1
|
Xu M, Lv D, Wei H, Li Z, Jin S, Liu Q, Zhang Y, Liu Y. Effects of antidiabetic agents on lipid metabolism of skeletal muscle: A narrative review. Diabetes Obes Metab 2025; 27:1693-1707. [PMID: 39807619 DOI: 10.1111/dom.16189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 12/17/2024] [Accepted: 12/26/2024] [Indexed: 01/16/2025]
Abstract
Metabolic syndrome-related diseases frequently involve disturbances in skeletal muscle lipid metabolism. The accumulation of lipid metabolites, lipid-induced mitochondrial stress in skeletal muscle cells, as well as the inflammation of adjacent adipose tissue, are associated with the development of insulin resistance and metabolic dysfunction. Consequently, when antidiabetic medications are used to treat various chronic conditions related to hyperglycaemia, the impact on skeletal muscle lipid metabolism should not be overlooked. However, current research has predominantly focused on muscle mass rather than skeletal muscle lipid metabolism and its interplay with glucose metabolism. In this review, we summarised the latest research on the effects of antidiabetic drugs and certain natural compounds with antidiabetic activity on skeletal muscle lipid metabolism, focusing on data from preclinical to clinical studies. Given the widespread use of antidiabetic drugs, a better understanding of their effects on skeletal muscle lipid metabolism merits further attention in future research.
Collapse
Affiliation(s)
- Ming Xu
- Department of Endocrinology, First Hospital of Shanxi Medical University, Taiyuan, China
- The First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Dongqing Lv
- Department of Endocrinology, First Hospital of Shanxi Medical University, Taiyuan, China
- The First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Hongxia Wei
- Department of Endocrinology, First Hospital of Shanxi Medical University, Taiyuan, China
- The First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Zhe Li
- Department of Endocrinology, First Hospital of Shanxi Medical University, Taiyuan, China
- The First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Shuqing Jin
- Department of Endocrinology, First Hospital of Shanxi Medical University, Taiyuan, China
- The First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Qinhao Liu
- Department of Endocrinology, First Hospital of Shanxi Medical University, Taiyuan, China
- The First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Yi Zhang
- Department of Pharmacology, Shanxi Medical University, Taiyuan, China
- Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan, China
| | - Yunfeng Liu
- Department of Endocrinology, First Hospital of Shanxi Medical University, Taiyuan, China
- The First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
2
|
Neto AC, Botelho M, Rodrigues AR, Lamas S, Araújo B, Guimarães JT, Gouveia AM, Almeida H, Neves D. Metformin reverses infertility in a mouse model of endometriosis: unveiling disease pathways and implications for future clinical approaches. Reprod Biomed Online 2025; 50:104474. [PMID: 39847839 DOI: 10.1016/j.rbmo.2024.104474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 09/20/2024] [Accepted: 09/25/2024] [Indexed: 01/25/2025]
Abstract
RESEARCH QUESTION Does metformin reverse endometriosis-associated infertility? DESIGN Endometriosis was induced by transplanting uterus fragments from B6CBAF1 mice into recipients of the same strain. The mice were divided into groups: endometriosis (End, n = 24), sham-operated (Sham, n = 12), endometriosis with metformin (0.5mg/ml) orally administered for 3 months (EndMet, n = 21) and sham-operated metformin-treated (ShamMet, n = 16). Implant growth was monitored using ultrasonography. Fibrosis was computer-assisted quantified in Masson's trichrome-stained sections of eutopic (EuEnd) and ectopic (EcEnd) endometrium. PCNA, CYP17a1, F4/80 and galectin-3 were analysed by immunofluorescence and western blotting, and NFkB, GPX-1 and HO-1 only by western blotting. Statistical significance was set at P <0.05. RESULTS The endometriosis model was successfully established. The End groups showed lower fertility rates than sham-operated mice (P = 0.0034), whereas metformin treatment increased the number of fetuses per pregnant mouse (P = 0.0295), restoring fertility to control levels; it also slowed implant growth and vascularization. Metformin also restored PCNA expression and fibrosis levels to those of non-treated EuSham mice. PCNA expression decreased in pregnant mice (P <0.0178). Metformin diminished CYP17a1 expression in EcEnd versus EuEnd non-treated tissues and conversely up-regulated F4/80 in EuEnd tissue (P <0.0170), and galectin-3, NFkB and the antioxidant enzymes HO-1 and GPX-1 in EcEnd tissue (P <0.0293), in non-mated mice. CONCLUSIONS These results indicate that application of metformin can alleviate oxidative stress and mitigate fibrosis in endometriosis lesions in a murine model of endometriosis, which highlights metformin's potential as a pharmacological intervention for improving infertility in endometriosis.
