh
- Department of Food Science and Nutrition, Tongmyong University, Busan 48520, Korea
- Correspondence: ; Tel.: +82-51-629-1716; Fax: +82-51-629-1709
| |
Collapse
|
10
|
Zhang L, Li F, Guo Q, Duan Y, Wang W, Zhong Y, Yang Y, Yin Y. Leucine Supplementation: A Novel Strategy for Modulating Lipid Metabolism and Energy Homeostasis. Nutrients 2020; 12:E1299. [PMID: 32370170 PMCID: PMC7282259 DOI: 10.3390/nu12051299] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 04/25/2020] [Accepted: 04/27/2020] [Indexed: 02/07/2023] Open
Abstract
Lipid metabolism is an important and complex biochemical process involved in the storage of energy and maintenance of normal biological functions. Leucine, a branched amino acid, has anti-obesity effects on glucose tolerance, lipid metabolism, and insulin sensitivity. Leucine also modulates mitochondrial dysfunction, representing a new strategy to target aging, neurodegenerative disease, obesity, diabetes, and cardiovascular disease. Although various studies have been carried out, much uncertainty still exists and further studies are required to fully elucidate the relationship between leucine and lipid metabolism. This review offers an up-to-date report on leucine, as key roles in both lipid metabolism and energy homeostasis in vivo and in vitro by acceleration of fatty acid oxidation, lipolysis, activation of the adenosine 5'-monophosphate-activated protein kinase (AMPK)-silent information regulator of transcription 1 (SIRT1)-proliferator-activated receptor γ coactivator-1α (PGC-1α) pathway, synthesis, and/or secretion of adipokines and stability of the gut microbiota.
Collapse
Affiliation(s)
- Lingyu Zhang
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; (L.Z.); (Q.G.); (Y.D.); (W.W.); (Y.Y.)
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100039, China
| | - Fengna Li
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; (L.Z.); (Q.G.); (Y.D.); (W.W.); (Y.Y.)
| | - Qiuping Guo
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; (L.Z.); (Q.G.); (Y.D.); (W.W.); (Y.Y.)
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100039, China
| | - Yehui Duan
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; (L.Z.); (Q.G.); (Y.D.); (W.W.); (Y.Y.)
| | - Wenlong Wang
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; (L.Z.); (Q.G.); (Y.D.); (W.W.); (Y.Y.)
- Laboratory of Animal Nutrition and Human Health, School of Biology, Hunan Normal University, Changsha 410018, China
| | - Yinzhao Zhong
- Guangdong Provincial Key Laboratory of Animal Nutrition Regulation, South China Agricultural University, Guangzhou 510642, China;
| | - Yuhuan Yang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China;
| | - Yulong Yin
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; (L.Z.); (Q.G.); (Y.D.); (W.W.); (Y.Y.)
| |
Collapse
|
11
|
Administration of Nicotinamide Mononucleotide (NMN) Reduces Metabolic Impairment in Male Mouse Offspring from Obese Mothers. Cells 2020; 9:cells9040791. [PMID: 32218167 PMCID: PMC7226525 DOI: 10.3390/cells9040791] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 03/19/2020] [Accepted: 03/23/2020] [Indexed: 12/20/2022] Open
Abstract
Maternal obesity impacts offspring metabolism. We sought to boost mitochondrial energy metabolism using the nicotinamide adenine dinucleotide (NAD+) precursor nicotinamide mononucleotide (NMN) to treat metabolic impairment induced by maternal and long-term post weaning over-nutrition. Male offspring of lean or obese mothers, fed chow or high fat diet (HFD) for 30 weeks post-weaning, were given NMN injection, starting at 31 weeks of age, daily for 3 weeks before sacrifice. Glucose tolerance was tested at 10, 29 and 32 weeks of age to measure short and long term effects of post-weaning HFD, and NMN treatment. Plasma insulin and triglycerides, liver triglycerides and expression of mitochondrial metabolism-related genes were measured at 34 weeks. Impaired glucose tolerance due to maternal and post weaning HFD was significantly improved by only 8 days of NMN treatment. Furthermore, in offspring of obese mothers hepatic lipid accumulation was reduced due to NMN treatment by 50% and 23% in chow and HFD fed offspring respectively. Hepatic genes involved in fat synthesis, transport and uptake were reduced, while those involved in fatty acid oxidation were increased by NMN. Overall this finding suggests short term administration of NMN could be a therapeutic approach for treating metabolic disease due to maternal and post weaning over-nutrition, even in late adulthood.
Collapse
|
12
|
Ma C, Liu Y, Liu S, Lévesque CL, Zhao F, Yin J, Dong B. Branched chain amino acids alter fatty acid profile in colostrum of sows fed a high fat diet. J Anim Sci Biotechnol 2020; 11:9. [PMID: 32095236 PMCID: PMC7025410 DOI: 10.1186/s40104-019-0423-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 12/24/2019] [Indexed: 02/02/2023] Open
Abstract
Background Branched chain amino acids (BCAAs) are important substrates for milk protein synthesis in the mammary gland, and are tightly related to lipid metabolism. No study has been performed examining the role of BCAAs with high fat diets on milk fat synthesis. This study was designed to investigate the effect of dietary BCAAs on growth performance of piglets, progeny body weight, and milk fat composition in sows fed a high fat diet. Four diets (CON = control diet; HF = high fat diet with 8% soybean oil; HF-MB=HF plus 0.39% BCAAs; HF-HB=HF plus 0.78% BCAAs) were fed to sows from late gestation to weaning. Results Compared to HF, BCAAs (HF-MB and HF-HB) increased the litter weight (P < 0.05) and overall litter weight gain (P < 0.05) at weaning and increased colostrum fat content by 27.3–35.8% (P < 0.01). Fatty acid profiles between the two doses of BCAAs were similar. Compared with HF, HF-MB tended to decrease the percentage of C18:3n3 (P = 0.063) and increased the percentage of C18:1n9c (P = 0.03). In addition, BCAAs in HF-MB increased the concentration of total fatty acid by 22.1% in colostrum (P = 0.03) but decreased that in serum at parturition by 53.2% (P = 0.027). The fatty acids in colostrum that increased with BCAAs were C15:0, C17:0, C20:3n6, C20:4n6, C20:5n3 and C22:6n3 (P = 0.00~0.04). Colostrum fatty acids of C20:0, C21:0, C22:0, C16:1, C20:1, C18:1n9c also tended to be increased (0.05 < P < 0.1) with BCAAs. The change in sow serum fatty acid profile due to BCAAs was different from that in colostrum. Conclusions BCAAs in high fat diet of sows altered the fatty acid composition in colostrum and enhanced litter growth. Our study indicated that BCAAs supplementation can enhance mammary fatty acid uptake and mammary fat synthesis and that supplemental BCAAs and fat in late gestation and lactation diets for sows can improve reproductive performance.
Collapse
Affiliation(s)
- Chang Ma
- 1State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 China
| | - Yajng Liu
- 1State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 China
| | - Shaoshuai Liu
- 1State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 China
| | - Crystal L Lévesque
- 2Department of Animal Science, College of Agriculture and Biological Sciences, South Dakota State University, Brookings, SD 57007 USA
| | - Fengqi Zhao
- 3Department of Animal and Veterinary Sciences, University of Vermont, Burlington, VT 05405 USA
| | - Jindong Yin
- 1State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 China
| | - Bing Dong
- 1State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 China
| |
Collapse
|
13
|
Ma Q, Zhou X, Hu L, Chen J, Zhu J, Shan A. Leucine and isoleucine have similar effects on reducing lipid accumulation, improving insulin sensitivity and increasing the browning of WAT in high-fat diet-induced obese mice. Food Funct 2020; 11:2279-2290. [DOI: 10.1039/c9fo03084k] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Leucine (Leu) and isoleucine (Ile) have similar effects in the management of obesity and related disorders.