Collapse
Affiliation(s)
- A Catarina Neto
- Department of Biomedicine Experimental Biology Unit, Faculty of Medicine of the University of Porto, Porto, Portugal.; Instituto de Investigação e Inovação em Saúde (i3S), Porto, Portugal
| | - Maria Botelho
- Department of Biomedicine Experimental Biology Unit, Faculty of Medicine of the University of Porto, Porto, Portugal.; Instituto de Investigação e Inovação em Saúde (i3S), Porto, Portugal
| | - Adriana R Rodrigues
- Department of Biomedicine Experimental Biology Unit, Faculty of Medicine of the University of Porto, Porto, Portugal.; Instituto de Investigação e Inovação em Saúde (i3S), Porto, Portugal.; Faculty of Nutrition and Food Sciences, University of Porto, Porto, Portugal
| | - Sofia Lamas
- Instituto de Investigação e Inovação em Saúde (i3S), Porto, Portugal
| | - Beatriz Araújo
- Serviço de Patologia Clínica da ULS de Braga, Braga, Portugal
| | - J Tiago Guimarães
- Department of Biomedicine Biochemistry Unit, Faculty of Medicine, University of Porto, Porto, Portugal.; Clinical Pathology, São João University Hospital Center, Porto, Portugal.; EPIUnit, Institute of Public Health, University of Porto, Porto, Portugal
| | - Alexandra M Gouveia
- Department of Biomedicine Experimental Biology Unit, Faculty of Medicine of the University of Porto, Porto, Portugal.; Instituto de Investigação e Inovação em Saúde (i3S), Porto, Portugal
| | - Henrique Almeida
- Department of Biomedicine Experimental Biology Unit, Faculty of Medicine of the University of Porto, Porto, Portugal.; Instituto de Investigação e Inovação em Saúde (i3S), Porto, Portugal.; Obstetrics and Gynaecology, Hospital-CUF Porto, Porto, Portugal
| | - Delminda Neves
- Department of Biomedicine Experimental Biology Unit, Faculty of Medicine of the University of Porto, Porto, Portugal.; Instituto de Investigação e Inovação em Saúde (i3S), Porto, Portugal..
| |
Collapse
|
3
|
Lawler JM, Kamal KY, Botchlett RE, Woo SL, Li H, Hord JM, Fluckey JD, Wu C. Metformin ablates high fat diet-induced skeletal muscle hypertrophy and elevation of sarcolemmal GLUT4 when feeding is initiated in young adult male mice. Connect Tissue Res 2025; 66:121-135. [PMID: 40052722 DOI: 10.1080/03008207.2025.2471853] [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: 06/26/2024] [Accepted: 02/20/2025] [Indexed: 03/25/2025]
Abstract
A high-fat diet (HFD) and metabolic disease can impair insulin signaling in skeletal muscle, including a reduction in IRS-1 and GLUT-4 at the cell membrane. Other sarcolemmal proteins (e.g. caveolin-3, nNOS) within the dystrophin-glycoprotein complex (DGC) are partially lost with Type II diabetes. Thus, we hypothesized that a HFD would cause a significant loss of sarcolemmal DGC proteins and GLUT4, and the anti-diabetic drug metformin would mitigate the disruption of the DGC and preserve sarcolemmal GLUT4 on the soleus muscle. Eight-week-old mice were fed a high-fat diet for 12 weeks. After 8 weeks, one-half of the HFD mice received metformin for the remaining 4 weeks. HFD caused a marked increase in soleus muscle mass and fiber cross-sectional area and elevated sarcolemmal GLUT4, even though systemic insulin resistance was greater. HFD-induced muscle hypertrophy and elevated membrane GLUT4 were unexpectedly attenuated by metformin. In addition, IRS-1 positive staining was not reduced by HFD but rather enhanced in the metformin mice fed a high-fat diet. Sarcolemmal staining of dystrophin and caveolin-3 was reduced by HFD but not in the metformin group, while nNOS intensity was unaffected by HFD and metformin. These findings suggest that skeletal muscles in young adult mice can compensate for a high-fat diet and insulin resistance, with a minor disruption of the DGC, by maintaining cell membrane nNOS and IRS-1 and elevating GLUT4. We postulate that a window of compensatory GLUT4 and nNOS signaling allows calorically dense food to enhance skeletal muscle fiber size when introduced in adolescence.
Collapse
Affiliation(s)
- John M Lawler
- Redox Biology & Cell Signaling Laboratory, Kinesiology and Sport Management Department, Texas A&M University, College Station, TX, USA
- Department of Nutrition, Texas A&M University, College Station, TX, USA
| | - Khaled Y Kamal
- Redox Biology & Cell Signaling Laboratory, Kinesiology and Sport Management Department, Texas A&M University, College Station, TX, USA
- Department of Kinesiology, Iowa State University, Ames, IA, USA
| | - Rachel E Botchlett
- Redox Biology & Cell Signaling Laboratory, Kinesiology and Sport Management Department, Texas A&M University, College Station, TX, USA
| | - Shih Lung Woo
- Department of Nutrition, Texas A&M University, College Station, TX, USA
| | - Honggui Li
- Department of Nutrition, Texas A&M University, College Station, TX, USA
| | - Jeff M Hord
- Redox Biology & Cell Signaling Laboratory, Kinesiology and Sport Management Department, Texas A&M University, College Station, TX, USA
- Department of Molecular Physiology and Biophysics, Carver School of Medicine, University of Iowa, Iowa City, IA, USA
| | - James D Fluckey
- Muscle Biology Laboratory, Department of Health and Kinesiology, Texas A&M University, College Station, TX, USA
| | - Chaodong Wu
- Department of Nutrition, Texas A&M University, College Station, TX, USA
| |
Collapse
|
4
|
Guerbette T, Ciesielski V, Brien M, Catheline D, Viel R, Bostoën M, Perrin JB, Burel A, Janvier R, Rioux V, Lan A, Boudry G. Bioenergetic adaptations of small intestinal epithelial cells reduce cell differentiation enhancing intestinal permeability in obese mice. Mol Metab 2025; 92:102098. [PMID: 39814101 PMCID: PMC11795564 DOI: 10.1016/j.molmet.2025.102098] [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: 11/29/2024] [Accepted: 01/08/2025] [Indexed: 01/18/2025] Open
Abstract
OBJECTIVE Obesity and overweight are associated with low-grade inflammation induced by adipose tissue expansion and perpetuated by altered intestinal homeostasis, including increased epithelial permeability. Intestinal epithelium functions are supported by intestinal epithelial cells (IEC) mitochondria function. However, diet-induced obesity (DIO) may impair mitochondrial activity of IEC and consequently, intestinal homeostasis. The aim of the project was to determine whether DIO alters the mitochondrial function of IEC, and what are the consequences on intestinal homeostasis. METHODS C57Bl/6J mice were fed a control diet for 22 weeks or a high fat diet (58 kcal% fat). Bioenergetic adaptations of IEC were evaluated on isolated crypts and villi from mouse jejunum. To determine the link between mitochondrial function and alterations of intestinal homeostasis in response to lipid overload, we used the jejunal epithelial cell line IPEC-J2 in vitro and mouse jejunum organoids. RESULTS Here, we report that DIO in mice induced lipid metabolism adaptations favoring lipid storage in IEC together with reduced number, altered dynamics and diminished oxidative phosphorylation activity of IEC mitochondria. Using the IPEC-J2 cell line, we showed that IEC lipid metabolism and oxidative stress machinery adaptations preceded mitochondrial bioenergetic ones. Moreover, we unraveled the intricate link between IEC energetic status and proliferation / differentiation balance since enhancing mitochondrial function with the AMPK activator AICAR in jejunal organoids reduced proliferation and initiated IEC differentiation and conversely. We confirmed that the reduced IEC mitochondrial function observed in DIO mice was associated with increased proliferation and reduced differentiation, promoting expression of the permissive Cldn2 in the jejunal epithelium of DIO mice. CONCLUSIONS Our study provides new insights into metabolic adaptations of IEC in obesity by revealing that excess lipid intake diminishes mitochondrial number in IEC, reducing IEC differentiation that contribute to increased epithelial permeability.
Collapse
Affiliation(s)
| | - Vincent Ciesielski
- Institut Numecan, INRAE, INSERM, Univ Rennes, Rennes, France; Institut Agro Rennes Angers, Rennes, France
| | - Manon Brien
- Institut Numecan, INRAE, INSERM, Univ Rennes, Rennes, France
| | - Daniel Catheline
- Institut Numecan, INRAE, INSERM, Univ Rennes, Rennes, France; Institut Agro Rennes Angers, Rennes, France
| | - Roselyne Viel
- Univ Rennes, CNRS, Inserm, Biosit UAR 3480 US_S 018, France-BioImaging (ANR-10-INBS-04), plateforme H2P2, Rennes, France
| | - Mégane Bostoën
- Institut Numecan, INRAE, INSERM, Univ Rennes, Rennes, France
| | | | - Agnès Burel
- Plateforme MRic, UMS 3480 BIOSIT, Rennes, France
| | - Régis Janvier
- Institut Numecan, INRAE, INSERM, Univ Rennes, Rennes, France
| | - Vincent Rioux
- Institut Numecan, INRAE, INSERM, Univ Rennes, Rennes, France; Institut Agro Rennes Angers, Rennes, France
| | - Annaïg Lan
- Institut Numecan, INRAE, INSERM, Univ Rennes, Rennes, France; Université Paris-Saclay, AgroParisTech, INRAE, UMR PNCA, 91120, Palaiseau, France
| | - Gaëlle Boudry
- Institut Numecan, INRAE, INSERM, Univ Rennes, Rennes, France.