Collapse
Affiliation(s)
- Qingquan Ma
- Institute of Animal Nutrition
- Northeast Agricultural University
- Harbin
- China
| | - Xinbo Zhou
- Institute of Animal Nutrition
- Northeast Agricultural University
- Harbin
- China
| | - Linlin Hu
- Institute of Animal Nutrition
- Northeast Agricultural University
- Harbin
- China
| | - Jiayi Chen
- Institute of Animal Nutrition
- Northeast Agricultural University
- Harbin
- China
| | - Jialiang Zhu
- Institute of Animal Nutrition
- Northeast Agricultural University
- Harbin
- China
| | - Anshan Shan
- Institute of Animal Nutrition
- Northeast Agricultural University
- Harbin
- China
| |
Collapse
|
14
|
Laskar J, Sengupta M, Choudhury Y. Treatment with the anti-diabetic drug metformin ameliorates betel-nut induced carcinogenesis in a murine model. Pharmacol Rep 2019; 71:1115-1124. [PMID: 31645006 DOI: 10.1016/j.pharep.2019.06.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 05/10/2019] [Accepted: 06/28/2019] [Indexed: 01/18/2023]
Abstract
BACKGROUND Metformin, a widely used anti-diabetic drug has gained enormous attention as an anticancer agent. This study seeks to investigate the efficacy of metformin in ameliorating aqueous extract of betel-nut (AEBN) and arecoline induced carcinogenesis in a murine model. METHODS Swiss albino mice were exposed to AEBN (2 mg ml-1) and arecoline (10 μg ml-1) in drinking water for 16 weeks followed by co-administration of metformin (75 mg kg-1 or 150 mg kg-1) for 4 or 8 weeks. Histological changes and oxidative stress were assessed by haematoxylin and eosin staining, TBARS assay and protein carbonylation assay respectively. Lipid profile was determined using an automated analyzer. Expression of total and phosphorylated AMPK, ACC and p53 were determined by immunoblotting. RESULTS AEBN and arecoline induced dyslipidemia by downregulating AMPK (Thr-172) and activating ACC (Ser-79); they also downregulated tumor suppressor p53 (Ser-15). Metformin treatment induced AMPK-dependent alleviation of dyslipidemia in a dose and time dependent manner, upregulated p53 (Ser-15), restored tissue architecture and reduced oxidative stress in tissues of AEBN and arecoline treated mice. CONCLUSION This study establishes that betel nut induces dyslipidemia through its alkaloid, arecoline by inhibition of AMPK (Thr-172) and activation of ACC (Ser-79) and highlights the therapeutic potential of metformin for treatment of betel-nut induced carcinogenesis, indicating the repurposing of the old drug in a new avenue.
Collapse
Affiliation(s)
- Jeny Laskar
- Department of Biotechnology, Assam University, Silchar, India
| | - Mahuya Sengupta
- Department of Biotechnology, Assam University, Silchar, India
| | | |
Collapse
|
15
|
Ginseng berry extract enhances metformin efficacy against obesity and hepatic steatosis in mice fed high-fat diet through increase of metformin uptake in liver. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.103551] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
16
|
Banerjee J, Bruckbauer A, Thorpe T, Zemel MB. Biphasic Effect of Sildenafil on Energy Sensing is Mediated by Phosphodiesterases 2 and 3 in Adipocytes and Hepatocytes. Int J Mol Sci 2019; 20:ijms20122992. [PMID: 31248114 PMCID: PMC6627652 DOI: 10.3390/ijms20122992] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 06/17/2019] [Indexed: 12/14/2022] Open
Abstract
Sirt1 (Sirtuin 1), AMPK (AMP-activated protein kinase), and eNOS (endothelial nitric oxide synthase) modulate hepatic energy metabolism and inflammation and play a major role in the development of NASH. Cyclic nucleotide phosphodiesterases (PDEs) play an important role in signal transduction by modulating intracellular levels of cyclic nucleotides. We previously found the PDE5 inhibitor sildenafil to synergize with leucine and leucine-metformin combinations in preclinical studies of NASH and obesity. However, efficacy is diminished at higher sildenafil concentrations. Herein, we have successfully modeled the U-shaped sildenafil dose-response in vitro and utilized this model to assess potential mechanisms of this dose-response relationship. Adipocytes and liver cells were treated with leucine (0.5 mM) and different concentrations of sildenafil (1 nM to 100 µM). cAMP, cGMP, and P-AMPK protein expression were used to demonstrate the biphasic response for increasing concentrations of sildenafil. The reversal with higher sildenafil levels was blunted by PDE2 inhibition. These data indicate that sildenafil-mediated increases in cGMP inhibits PDE3 at lower concentrations, which increases cAMP. However, further increases in cGMP from higher sildenafil concentrations activate PDE2 and consequently decrease cAMP, which demonstrates crosstalk between cAMP and cGMP via PDE2, PDE3, and PDE5. These changes in cAMP concentration are further reflected in downstream effects, including AMPK activation.
Collapse
Affiliation(s)
- Jheelam Banerjee
- NuSirt Biopharma Inc., 11020 Solway School Rd, Knoxville, TN 37931, USA.
| | - Antje Bruckbauer
- NuSirt Biopharma Inc., 11020 Solway School Rd, Knoxville, TN 37931, USA.
| | - Teresa Thorpe
- NuSirt Biopharma Inc., 11020 Solway School Rd, Knoxville, TN 37931, USA.
| | - Michael B Zemel
- NuSirt Biopharma Inc., 11020 Solway School Rd, Knoxville, TN 37931, USA.