| |
Collapse
|
5
|
Guo X, Li H, Zhu B, Wang X, Xu Q, Aquino E, Koo M, Li Q, Cai J, Glaser S, Wu C. HFD feeding for seven months abolishes STING disruption-driven but not female sex-based protection against hepatic steatosis and inflammation in mice. J Nutr Biochem 2025; 135:109770. [PMID: 39284534 PMCID: PMC11620956 DOI: 10.1016/j.jnutbio.2024.109770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/07/2024] [Accepted: 09/11/2024] [Indexed: 10/06/2024]
Abstract
Stimulator of interferon genes (STING) is positively correlated with the degrees of liver inflammation in human metabolic dysfunction-associated steatotic liver disease (MASLD). In addition, STING disruption alleviates MASLD in mice fed a high-fat diet (HFD) for 3 months (3-m-HFD). Here we investigated the role of the duration of dietary feeding in regulating MASLD in mice and explored the involvement of STING in sex differences in MASLD. Both male and female STING-disrupted (STINGgt) and wild-type C57BL/6J mice were fed an HFD for 3 or 7 months (7-m-HFD). Additionally, female STINGgt mice upon ovariectomy (OVX) and 3-m-HFD were analyzed for MASLD. Upon 3-m-HFD, STINGgt mice exhibited decreased severity of MASLD compared to control. However, upon 7-m-HFD, STINGgt mice were comparable with wild-type mice in body weight, fat mass, and MASLD. Regarding regulating the liver RNA transcriptome, 7-m-HFD increased the expression of genes indicating proinflammatory activation of various liver cells. Interestingly, the severity of MASLD in female mice was much lighter than in male mice, regardless of STING disruption. Upon OVX, female STINGgt mice showed significantly increased severity of MASLD relative to sham control but were comparable with male STINGgt mice. Upon treatment with 17-beta estradiol (E2), hepatocytes revealed decreased fat deposition while macrophages displayed decreases in lipopolysaccharide-induced phosphorylation of Nfkb p65 and Jnk p46 independent of STING. These results suggest that 7-m-HFD, without altering female sex-based protection, abolishes STING disruption-driven protection of MASLD, likely through causing proinflammatory activation of multiple types of liver cells to offset the effect of STING disruption.
Collapse
Affiliation(s)
- Xinlei Guo
- Department of Nutrition, Texas A&M University, College Station, Texas, USA
| | - Honggui Li
- Department of Nutrition, Texas A&M University, College Station, Texas, USA
| | - Bilian Zhu
- Department of Nutrition, Texas A&M University, College Station, Texas, USA
| | - Xiaoxiao Wang
- Department of Nutrition, Texas A&M University, College Station, Texas, USA
| | - Qian Xu
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas, USA
| | - Eduardo Aquino
- Department of Nutrition, Texas A&M University, College Station, Texas, USA
| | - Minji Koo
- Department of Nutrition, Texas A&M University, College Station, Texas, USA
| | - Qingsheng Li
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - James Cai
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas, USA
| | - Shannon Glaser
- Department of Medical Physiology, Texas A&M University College of Medicine, Byran, Texas, USA.
| | - Chaodong Wu
- Department of Nutrition, Texas A&M University, College Station, Texas, USA.
| |
Collapse
|
6
|
Misceo D, Mocciaro G, D'Amore S, Vacca M. Diverting hepatic lipid fluxes with lifestyles revision and pharmacological interventions as a strategy to tackle steatotic liver disease (SLD) and hepatocellular carcinoma (HCC). Nutr Metab (Lond) 2024; 21:112. [PMID: 39716321 DOI: 10.1186/s12986-024-00871-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 11/13/2024] [Indexed: 12/25/2024] Open
Abstract
Steatotic liver disease (SLD) and Hepatocellular Carcinoma (HCC) are characterised by a substantial rewiring of lipid fluxes caused by systemic metabolic unbalances and/or disrupted intracellular metabolic pathways. SLD is a direct consequence of the interaction between genetic predisposition and a chronic positive energy balance affecting whole-body energy homeostasis and the function of metabolically-competent organs. In this review, we discuss how the impairment of the cross-talk between peripheral organs and the liver stalls glucose and lipid metabolism, leading to unbalances in hepatic lipid fluxes that promote hepatic fat accumulation. We also describe how prolonged metabolic stress builds up toxic lipid species in the liver, and how lipotoxicity and metabolic disturbances drive disease progression by promoting a chronic activation of wound healing, leading to fibrosis and HCC. Last, we provide a critical overview of current state of the art (pre-clinical and clinical evidence) regarding mechanisms of action and therapeutic efficacy of candidate SLD treatment options, and their potential to interfere with SLD/HCC pathophysiology by diverting lipids away from the liver therefore improving metabolic health.
Collapse
Affiliation(s)
- Davide Misceo
- Department of Interdisciplinary Medicine, Clinica Medica "C. Frugoni", "Aldo Moro" University of Bari, Piazza Giulio Cesare 11, 70124, Bari, Italy
| | - Gabriele Mocciaro
- Roger Williams Institute of Liver Studies, Foundation for Liver Research, London, SE5 9NT, UK
| | - Simona D'Amore
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), Clinica Medica "G. Baccelli", "Aldo Moro" University of Bari, 70124, Bari, Italy.
| | - Michele Vacca
- Department of Interdisciplinary Medicine, Clinica Medica "C. Frugoni", "Aldo Moro" University of Bari, Piazza Giulio Cesare 11, 70124, Bari, Italy.
- Roger Williams Institute of Liver Studies, Foundation for Liver Research, London, SE5 9NT, UK.
| |
Collapse
|
7
|
Mohamed AS, Ahmad HM, Sharawy MA, Kamel FMM. The effect of vildagliptin versus metformin on hepatic steatosis in type 2 diabetic patients: a randomized controlled trial. BMC Pharmacol Toxicol 2024; 25:94. [PMID: 39673064 PMCID: PMC11645785 DOI: 10.1186/s40360-024-00818-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 11/20/2024] [Indexed: 12/15/2024] Open
Abstract
BACKGROUND The risk of hepatic steatosis (HS) is elevated in patients with type 2 diabetes mellitus (T2D). Antidiabetic medications may contribute to the prevention or treatment of HS. This study aimed to compare the effects of vildagliptin and metformin on hepatic steatosis in newly diagnosed T2D patients, using the Hepatic Steatosis Index (HSI) and ultrasound grading. METHODS The study included 246 newly diagnosed T2D patients who were randomly assigned to two groups. The first group (117 patients) received 50 mg of vildagliptin orally twice daily. The second group (129 patients) received 500 mg of metformin orally twice daily with meals, and the dosage could be gradually increased by 500 mg per week, up to a maximum daily dose of 2000 mg. Baseline and 6-month follow-up assessments included fasting blood glucose (FBG), HbA1c, weight, body mass index (BMI), waist circumference (WC), hip circumference (HC), the Hepatic Steatosis Index (HSI), and hepatic steatosis grading via ultrasound. RESULTS Both groups showed significant improvements in FBG, HbA1c, weight, BMI, WC, HC, HSI, and ultrasound grading of hepatic steatosis from baseline to the 6-month follow-up (p < 0.001). The metformin group demonstrated significantly greater reductions in weight and BMI compared to the vildagliptin group (p = 0.001 and p = 0.009, respectively). However, there was no significant difference between the two groups in terms of hepatic steatosis improvement on ultrasound. Correlation analysis revealed that HSI was significantly associated with HbA1c, BMI, WC, and HC (p < 0.001 for all), as well as FBG (p = 0.008), but not with age. The lipid profile, particularly total cholesterol and LDL, was identified as a stronger predictor of hepatic steatosis, based on high AUC, sensitivity, and specificity values. CONCLUSION Both vildagliptin and metformin are effective in improving glycemic control in newly diagnosed T2D patients, as evidenced by reductions in FBG and HbA1c levels. Additionally, both drugs significantly reduced the HSI, body weight, and BMI, with metformin showing a more pronounced effect on weight and BMI. Both vildagliptin and metformin effectively decreased hepatic steatosis in T2D patients. Total cholesterol and LDL are important predictors of hepatic steatosis. TRIAL REGISTRATION Trial Registration ID: UMIN000055121, registered on 30/07/2024 (retrospectively registered).
Collapse
Affiliation(s)
- Asmaa S Mohamed
- Clinical Pharmacy and Pharmacy Practice Department, Faculty of Pharmacy, Port said University, Port said, Egypt.
| | - Hosam M Ahmad
- Internal Medicine and Biomedical Chemistry Departments, Egypt Ministry of Health and Population, Minia, Egypt
| | - Mohammed A Sharawy
- Internal Medicine Department, Faculty of Medicine, Minia University, Minia, Egypt
| | - Fatma M M Kamel
- Internal Medicine Department, Faculty of Medicine, Minia University, Minia, Egypt
| |
Collapse
|
8
|
Siddique AHH, Kale PP. Importance of glucose and its metabolism in neurodegenerative disorder, as well as the combination of multiple therapeutic strategies targeting α-synuclein and neuroprotection in the treatment of Parkinson's disease. Rev Neurol (Paris) 2024; 180:736-753. [PMID: 38040547 DOI: 10.1016/j.neurol.2023.08.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 04/04/2023] [Accepted: 08/18/2023] [Indexed: 12/03/2023]
Abstract
According to recent findings, Phosphoglycerate Kinase 1 (pgk-1) enzyme is linked to Parkinson's disease (PD). Mutations in the PGK-1 gene lead to decreases in the pgk-1 enzyme which causes an imbalance in the levels of energy demand and supply. An increase in glycolytic adenosine triphosphate (ATP) production would help alleviate energy deficiency and sustain the acute energetic need of neurons. Neurodegeneration is caused by an imbalance or reduction in ATP levels. Recent data suggest that medications that increase glycolysis and neuroprotection can be used to treat PD. The current study focuses on treatment options for disorders associated with the pgk-1 enzyme, GLP-1, and A2A receptor which can be utilized to treat PD. A combination of metformin and terazosin, exenatide and meclizine, istradefylline and salbutamol treatments may benefit parkinsonism. The review also looked at potential target-specific new techniques that might assist in satisfying unfulfilled requirements in the treatment of PD.