| |
Collapse
|
17
|
Duan Y, Zhong Y, Xiao H, Zheng C, Song B, Wang W, Guo Q, Li Y, Han H, Gao J, Xu K, Li T, Yin Y, Li F, Yin J, Kong X. Gut microbiota mediates the protective effects of dietary β‐hydroxy‐β‐methylbutyrate (HMB) against obesity induced by high‐fat diets. FASEB J 2019; 33:10019-10033. [DOI: 10.1096/fj.201900665rr] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- Yehui Duan
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic ProcessKey Laboratory of Agro‐ecological Processes in Subtropical RegionInstitute of Subtropical AgricultureChinese Academy of SciencesHunan Provincial Engineering Research Center for Healthy Livestock and Poultry ProductionScientific Observing and Experimental Station of Animal Nutrition and Feed Science in South‐CentralMinistry of Agriculture Changsha China
| | - Yinzhao Zhong
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic ProcessKey Laboratory of Agro‐ecological Processes in Subtropical RegionInstitute of Subtropical AgricultureChinese Academy of SciencesHunan Provincial Engineering Research Center for Healthy Livestock and Poultry ProductionScientific Observing and Experimental Station of Animal Nutrition and Feed Science in South‐CentralMinistry of Agriculture Changsha China
- Guangdong Provincial Key Laboratory of Animal Nutrition RegulationSouth China Agricultural University Guangzhou China
| | - Hao Xiao
- Guangdong Academy of Agricultural SciencesKey Laboratory of Animal Nutrition and Feed Science in South ChinaInstitute of Animal ScienceMinistry of Agriculture Guangzhou China
| | - Changbing Zheng
- Guangdong Provincial Key Laboratory of Animal Nutrition RegulationSouth China Agricultural University Guangzhou China
| | - Bo Song
- Guangdong Provincial Key Laboratory of Animal Nutrition RegulationSouth China Agricultural University Guangzhou China
| | - Wenlong Wang
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic ProcessKey Laboratory of Agro‐ecological Processes in Subtropical RegionInstitute of Subtropical AgricultureChinese Academy of SciencesHunan Provincial Engineering Research Center for Healthy Livestock and Poultry ProductionScientific Observing and Experimental Station of Animal Nutrition and Feed Science in South‐CentralMinistry of Agriculture Changsha China
| | - Qiuping Guo
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic ProcessKey Laboratory of Agro‐ecological Processes in Subtropical RegionInstitute of Subtropical AgricultureChinese Academy of SciencesHunan Provincial Engineering Research Center for Healthy Livestock and Poultry ProductionScientific Observing and Experimental Station of Animal Nutrition and Feed Science in South‐CentralMinistry of Agriculture Changsha China
| | - Yuying Li
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic ProcessKey Laboratory of Agro‐ecological Processes in Subtropical RegionInstitute of Subtropical AgricultureChinese Academy of SciencesHunan Provincial Engineering Research Center for Healthy Livestock and Poultry ProductionScientific Observing and Experimental Station of Animal Nutrition and Feed Science in South‐CentralMinistry of Agriculture Changsha China
| | - Hui Han
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic ProcessKey Laboratory of Agro‐ecological Processes in Subtropical RegionInstitute of Subtropical AgricultureChinese Academy of SciencesHunan Provincial Engineering Research Center for Healthy Livestock and Poultry ProductionScientific Observing and Experimental Station of Animal Nutrition and Feed Science in South‐CentralMinistry of Agriculture Changsha China
| | - Jing Gao
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic ProcessKey Laboratory of Agro‐ecological Processes in Subtropical RegionInstitute of Subtropical AgricultureChinese Academy of SciencesHunan Provincial Engineering Research Center for Healthy Livestock and Poultry ProductionScientific Observing and Experimental Station of Animal Nutrition and Feed Science in South‐CentralMinistry of Agriculture Changsha China
| | - Kang Xu
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic ProcessKey Laboratory of Agro‐ecological Processes in Subtropical RegionInstitute of Subtropical AgricultureChinese Academy of SciencesHunan Provincial Engineering Research Center for Healthy Livestock and Poultry ProductionScientific Observing and Experimental Station of Animal Nutrition and Feed Science in South‐CentralMinistry of Agriculture Changsha China
| | - Tiejun Li
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic ProcessKey Laboratory of Agro‐ecological Processes in Subtropical RegionInstitute of Subtropical AgricultureChinese Academy of SciencesHunan Provincial Engineering Research Center for Healthy Livestock and Poultry ProductionScientific Observing and Experimental Station of Animal Nutrition and Feed Science in South‐CentralMinistry of Agriculture Changsha China
| | - Yulong Yin
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic ProcessKey Laboratory of Agro‐ecological Processes in Subtropical RegionInstitute of Subtropical AgricultureChinese Academy of SciencesHunan Provincial Engineering Research Center for Healthy Livestock and Poultry ProductionScientific Observing and Experimental Station of Animal Nutrition and Feed Science in South‐CentralMinistry of Agriculture Changsha China
- Guangdong Provincial Key Laboratory of Animal Nutrition RegulationSouth China Agricultural University Guangzhou China
| | - Fengna Li
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic ProcessKey Laboratory of Agro‐ecological Processes in Subtropical RegionInstitute of Subtropical AgricultureChinese Academy of SciencesHunan Provincial Engineering Research Center for Healthy Livestock and Poultry ProductionScientific Observing and Experimental Station of Animal Nutrition and Feed Science in South‐CentralMinistry of Agriculture Changsha China
- Hunan Collaborative Innovation Center for Utilization of Botanical Functional IngredientsHunan Co‐Innovation Center of Animal Production Safety (CICAPS) Changsha China
| | - Jie Yin
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic ProcessKey Laboratory of Agro‐ecological Processes in Subtropical RegionInstitute of Subtropical AgricultureChinese Academy of SciencesHunan Provincial Engineering Research Center for Healthy Livestock and Poultry ProductionScientific Observing and Experimental Station of Animal Nutrition and Feed Science in South‐CentralMinistry of Agriculture Changsha China
| | - Xiangfeng Kong
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic ProcessKey Laboratory of Agro‐ecological Processes in Subtropical RegionInstitute of Subtropical AgricultureChinese Academy of SciencesHunan Provincial Engineering Research Center for Healthy Livestock and Poultry ProductionScientific Observing and Experimental Station of Animal Nutrition and Feed Science in South‐CentralMinistry of Agriculture Changsha China
| |
Collapse
|
18
|
Szuster-Ciesielska A, Zwolak A, Daniluk J, Kandefer-Szerszeń M. Metformin inhibits both oleic acid-induced and CB1R receptor agonist-induced lipid accumulation in Hep3B cells: The preliminary report. Int J Immunopathol Pharmacol 2019; 33:2058738419832714. [PMID: 30880507 PMCID: PMC6423672 DOI: 10.1177/2058738419832714] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Fatty liver is characterized by excessive accumulation of triglycerides within
hepatocytes. Recent findings indicate that natural history of nonalcoholic fatty
liver is regulated, in part, by endogenous cannabinoids. Metformin is an oral
hypoglycemic medication which inhibits gluconeogenesis and glycogenolysis in
hepatocytes and limits lipid storage in the liver through the inhibition of free
fatty acid formation via induction of activated protein kinase activity (AMPK).
Both endocannabinoids and metformin may modulate hepatosteatosis; therefore, it
was interesting to examine whether metformin may affect lipid accumulation in
hepatocytes by acting on cannabinoid receptors, CB1 and CB2, in in vitro study.
Hep3B cells were incubated with or without metformin (Met), phosphatidylcholine
(PC), and oleic acid (OA). Cells without any of the examined substances served
as negative control. Cells treated only with OA served as positive control. The
quantity of intracellular lipids was assessed using Oil-Red-O staining.
Selective CB1R agonist, arachidonyl-2-chloromethylamide (ACEA), and CB2R
agonist, AM1241
(2-iodo-5-nitrophenyl)-[1-(methylpiperidin-2-ylmethyl)-1H-indol-3-yl]methanone,
were also used to treat Hep3B cells. In some experiments, antagonist for CB1R,
AM6545, or SR144528 as selective antagonist of CB2R were used. In the study, Met
decreased lipid accumulation in cells treated with OA and inhibited CB1R
agonist–induced lipid accumulation in hepatocytes. The CB2R agonist–induced
hepatic lipid accumulation was not inhibited by metformin. The results indicate
that metformin may interact with endocannabinoid system in the liver by
inhibiting CB1R agonist–stimulated fat accumulation in hepatocytes.
Collapse
Affiliation(s)
| | - Agnieszka Zwolak
- 2 Chair of Internal Medicine and Department of Internal Medicine in Nursing, Medical University of Lublin, Lublin, Poland.,3 Department and Clinic of Endocrinology, Medical University of Lublin, Lublin, Poland
| | - Jadwiga Daniluk
- 2 Chair of Internal Medicine and Department of Internal Medicine in Nursing, Medical University of Lublin, Lublin, Poland.,4 Department of Health, Pope John Paul II State School of Higher Education in Biała Podlaska, Biała Podlaska, Poland
| | | |
Collapse
|
19
|
Park JH, Ahn EK, Kim JK, Oh JS. Antihyperlipidemic Activity of Ligularia fischeri Extract in Mice Fed a High-Carbohydrate Diet. J Med Food 2019; 22:374-383. [PMID: 30801226 DOI: 10.1089/jmf.2018.4248] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Ligularia fischeri, indigenous to eastern Asia, has been used as a traditional herbal medicine. Ligularia fischeri reportedly possesses a number of biological activities such as antimutagenic, antioxidant, antigenotoxic, and anti-inflammation. This study demonstrated the effects of ethanol extracts of Ligularia fischeri (ELF) on a high-carbohydrate diet (HCD)-induced hyperlipidemia in C57BL/6 mice. The mice were divided into six groups (n = 7/group) as follows: normal diet, HCD, or HCD+ELF (100, 200, 400, and 800 mg/kg/day), which were orally administered daily for 12 weeks. Various lipid parameters and histological changes in liver and fat tissue were compared among the treatment and control groups. ELF remarkably reduced body weight gain and attenuated hyperlipidemia by improving the plasma levels of total cholesterol, triglycerides, high-density lipoprotein cholesterol, low-density lipoprotein cholesterol, atherogenic index, and cardiac risk factor. Moreover, ELF decreased the HCD-induced hepatic accumulation of lipid droplets and adipocyte hypertrophy. These regulatory effects of ELF appeared to be mediated through the phosphorylation of AMP-activated protein kinase, acetyl-CoA carboxylase, sterol regulatory element-binding protein-1c, and expression of fatty acid synthase. Taken together, these findings indicate a functional role for ELF in the regulation of HCD-induced obesity and hyperlipidemia.