Collapse
Affiliation(s)
- A H H Siddique
- Department of Pharmacology, SVKM's Dr Bhanuben Nanavati College of Pharmacy, V. L. Mehta Road, Vile Parle west, 400056 Mumbai, India.
| | - P P Kale
- Department of Pharmacology, SVKM's Dr Bhanuben Nanavati College of Pharmacy, V. L. Mehta Road, Vile Parle west, 400056 Mumbai, India.
| |
Collapse
|
9
|
Li L, Pan L, Lin Z, Wen J, Tan B, Liu H, Hu Y. Metformin improves insulin resistance, liver healthy and abnormal hepatic glucolipid metabolism via IR/PI 3K/AKT pathway in Ctenopharyngodon idella fed a high-carbohydrate diet. Comp Biochem Physiol C Toxicol Pharmacol 2024; 283:109976. [PMID: 38987002 DOI: 10.1016/j.cbpc.2024.109976] [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: 03/12/2024] [Revised: 06/13/2024] [Accepted: 07/07/2024] [Indexed: 07/12/2024]
Abstract
The effects and underlying mechanisms of metformin which can improve glucose homeostasis of fish have rarely been explored. This experiment aimed to explore the influence of metformin on growth performance, body composition, liver health, hepatic glucolipid metabolic capacity and IR/PI3K/AKT pathway in grass carp (Ctenopharyngodon idella) fed high-carbohydrate diets. A normal diet (Control) and high carbohydrate diets with metformin supplementation (0.00 %, 0.20 %, 0.40 %, 0.60 % and 0.80 %) were configured. Six groups of healthy fish were fed with the experimental diet for eight weeks. The results showed that the growth performance of grass carp was impaired in high carbohydrate diet. Impairment of IR/PI3K/AKT signalling pathway reduced insulin sensitivity, while hepatic oxidative stress damage and decreased immunity affected liver metabolic function. The glycolysis and lipolysis decrease while the gluconeogenesis and fat synthesis increase, which triggers hyperglycaemia and lipid deposition in the body. Metformin supplementation restored the growth performance of grass carp. Metformin improved IR/PI3K/AKT pathway signalling and alleviated insulin resistance, while liver antioxidant capacity and immunity were enhanced resulting in the restoration of liver health. The elevation of glycolysis and lipolysis maintains glycaemic homeostasis and reduces lipid deposition, respectively. These results suggest that metformin supplementation restores liver health and activates the IR/PI3K/AKT signalling pathway, ameliorating insulin resistance and glucose-lipid metabolism disorders caused by a high-carbohydrate diet. As judged by HOMA-IR, the optimum supplementation level of metformin in grass carp (C. idella) fed a high-carbohydrate diet is 0.67 %.
Collapse
Affiliation(s)
- Lixian Li
- Laboratory of Aquatic Animal Nutrition and Feed, Fisheries College, Guangdong Ocean University, Zhanjiang 524088, PR China; Key Laboratory of Aquatic, Livestock and Poultry Feed Science and Technology in South China, Ministry of Agriculture and Rural Affairs, Zhanjiang 524088, PR China; Aquatic Animals Precision Nutrition and High Efficiency Feed Engineering Research Center of Guangdong Province, Zhanjiang 524088, PR China
| | - Ling Pan
- Laboratory of Aquatic Animal Nutrition and Feed, Fisheries College, Guangdong Ocean University, Zhanjiang 524088, PR China; Key Laboratory of Aquatic, Livestock and Poultry Feed Science and Technology in South China, Ministry of Agriculture and Rural Affairs, Zhanjiang 524088, PR China; Aquatic Animals Precision Nutrition and High Efficiency Feed Engineering Research Center of Guangdong Province, Zhanjiang 524088, PR China; Zhanjiang Experimental Station, CATAS. Zhanjiang 524013, PR China
| | - Zhixuan Lin
- Laboratory of Aquatic Animal Nutrition and Feed, Fisheries College, Guangdong Ocean University, Zhanjiang 524088, PR China; Key Laboratory of Aquatic, Livestock and Poultry Feed Science and Technology in South China, Ministry of Agriculture and Rural Affairs, Zhanjiang 524088, PR China; Aquatic Animals Precision Nutrition and High Efficiency Feed Engineering Research Center of Guangdong Province, Zhanjiang 524088, PR China
| | - Jiasheng Wen
- Laboratory of Aquatic Animal Nutrition and Feed, Fisheries College, Guangdong Ocean University, Zhanjiang 524088, PR China; Key Laboratory of Aquatic, Livestock and Poultry Feed Science and Technology in South China, Ministry of Agriculture and Rural Affairs, Zhanjiang 524088, PR China; Aquatic Animals Precision Nutrition and High Efficiency Feed Engineering Research Center of Guangdong Province, Zhanjiang 524088, PR China
| | - Beiping Tan
- Laboratory of Aquatic Animal Nutrition and Feed, Fisheries College, Guangdong Ocean University, Zhanjiang 524088, PR China; Key Laboratory of Aquatic, Livestock and Poultry Feed Science and Technology in South China, Ministry of Agriculture and Rural Affairs, Zhanjiang 524088, PR China; Aquatic Animals Precision Nutrition and High Efficiency Feed Engineering Research Center of Guangdong Province, Zhanjiang 524088, PR China
| | - Hongyu Liu
- Laboratory of Aquatic Animal Nutrition and Feed, Fisheries College, Guangdong Ocean University, Zhanjiang 524088, PR China; Key Laboratory of Aquatic, Livestock and Poultry Feed Science and Technology in South China, Ministry of Agriculture and Rural Affairs, Zhanjiang 524088, PR China; Aquatic Animals Precision Nutrition and High Efficiency Feed Engineering Research Center of Guangdong Province, Zhanjiang 524088, PR China.
| | - Yi Hu
- Hunan Research Center of Engineering Technology for Utilization of Distinctive Aquatic Resource, Hunan Agricultural University, Changsha 410128, PR China
| |
Collapse
|
10
|
Raggi P, Milic J, Manicardi M, Cinque F, Swain MG, Sebastiani G, Guaraldi G. Metabolic dysfunction-associated steatotic liver disease: An opportunity for collaboration between cardiology and hepatology. Atherosclerosis 2024; 392:117523. [PMID: 38522165 DOI: 10.1016/j.atherosclerosis.2024.117523] [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: 01/23/2024] [Revised: 03/07/2024] [Accepted: 03/14/2024] [Indexed: 03/26/2024]
Abstract
Altered metabolic function has many detrimental effects on the body that can manifest as cardiovascular and liver diseases. Traditional approaches to understanding and treating metabolic dysfunction-associated disorders have been organ-centered, leading to silo-type disease care. However, given the broad impact that systemic metabolic dysfunction has on the human body, approaches that simultaneously involve multiple medical specialists need to be developed and encouraged to optimize patient outcomes. In this review, we highlight how several of the treatments developed for cardiac care may have a beneficial effect on the liver and vice versa, suggesting that there is a need to target the disease process, rather than specifically target the cardiovascular or liver specific sequelae of metabolic dysfunction.
Collapse
Affiliation(s)
- Paolo Raggi
- Department of Medicine and Division of Cardiology, University of Alberta, Edmonton, Alberta, Canada.
| | - Jovana Milic
- Department of Surgical, Medical, Dental and Morphological Sciences, University of Modena and Reggio Emilia, Italy
| | - Marcella Manicardi
- Cardiology Department, University of Modena and Reggio Emilia, Policlinico di Modena, Modena, Italy
| | - Felice Cinque
- SC-Medicina Indirizzo Metabolico, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico of Milan, Department of Pathophysiology and Transplantation, University of Milan, Italy; Division of Gastroenterology and Hepatology and Chronic Viral Illness Service, McGill University Health Centre, Montreal, QC, Canada
| | - Mark G Swain
- Department of Medicine, University of Calgary Liver Unit, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Giada Sebastiani
- Division of Gastroenterology and Hepatology and Chronic Viral Illness Service, McGill University Health Centre, Montreal, QC, Canada; Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Giovanni Guaraldi
- Department of Surgical, Medical, Dental and Morphological Sciences, University of Modena and Reggio Emilia, Italy; Department of Infectious Diseases, Azienda Ospedaliero-Universitaria, Policlinico of Modena, Modena, Italy
| |
Collapse
|
11
|
Kumawat BL, Kumar P, Mahla AS, Kumar A, Kumar A, Singh R, Kumar A. A novel action of insulin sensitizing drug as a potential promotor of preovulatory follicles, ovulation rate and prolificacy in sheep. Vet Res Commun 2024; 48:849-863. [PMID: 37957451 DOI: 10.1007/s11259-023-10259-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 11/08/2023] [Indexed: 11/15/2023]
Abstract
The effect of the insulin-sensitizing drug metformin on preovulatory follicle (POF) number, ovulation rate, fetal rate and prolificacy was studied in forty-six cyclic Malpura ewes. After estrus synchronization, the ewes were equally divided into two groups (n = 23). The treatment group (MET) received a daily oral dose of metformin at a rate of 500 mg/animal for approximately 12 weeks, spanning five estrous cycles, as against untreated control (CON). All the ewes were bred to proven rams at the end of treatment. Ovarian ultrasound scans were performed at each estrus and day 9 of each cycle to assess the number and diameter of POFs and corpora lutea (CL), respectively. A comprehensive assessment of circulating hormones including, estradiol, progesterone, androstenedione, and insulin as well as metabolic indicators such as glucose, and lipid profile parameters was performed. At the end of treatment on the day of estrus (E5D0), the treatment showed a stimulatory effect on follicular development with a 53.2% (P < 0.001) increase in the number of POFs. It also increased the ovulation rate by 67.4% (P < 0.01), with a higher proportion (χ2df1 = 10.7, P < 0.001) of ewes in the MET group having multiple ovulations compared to the CON group (82.6 vs. 30.4%). With 1.48 ± 0.12 prolificacy rate in MET ewes, the proportion of ewes giving birth to multiple lambs was 2.9-fold higher than in the CON group. Plasma estradiol, insulin, glucose, total cholesterol, and LDL-cholesterol concentrations were lower (P < 0.05) in the MET ewes than in the CON. The results of the present study indicate that metformin can increase the number of POF, ovulation rate, fetal rate and prolificacy in ewes, while reducing the plasma estradiol, insulin, glucose and cholesterol in MET ewes.