Collapse
Affiliation(s)
- Ju-Hyoung Park
- 1 Department of Pharmacy, Dankook University, Cheonan, Chungnam, Korea
| | - Eun-Kyung Ahn
- 2 Department of Bio-Center, Gyeonggido Business and Science Accelerator, Suwon, Gyeonggi, Korea
| | - Jin-Kyu Kim
- 2 Department of Bio-Center, Gyeonggido Business and Science Accelerator, Suwon, Gyeonggi, Korea
| | - Joa Sub Oh
- 1 Department of Pharmacy, Dankook University, Cheonan, Chungnam, Korea
| |
Collapse
|
20
|
Zemel MB, Kolterman O, Rinella M, Vuppalanchi R, Flores O, Barritt AS, Siddiqui M, Chalasani N. Randomized Controlled Trial of a Leucine-Metformin-Sildenafil Combination (NS-0200) on Weight and Metabolic Parameters. Obesity (Silver Spring) 2019; 27:59-67. [PMID: 30569637 DOI: 10.1002/oby.22346] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 09/20/2018] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Leucine was previously demonstrated to allosterically activate mammalian sirtuin 1 and synergize with other sirtuin 1/AMP-activated protein kinase/nitric oxide pathway activators to modulate energy metabolism. The objective of this study was to evaluate the effects of a triple combination of leucine, metformin, and sildenafil (NS-0200) on body weight and obesity comorbidities in a phase 2 randomized trial. METHODS A total of 91 subjects with obesity were randomized to placebo, low dose (1.1 g leucine/0.5 g metformin/0.5 mg sildenafil), or high dose (1.1 g leucine/0.5 g metformin/1.0 mg sildenafil) twice daily for 16 weeks. Seventy subjects completed the trial and met all a priori compliance criteria. Hypertensive (n = 35) and hypertriglyceridemic (n = 22) subcohorts were also analyzed. RESULTS NS-0200 dose-responsively reduced weight; high dose reduced weight by 2.4 and 5.0 kg in the full and high-triglyceride cohorts, respectively (P < 0.0001). High-dose NS-0200 treatment also decreased blood pressure (-5.5 mm Hg diastolic pressure; P = 0.011), with greater effects among hypertensive subjects. NS-0200 also significantly reduced triglycerides and hemoglobin A1c. Significant improvement in ≥ 2 comorbidities was exhibited by 54% of subjects in the high-dose arm versus 5% of placebo subjects (P = 0.0009). Treatment-emergent adverse events did not significantly differ among groups. CONCLUSIONS These data support further study of NS-0200 as a therapy for obesity and associated comorbidities.
Collapse
Affiliation(s)
| | | | - Mary Rinella
- Division of Gastroenterology and Hepatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Raj Vuppalanchi
- Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, Illinois, USA
| | | | - A Sidney Barritt
- Division of Gastroenterology and Hepatology, UNC School of Medicine, Chapel Hill, North Carolina, USA
| | - Mohammad Siddiqui
- Department of Internal Medicine, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Naga Chalasani
- Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, Illinois, USA
| |
Collapse
|
21
|
Effects of Metformin Combined with Lactoferrin on Lipid Accumulation and Metabolism in Mice Fed with High-Fat Diet. Nutrients 2018; 10:nu10111628. [PMID: 30400147 PMCID: PMC6265902 DOI: 10.3390/nu10111628] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 10/15/2018] [Accepted: 10/22/2018] [Indexed: 12/17/2022] Open
Abstract
Metformin (Met) and lactoferrin (Lf) both exhibit beneficial effects on body weight management and lipid accumulation. However, the synergistical action of Met and Lf remains unclear. In this study, 64 mice were divided into five groups, namely, the control group, high-fat diet (HFD group), HFD with Met (Met group), Lf (Lf group), and a combination of Met and Lf (Met + Lf group). Met (200 mg/kg body weight) and Lf (2 g/100 mL) were administrated in drinking water. The experiment lasted for 12 weeks. Body weight, serum, and hepatic lipids were determined. Histology of the liver and perirenal fat was observed. Protein expression related to hepatic lipid metabolism was also measured. HFD significantly increased body weight, visceral fat weight, and lipid profiles, which lead to obesity and dyslipidemia in mice. Compared with the HFD group, the treatments significantly decreased body weight and Lee’s index (body mass index of mice) with the lowest values in the Met + Lf group. The treatments also decreased the weight of visceral fat, and improved circulating lipid profile and the ability for regulating glucose intake. The adipocyte size and serum TC level were significantly lower in the Met + Lf group as compared with those in the Met or Lf group. The treatments alleviated hepatic lipid accumulation, especially in the Met + Lf group. For protein expression, the p-AMPK/AMPK ratio, a key kinase-regulating cellular energy homeostasis, was significantly higher in the Met + Lf group than the ratio in the HFD group. Similarly, the treatments significantly downregulated the protein expression of lipogenic enzymes (FAS, ACC, and SREBP-1) and upregulated the protein expression of lipolytic enzyme (ATGL). The protein expression of HMGCoAR, which is an important rate limiting enzyme in cholesterol biosynthesis, was only significantly lower in the Met + Lf group than in the HFD group. In conclusion, Met and Lf, either alone or in combination, prevented HFD-induced obesity and improved lipid metabolism.
Collapse
|
22
|
Noureddin M, Loomba R. Editorial: role of leucine-metformin-sildenafil combination in the treatment of nonalcoholic fatty liver disease (NAFLD). Aliment Pharmacol Ther 2018; 48:378-379. [PMID: 29998503 DOI: 10.1111/apt.14819] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- M Noureddin
- Division of Digestive and Liver Diseases, Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - R Loomba
- Division of Gastroenterology, NAFLD Research Center, University of California at San Diego, San Diego, CA, USA
| |
Collapse
|
23
|
Dietary Leucine Supplement Ameliorates Hepatic Steatosis and Diabetic Nephropathy in db/db Mice. Int J Mol Sci 2018; 19:ijms19071921. [PMID: 29966331 PMCID: PMC6073714 DOI: 10.3390/ijms19071921] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 06/25/2018] [Accepted: 06/29/2018] [Indexed: 12/14/2022] Open
Abstract
Dietary leucine supplementation has been explored for the therapeutic intervention of obesity and obesity-induced metabolic dysfunctions. In this study, we aim to examine the effects of dietary leucine supplementation in db/db mice. Mice were treated with or without leucine (1.5% w/v) in drinking water for 12 weeks. The leucine supplement was found to reduce insulin resistance and hepatic steatosis in db/db mice. Using Nuclear Magnetic Resonance (NMR)-based lipidomics, we found that the reduction of hepatic triglyceride synthesis was correlated with attenuated development of fatty liver. In addition, diabetic nephropathy (DN) was also ameliorated by leucine. Using liquid chromatography–time-of-flight mass spectrometry (LC-TOF MS)-based urine metabolomics analysis, we found that the disturbance of the tricarboxylic acid (TCA) cycle was reversed by leucine. The beneficial effects of leucine were probably due to AMP-activated protein kinase (AMPK) activation in the liver and kidneys of db/db mice. Thus, dietary leucine supplementation may potentially be a nutritional intervention to attenuate hepatic steatosis and early DN in type II diabetes.
Collapse
|
24
|
Chalasani N, Vuppalanchi R, Rinella M, Middleton MS, Siddiqui MS, Barritt AS, Kolterman O, Flores O, Alonso C, Iruarrizaga‐Lejarreta M, Gil‐Redondo R, Sirlin CB, Zemel MB. Randomised clinical trial: a leucine-metformin-sildenafil combination (NS-0200) vs placebo in patients with non-alcoholic fatty liver disease. Aliment Pharmacol Ther 2018; 47:1639-1651. [PMID: 29696666 PMCID: PMC6001629 DOI: 10.1111/apt.14674] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 02/24/2018] [Accepted: 03/27/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Sirtuin 1 (Sirt1) is suppressed in non-alcoholic fatty liver disease (NAFLD), while its' stimulation or overexpression results in reduced disease severity in pre-clinical NAFLD models. Leucine allosterically activates Sirt1 and synergise with other Sirt/AMPK/NO pathway activators. We developed a triple combination of leucine, metformin and sildenafil (NS-0200), which was effective in a mouse model of non-alcoholic steatohepatitis (NASH). AIM To report the results from a Phase 2, randomised clinical trial of of NS-0200 in 91 subjects with NAFLD (liver fat ≥15% by magnetic resonance imaging-proton-density fat fraction (MRI-PDFF)). METHODS Subjects were randomised to placebo, low-dose (1.1 g leucine/0.5 g metformin/0.5 mg sildenafil) or high-dose NS-0200 (1.1 g leucine/0.5 g metformin/1.0 mg sildenafil) b.d. for 16 weeks; change in hepatic fat was assessed via MRI-PDFF, and lipid metabolism was assessed via changes in the lipidomic signature. Seventy subjects completed the trial and met a priori compliance criteria. Analyses were conducted on the full cohort and on those with alanine aminotransferase (ALT) values above median (50 U/L; n = 35). RESULTS In the full cohort, active treatments did not separate from placebo. High dose NS-0200 reduced hepatic fat by 15.7% (relative change from baseline) in the high ALT group (P < 0.005) while low dose NS-0200 and placebo did not significantly change hepatic fat. Lipidomic analysis showed dose-responsive treatment effects in both overall and high ALT cohorts, with significant decreases in metabolically active lipids and up-regulation of fatty acid oxidation. CONCLUSION These data support further evaluation of high-dose NS-0200 for treating NASH, especially in those with elevated ALT (NCT 02546609).
Collapse
Affiliation(s)
- N. Chalasani
- Indiana University School of MedicineIndianapolisINUSA
| | | | | | | | | | | | | | | | | | | | | | - C. B. Sirlin
- University of California at San DiegoSan DiegoCAUSA
| | | |
Collapse
|
25
|
Branched-chain amino acid ratios modulate lipid metabolism in adipose tissues of growing pigs. J Funct Foods 2018. [DOI: 10.1016/j.jff.2017.12.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
26
|
Liu Y, Jiang X, Chen X. Liraglutide and Metformin alone or combined therapy for type 2 diabetes patients complicated with coronary artery disease. Lipids Health Dis 2017; 16:227. [PMID: 29197387 PMCID: PMC5712174 DOI: 10.1186/s12944-017-0609-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 11/05/2017] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND This study is to compare the effects of Liraglutide and Metformin alone or combined treatment on the cardiac function in T2DM patients complicated with CAD. METHODS 120 T2DM patients were included at Endocrinology Department of Tianjin First Center Hospital (Tianjin, China) from April 2012 to September 2013. The study contained two sections. Section 1: 30 patients in group 1 was treated with Liraglutide (Novo Nordisk) (1.2 mg/d), and 30 patients in group 2 with Metformin (Shiguibao) (1500 mg/d) for 24 weeks. Section 2: 30 patients in group1 was treated with Liraglutide (1.8 mg/d) and 30 in group 2 with Liraglutide (1.2 mg/d) plus Metformin (1500 mg/d) for 24 weeks. Fasting blood glucose (FBG), postprandial glucose (PPG), glycated hemoglobin (HbA1c), body mass index (BMI), blood pressure (BP), triglyceride (TG), total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), C reactive protein (CRP), left ventricular end-diastolic diameter (LVEDD), ejection fraction (EF) and the ratio of early (E) to late (A) ventricular filling velocities (E/A ratio) were measured before and after the 24-week treatment. RESULTS After 24-week treatment, when blood glucose level was controlled in 4 groups, Liraglutide alone treatment showed better improvements than on all measuring except TG in Section 1, however, combined treatment of Liraglutide and Metformin showed better improvements on all measuring except BMI, TG and BP in Section 2. CONCLUSIONS With similar glycemic control, the Liraglutide (1.2 mg/d) monotherapy showed the better effects than either Metformin alone, or combination of Liraglutide and Metformin on lipid metabolism and cardiovascular function. TRIAL REGISTRATION This trial was registered at Chinese Clinical Trial Registry ( chictr.org.cn ) # ChiCTR-IPR-16008578 .
Collapse
Affiliation(s)
- Ying Liu
- Department of Endocrinology, Tianjin First Center Hospital, 24 Fukang Road, Nankai District, Tianjin, 300192, China.
| | - Xia Jiang
- Department of Endocrinology, Tianjin First Center Hospital, 24 Fukang Road, Nankai District, Tianjin, 300192, China
| | - Xin Chen
- Department of Cardiovascular Medicine, Tianjin First Center Hospital, Tianjin, 300192, China
| |
Collapse
|
27
|
Sadri H, Larki NN, Kolahian S. Hypoglycemic and Hypolipidemic Effects of Leucine, Zinc, and Chromium, Alone and in Combination, in Rats with Type 2 Diabetes. Biol Trace Elem Res 2017; 180:246-254. [PMID: 28409409 DOI: 10.1007/s12011-017-1014-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 04/04/2017] [Indexed: 01/08/2023]
Abstract
For the increasing development of diabetes, dietary habits and using appropriate supplements can play important roles in the treatment or reduction of risk for this disease. The objective of this study was to investigate the effects of leucine (Leu), zinc (Zn), and chromium (Cr) supplementation, alone or in combination, in rats with type 2 diabetes (T2D). Seventy-seven adult male Wistar rats were randomly assigned in 11 groups, using nutritional supplements and insulin (INS) or glibenclamide (GLC). Supplementing Leu significantly reduced blood glucose, triglycerides (TG), nonesterified fatty acids (NEFA), low-density lipoprotein (LDL), and increased high-density lipoprotein (HDL) concentrations compared to vehicle-treated T2D animals, and those improvements were associated with reduced area under the 2-h blood glucose response curve (AUC). Supplementation of T2D animals with Zn improved serum lipid profile as well as blood glucose concentrations but was not comparable with the INS, GLC, and Leu groups. Supplementary Cr did not improve blood glucose and AUC in T2D rats, whereas it reduced serum TG and LDL and increased HDL concentrations. In conclusion, supplementation of diabetic rats with Leu was more effective in improving blood glucose and consequently decreasing glucose AUC than other nutritional supplements. Supplementary Zn and Cr only improved serum lipid profile. The combination of the nutritional supplements did not improve blood glucose level. Nevertheless, supplementation with Leu-Zn, Leu-Cr, Zn-Cr, and Leu-Zn-Cr led to an improved response in serum lipid profile over each supplement given alone.
Collapse
Affiliation(s)
- Hassan Sadri
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, 516616471, Iran.
| | - Negar Nowroozi Larki
- Department of Pharmacology and Experimental Therapy, Institute of Experimental and Clinical Pharmacology and Toxicology and ICePhA, University of Tuebingen, 72074, Tuebingen, Germany
- Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, 516616471, Iran
| | - Saeed Kolahian
- Department of Pharmacology and Experimental Therapy, Institute of Experimental and Clinical Pharmacology and Toxicology and ICePhA, University of Tuebingen, 72074, Tuebingen, Germany
- Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, 516616471, Iran
| |
Collapse
|
28
|
Luan Y, Zhang F, Cheng Y, Liu J, Huang R, Yan M, Wang Y, He Z, Lai H, Wang H, Ying H, Guo F, Zhai Q. Hemin Improves Insulin Sensitivity and Lipid Metabolism in Cultured Hepatocytes and Mice Fed a High-Fat Diet. Nutrients 2017; 9:nu9080805. [PMID: 28933767 PMCID: PMC5579599 DOI: 10.3390/nu9080805] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 07/05/2017] [Accepted: 07/17/2017] [Indexed: 12/18/2022] Open
Abstract
Hemin is a breakdown product of hemoglobin. It has been reported that the injection of hemin improves lipid metabolism and insulin sensitivity in various genetic models. However, the effect of hemin supplementation in food on lipid metabolism and insulin sensitivity is still unclear, and whether hemin directly affects cellular insulin sensitivity is yet to be elucidated. Here we show that hemin enhances insulin-induced phosphorylation of insulin receptors, Akt, Gsk3β, FoxO1 and cytoplasmic translocation of FoxO1 in cultured primary hepatocytes under insulin-resistant conditions. Furthermore, hemin diminishes the accumulation of triglyceride and increases in free fatty acid content in primary hepatocytes induced by palmitate. Oral administration of hemin decreases body weight, energy intake, blood glucose and triglyceride levels, and improves insulin and glucose tolerance as well as hepatic insulin signaling and hepatic steatosis in male mice fed a high-fat diet. In addition, hemin treatment decreases the mRNA and protein levels of some hepatic genes involved in lipogenic regulation, fatty acid synthesis and storage, and increases the mRNA level and enzyme activity of CPT1 involved in fatty acid oxidation. These data demonstrate that hemin can improve lipid metabolism and insulin sensitivity in both cultured hepatocytes and mice fed a high-fat diet, and show the potential beneficial effects of hemin from food on lipid and glucose metabolism.
Collapse
Affiliation(s)
- Yi Luan
- Key Laboratory of Nutrition and Metabolism, CAS Center for Excellence in Molecular Cell Sciences, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China.
| | - Fang Zhang
- Key Laboratory of Nutrition and Metabolism, CAS Center for Excellence in Molecular Cell Sciences, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China.
| | - Yalan Cheng
- Key Laboratory of Nutrition and Metabolism, CAS Center for Excellence in Molecular Cell Sciences, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China.
| | - Jun Liu
- Key Laboratory of Nutrition and Metabolism, CAS Center for Excellence in Molecular Cell Sciences, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China.
| | - Rui Huang
- Key Laboratory of Nutrition and Metabolism, CAS Center for Excellence in Molecular Cell Sciences, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China.
| | - Menghong Yan
- Key Laboratory of Nutrition and Metabolism, CAS Center for Excellence in Molecular Cell Sciences, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China.
| | - Yuangao Wang
- Key Laboratory of Nutrition and Metabolism, CAS Center for Excellence in Molecular Cell Sciences, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China.
| | - Zhishui He
- Key Laboratory of Nutrition and Metabolism, CAS Center for Excellence in Molecular Cell Sciences, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China.
| | - Hejin Lai
- Key Laboratory of Nutrition and Metabolism, CAS Center for Excellence in Molecular Cell Sciences, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China.
| | - Hui Wang
- Key Laboratory of Nutrition and Metabolism, CAS Center for Excellence in Molecular Cell Sciences, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China.
| | - Hao Ying
- Key Laboratory of Nutrition and Metabolism, CAS Center for Excellence in Molecular Cell Sciences, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China.
| | - Feifan Guo
- Key Laboratory of Nutrition and Metabolism, CAS Center for Excellence in Molecular Cell Sciences, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China.
| | - Qiwei Zhai
- Key Laboratory of Nutrition and Metabolism, CAS Center for Excellence in Molecular Cell Sciences, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China.
- School of Life Science and Technology, Shanghai Tech University, Shanghai 200093, China.
| |
Collapse
|
29
|
Farr OM, Mantzoros CS. Treatment options to prevent diabetes in subjects with prediabetes: Efficacy, cost effectiveness and future outlook. Metabolism 2017; 70:192-195. [PMID: 28095990 PMCID: PMC5871912 DOI: 10.1016/j.metabol.2016.12.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 12/31/2016] [Indexed: 01/18/2023]
Affiliation(s)
- Olivia M Farr
- Division of Endocrinology, Beth Israel Deaconess Medical Center, Boston, MA 02215.
| | - Christos S Mantzoros
- Division of Endocrinology, Beth Israel Deaconess Medical Center, Boston, MA 02215
| |
Collapse
|
30
|
Duan YH, Li FN, Wen CY, Wang WL, Guo QP, Li YH, Yin YL. Branched-chain amino acid ratios in low-protein diets regulate the free amino acid profile and the expression of hepatic fatty acid metabolism-related genes in growing pigs. J Anim Physiol Anim Nutr (Berl) 2017; 102:e43-e51. [DOI: 10.1111/jpn.12698] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 01/20/2017] [Indexed: 02/06/2023]
Affiliation(s)
- Y. H. Duan
- Key Laboratory of Agro-ecological Processes in Subtropical Region; Institute of Subtropical Agriculture; Chinese Academy of Sciences; Changsha Hunan China
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production; Changsha Hunan China
- Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production; Changsha Hunan China
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central; Ministry of Agriculture; Changsha Hunan China
- University of Chinese Academy of Sciences; Beijing China
| | - F. N. Li
- Key Laboratory of Agro-ecological Processes in Subtropical Region; Institute of Subtropical Agriculture; Chinese Academy of Sciences; Changsha Hunan China
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production; Changsha Hunan China
- Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production; Changsha Hunan China
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central; Ministry of Agriculture; Changsha Hunan China
- Hunan Co-Innovation Center of Animal Production Safety; CICAPS; Changsha Hunan China. Hunan Collaborative Innovation Center for Utilization of Botanical Functional Ingredients; Changsha Hunan China
| | - C. Y. Wen
- Laboratory of Animal Nutrition and Human Health; School of Biology; Hunan Normal University; Changsha Hunan China
| | - W. L. Wang
- Laboratory of Animal Nutrition and Human Health; School of Biology; Hunan Normal University; Changsha Hunan China
| | - Q. P. Guo
- Key Laboratory of Agro-ecological Processes in Subtropical Region; Institute of Subtropical Agriculture; Chinese Academy of Sciences; Changsha Hunan China
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production; Changsha Hunan China
- Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production; Changsha Hunan China
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central; Ministry of Agriculture; Changsha Hunan China
- University of Chinese Academy of Sciences; Beijing China
| | - Y. H. Li
- Key Laboratory of Agro-ecological Processes in Subtropical Region; Institute of Subtropical Agriculture; Chinese Academy of Sciences; Changsha Hunan China
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production; Changsha Hunan China
- Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production; Changsha Hunan China
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central; Ministry of Agriculture; Changsha Hunan China
- University of Chinese Academy of Sciences; Beijing China
| | - Y. L. Yin
- Key Laboratory of Agro-ecological Processes in Subtropical Region; Institute of Subtropical Agriculture; Chinese Academy of Sciences; Changsha Hunan China
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production; Changsha Hunan China
- Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production; Changsha Hunan China
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central; Ministry of Agriculture; Changsha Hunan China
- Laboratory of Animal Nutrition and Human Health; School of Biology; Hunan Normal University; Changsha Hunan China. College of Animal Science; South China Agricultural University; Guangzhou China
| |
Collapse
|
31
|
Kawaguchi T, Ueno T, Nogata Y, Hayakawa M, Koga H, Torimura T. Wheat-bran autolytic peptides containing a branched-chain amino acid attenuate non-alcoholic steatohepatitis via the suppression of oxidative stress and the upregulation of AMPK/ACC in high-fat diet-fed mice. Int J Mol Med 2016; 39:407-414. [DOI: 10.3892/ijmm.2016.2831] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 12/12/2016] [Indexed: 11/06/2022] Open
|
32
|
Kong Q, Zhang H, Zhao T, Zhang W, Yan M, Dong X, Li P. Tangshen formula attenuates hepatic steatosis by inhibiting hepatic lipogenesis and augmenting fatty acid oxidation in db/db mice. Int J Mol Med 2016; 38:1715-1726. [PMID: 27840945 PMCID: PMC5117754 DOI: 10.3892/ijmm.2016.2799] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 11/03/2016] [Indexed: 12/24/2022] Open
Abstract
Tangshen formula (TSF), a well-prescribed traditional Chinese formula, has been used in the treatment of diabetic nephropathy. However, whether TSF ameliorates dyslipidemia and liver injury associated with diabetes remains unclear. In this study, we examined the effects of TSF on lipid profiles and hepatic steatosis in db/db mice. For this purpose, 8‑week-old db/db mice were treated with TSF or saline for 12 weeks via gavage and db/m mice were used as controls. Body weight and blood glucose levels were monitored weekly and bi-weekly, respectively. Blood samples were obtained for the analysis of lipids and enzymes related to hepatic function, and liver tissues were analyzed by histology, immunohistochemistry and molecular examination. The results revealed that TSF markedly reduced body weight, liver index [liver/body weight (LW/BW)] and improved lipid profiles, hepatic function and steatosis in db/db mice. TSF induced the phosphoralation of AMP-activated protein kinase and inhibited the activity of sterol regulatory element-binding protein 1 together with the inhibition of the expression of genes involved in de novo lipogenesis (DNL) and gluconeogenesis, such as fatty acid synthase (FAS), acetyl-CoA carboxylase (ACC), stearoyl CoA desaturase 1 (SCD1), glucose-6-phosphatase (G6pc) and phosphoenolpyruvate carboxykinase 1 (Pck1). Additionally, the silent mating type information regulation 2 homolog 1 (Sirt1)/peroxisome proliferator-activated receptor α (PPARα)/malonyl-CoA decarboxylase (MLYCD) cascade was potently activated by TSF in the liver and skeletal muscle of db/db mice, which led to enhanced fatty acid oxidation. These findings demonstrated that TSF attenuated hepatic fat accumulation and steatosis in db/db mice by inhibiting lipogenesis and augmenting fatty acid oxidation.
Collapse
Affiliation(s)
- Qin Kong
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, P.R. China
| | - Haojun Zhang
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, P.R. China
| | - Tingting Zhao
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, P.R. China
| | - Weiku Zhang
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, P.R. China
| | - Meihua Yan
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, P.R. China
| | - Xi Dong
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, P.R. China
| | - Ping Li
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, P.R. China
| |
Collapse
|
33
|
Banerjee J, Bruckbauer A, Zemel MB. Activation of the AMPK/Sirt1 pathway by a leucine-metformin combination increases insulin sensitivity in skeletal muscle, and stimulates glucose and lipid metabolism and increases life span in Caenorhabditis elegans. Metabolism 2016; 65:1679-1691. [PMID: 27456392 DOI: 10.1016/j.metabol.2016.06.011] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 06/28/2016] [Accepted: 06/29/2016] [Indexed: 12/15/2022]
Abstract
BACKGROUND We have previously shown leucine (Leu) to activate Sirt1 by lowering its KM for NAD+, thereby amplifying the effects of other sirtuin activators and improving insulin sensitivity. Metformin (Met) converges on this pathway both indirectly (via AMPK) and by direct activation of Sirt1, and we recently found Leu to synergize with Met to improve insulin sensitivity and glycemic control while achieving ~80% dose-reduction in diet-induced obese mice. Accordingly, we sought here to define the mechanism of this interaction. METHODS Muscle cells C2C12 and liver cells HepG2 were used to test the effect of Met-Leu on Sirt1 activation. Caenorhabditis elegans was used for glucose utilization and life span studies. RESULTS Leu (0.5mmol/L)+Met (50-100μmol/L) synergistically activated Sirt1 (p<0.001) at low (≤100μmol/L) NAD+ levels while Met exerted no independent effect. This was associated with an increase in AMPK and ACC, phosphorylation, and increased fatty acid oxidation, which was prevented by AMPK or Sirt inhibition or silencing. Met-Leu also increased P-IRS1/IRS1 and P-AKT/AKT and in insulin-independent glucose disposal in myotubes (~50%, p<0.002) evident within 30 min as well as a 60% reduction in insulin EC50. In addition, in HepG2 liver cells nuclear CREB regulated transcription coactivator 2 (CRTC2) protein expression and phosphorylation of glycogen synthase was decreased, while glycogen synthase kinase phosphorylation was increased indicating decreased gluconeogenesis and glycogen synthesis. We utilized C. elegans to assess the metabolic consequences of this interaction. Exposure to high glucose impaired glucose utilization and shortened life span by ~25%, while addition of Leu+Met to high glucose worms increased median and maximal life span by 29 and 15%, respectively (p=0.023), restored normal glucose utilization and increased fat oxidation ~two-fold (p<0.005), while metformin exerted no independent effect at any concentration (0.1-0.5mmol/L). CONCLUSION Thus, Leu and Met synergize to enable Sirt1 activation at low NAD+ concentrations (typical of energy replete states). Sirt1 and AMPK activations are required for Met-Leu's full action, which result in improvements in energy metabolism and insulin sensitivity.
Collapse
Affiliation(s)
- Jheelam Banerjee
- NuSirt BioPharma Inc., 11020 Solway School Road, Knoxville, TN 37931, USA.
| | - Antje Bruckbauer
- NuSirt BioPharma Inc., 11020 Solway School Road, Knoxville, TN 37931, USA
| | - Michael B Zemel
- NuSirt BioPharma Inc., 11020 Solway School Road, Knoxville, TN 37931, USA
| |
Collapse
|
34
|
Yao K, Duan Y, Li F, Tan B, Hou Y, Wu G, Yin Y. Leucine in Obesity: Therapeutic Prospects. Trends Pharmacol Sci 2016; 37:714-727. [DOI: 10.1016/j.tips.2016.05.004] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 05/08/2016] [Accepted: 05/10/2016] [Indexed: 02/07/2023]
|
35
|
Polyzos SA, Mantzoros CS. Nonalcoholic fatty future disease. Metabolism 2016; 65:1007-16. [PMID: 26805015 DOI: 10.1016/j.metabol.2015.12.009] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 12/15/2015] [Accepted: 12/16/2015] [Indexed: 01/12/2023]
Affiliation(s)
- Stergios A Polyzos
- Second Medical Clinic, Aristotle University of Thessaloniki, Ippokration Hospital, Thessaloniki, Greece
| | - Christos S Mantzoros
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; Boston VA Healthcare system and Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
36
|
Jiang Tang Xiao Ke Granule, a Classic Chinese Herbal Formula, Improves the Effect of Metformin on Lipid and Glucose Metabolism in Diabetic Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 2016:1592731. [PMID: 27418937 PMCID: PMC4932160 DOI: 10.1155/2016/1592731] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 04/30/2016] [Accepted: 05/24/2016] [Indexed: 12/19/2022]
Abstract
In the present study, the hypoglycemic, hypolipidemic, and antioxidative effects of metformin (MET) combined with Jiang Tang Xiao Ke (JTXK) granule derived from the “Di Huang Tang” were evaluated in mice with type 2 diabetes mellitus (DM) induced by high-fat diet/streptozotocin. DM mice were orally treated with MET (0.19 g/kg) either alone or combined with different doses (1.75, 3.5, or 7 g/kg) of JTXK for 4 weeks. Results showed that the serum and hepatic glucose, lipids, and oxidative stress levels were elevated in DM mice, when compared with the normal mice. MET treatment decreased FBG and serum glucagon levels of DM mice. Combination treatment with MET and JTXK 3.5 g/kg increased the hypoglycemia and insulin sensitivity at 4 weeks when compared with the DM mice treated with MET alone. However, neither MET nor MET/JTXK treatment could completely reverse the hyperglycemia in DM mice. JTXK enhanced the serum triglyceride (TG) and hepatic lipid-lowering effect of MET in a dose-dependent manner in DM mice. JTXK 1.75 and 3.5 g/kg improved the hepatoprotective effect of MET in DM mice. Synergistic effect of combination treatment with MET and JTXK on antioxidant stress was also found in DM mice compared with MET alone.
Collapse
|
37
|
García-Heredia A, Riera-Borrull M, Fort-Gallifa I, Luciano-Mateo F, Cabré N, Hernández-Aguilera A, Joven J, Camps J. Metformin administration induces hepatotoxic effects in paraoxonase-1-deficient mice. Chem Biol Interact 2016; 249:56-63. [PMID: 26945512 DOI: 10.1016/j.cbi.2016.03.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 02/23/2016] [Accepted: 03/01/2016] [Indexed: 12/11/2022]
Abstract
Metformin is the first-line pharmacological treatment of diabetes. In these patients, metformin reduces body weight and decreases the risk of diabetes-related complications such as cardiovascular disease. However, whether metformin elicits beneficial effects on liver histology is a controversial issue and, as yet, there is no consensus. Paraoxonase-1 (PON1), an enzyme synthesized mainly by the liver, degrades lipid peroxides and reduces oxidative stress. PON1 activities are decreased in chronic liver diseases. We evaluated the effects of metformin in the liver of PON1-deficient mice which, untreated, present a mild degree of liver steatosis. Metformin administration aggravated inflammation in animals given a standard mouse chow and in those fed a high-fat diet. Also, it was associated with a higher degree of steatosis in animals fed a standard chow diet. This report is a cautionary note regarding the prescription of metformin for the treatment of diabetes in patients with concomitant liver impairment.
Collapse
Affiliation(s)
- Anabel García-Heredia
- Unitat de Recerca Biomèdica (CRB-URB), Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili, Reus, Spain
| | - Marta Riera-Borrull
- Unitat de Recerca Biomèdica (CRB-URB), Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili, Reus, Spain
| | - Isabel Fort-Gallifa
- Unitat de Recerca Biomèdica (CRB-URB), Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili, Reus, Spain; Laboratori de Referència Sud, Hospital Universitari de Sant Joan, Reus, Spain
| | - Fedra Luciano-Mateo
- Unitat de Recerca Biomèdica (CRB-URB), Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili, Reus, Spain
| | - Noemí Cabré
- Unitat de Recerca Biomèdica (CRB-URB), Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili, Reus, Spain
| | - Anna Hernández-Aguilera
- Unitat de Recerca Biomèdica (CRB-URB), Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili, Reus, Spain
| | - Jorge Joven
- Unitat de Recerca Biomèdica (CRB-URB), Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili, Reus, Spain
| | - Jordi Camps
- Unitat de Recerca Biomèdica (CRB-URB), Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili, Reus, Spain.
| |
Collapse
|
38
|
Paschou SA, Spyrou N, Mantzoros CS. Research advances in metabolism 2015. Metabolism 2016; 65:27-37. [PMID: 26892513 DOI: 10.1016/j.metabol.2015.10.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 10/15/2015] [Indexed: 11/16/2022]
Affiliation(s)
- Stavroula A Paschou
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| | - Nikolaos Spyrou
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Christos S Mantzoros
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
39
|
A Combination of Leucine, Metformin, and Sildenafil Treats Nonalcoholic Fatty Liver Disease and Steatohepatitis in Mice. Int J Hepatol 2016; 2016:9185987. [PMID: 28042486 PMCID: PMC5155097 DOI: 10.1155/2016/9185987] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 10/07/2016] [Accepted: 10/19/2016] [Indexed: 12/14/2022] Open
Abstract
Sirt1, AMPK, and eNOS modulate hepatic energy metabolism and inflammation and are key players in the development of NASH. L-leucine, an allosteric Sirt1 activator, synergizes with low doses of metformin or sildenafil on the AMPK-eNOS-Sirt1 pathway to reverse mild NAFLD in preclinical mouse models. Here we tested a possible multicomponent synergy to yield greater therapeutic efficacy in NAFLD/NASH. Liver cells and macrophages or an atherogenic diet induced NASH mouse model was treated with two-way and three-way combinations. The three-way combination Sild-Met-Leu increased hepatic fatty acid oxidation and reduced lipogenic gene expression and inflammatory marker in vitro. In mice, Sild-Met-Leu reduced the diet induced increases of ALT, TGFβ, PAI-1, IL1β, and TNFα, hepatic collagen expression, and nearly completely reversed hepatocyte ballooning and triglyceride accumulation, while all two-way combinations had only modest effects. Therefore, these data provide preclinical evidence for therapeutic efficacy of Sild-Met-Leu in the treatment of NAFLD and NASH.
Collapse
|
40
|
Liao PH, Kuo WW, Kuo CH, Yeh YL, Shen CY, Chen YH, Chen RJ, Padma VV, Chen YH, Huang CY. Lactobacillus reuteri GMNL-263 reduces hyperlipidaemia and the heart failure process in high-calorie diet-fed induced heart dysfunction in rats. J Funct Foods 2016. [DOI: 10.1016/j.jff.2015.11.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
41
|
Mathew H, Farr OM, Mantzoros CS. Metabolic health and weight: Understanding metabolically unhealthy normal weight or metabolically healthy obese patients. Metabolism 2016; 65:73-80. [PMID: 26683798 PMCID: PMC4750380 DOI: 10.1016/j.metabol.2015.10.019] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 10/01/2015] [Indexed: 02/07/2023]
Affiliation(s)
- Hannah Mathew
- Department of Internal Medicine, Boston Medical Center, 72 East Concord Street, Evans 124 Boston, MA 02118, USA; Section of Endocrinology, Beth-Israel Deaconess Medical Center/Harvard Medical School, Boston, MA, USA
| | - Olivia M Farr
- Section of Endocrinology, Beth-Israel Deaconess Medical Center/Harvard Medical School, Boston, MA, USA; Division of Endocrinology, Boston VA Healthcare System/Harvard Medical School, Boston, MA, USA.
| | - Christos S Mantzoros
- Section of Endocrinology, Beth-Israel Deaconess Medical Center/Harvard Medical School, Boston, MA, USA; Division of Endocrinology, Boston VA Healthcare System/Harvard Medical School, Boston, MA, USA
| |
Collapse
|