Collapse
Affiliation(s)
- Babu Lal Kumawat
- Animal Physiology and Biochemistry Division, ICAR-Central Sheep and Wool Research Institute, Avikanagar, Rajasthan, 304 501, India
- Department of Veterinary Gynaecology and Obstetrics, College of Veterinary and Animal Sciences, Bikaner (RAJUVAS), Rajasthan, 334 001, India
- Department of Animal Reproduction, Gynaecology and Obstetrics, College of Veterinary and Animal Sciences, Parbhani (MAFSU, Nagpur), Maharashtra, 431 402, India
| | - Pramod Kumar
- Department of Veterinary Gynaecology and Obstetrics, College of Veterinary and Animal Sciences, Bikaner (RAJUVAS), Rajasthan, 334 001, India
| | - Ajit Singh Mahla
- Animal Physiology and Biochemistry Division, ICAR-Central Sheep and Wool Research Institute, Avikanagar, Rajasthan, 304 501, India.
| | - Ashok Kumar
- ICAR-Central Sheep and Wool Research Institute, Arid Region Campus, Bikaner, Rajasthan, 334 006, India
| | - Amit Kumar
- Department of Veterinary Gynaecology and Obstetrics, College of Veterinary and Animal Sciences, Bikaner (RAJUVAS), Rajasthan, 334 001, India
| | - Raghvendar Singh
- Animal Physiology and Biochemistry Division, ICAR-Central Sheep and Wool Research Institute, Avikanagar, Rajasthan, 304 501, India
| | - Arun Kumar
- ICAR-Central Sheep and Wool Research Institute, Avikanagar, Rajasthan, 304 501, India
| |
Collapse
|
12
|
Al Hashmi K, Giglio RV, Pantea Stoian A, Patti AM, Al Waili K, Al Rasadi K, Ciaccio M, Rizzo M. Metabolic dysfunction-associated fatty liver disease: current therapeutic strategies. Front Nutr 2024; 11:1355732. [PMID: 38567250 PMCID: PMC10985255 DOI: 10.3389/fnut.2024.1355732] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 03/11/2024] [Indexed: 04/04/2024] Open
Abstract
The definition of "Metabolic Associated Fatty Liver Disease - MAFLD" has replaced the previous definition of Nonalcoholic Fatty Liver Disease (NAFLD), because cardiometabolic criteria have been added for the prevention of cardiological risk in these patients. This definition leads to an in-depth study of the bidirectional relationships between hepatic steatosis, Type 2 Diabetes Mellitus (T2DM), Cardiovascular Disease (CVD) and/or their complications. Lifestyle modification, which includes correct nutrition combined with regular physical activity, represents the therapeutic cornerstone of MAFLD. When therapy is required, there is not clear accord on how to proceed in an optimal way with nutraceutical or pharmacological therapy. Numerous studies have attempted to identify nutraceuticals with a significant benefit on metabolic alterations and which contribute to the improvement of hepatic steatosis. Several evidences are supporting the use of silymarin, berberine, curcumin, Nigella sativa, Ascophyllum nodosum, and Fucus vesiculosus, vitamin E, coenzyme Q10 and Omega-3. However, more evidence regarding the long-term efficacy and safety of these compounds are required. There is numerous evidence that highlights the use of therapies such as incretins or the use of Proprotein Convertase Subtilisin/Kexin type 9 (PCSK9) inhibitors or other similar therapies which, by assisting existing therapies for pathologies such as diabetes, hypertension, insulin resistance, have given a breakthrough in prevention and the reduction of cardiometabolic risk. This review gave an overview of the current therapeutic strategies that are expected to aid in the treatment and prevention of MAFLD.
Collapse
Affiliation(s)
- Khamis Al Hashmi
- Department of Physiology, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Rosaria Vincenza Giglio
- Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, Italy
- Department of Laboratory Medicine, University Hospital, Palermo, Italy
| | - Anca Pantea Stoian
- Department of Diabetes, Nutrition and Metabolic Diseases, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Angelo Maria Patti
- Internal Medicine Unit, “Vittorio Emanuele II” Hospital, Castelvetrano, Italy
| | - Khalid Al Waili
- Department of Biochemistry, Sultan Qaboos University Hospital, Muscat, Oman
| | - Khalid Al Rasadi
- Department of Biochemistry, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
- Medical Research Center, Sultan Qaboos University, Muscat, Oman
| | - Marcello Ciaccio
- Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, Italy
- Department of Laboratory Medicine, University Hospital, Palermo, Italy
| | - Manfredi Rizzo
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| |
Collapse
|
13
|
Lin H, Ao H, Guo G, Liu M. The Role and Mechanism of Metformin in Inflammatory Diseases. J Inflamm Res 2023; 16:5545-5564. [PMID: 38026260 PMCID: PMC10680465 DOI: 10.2147/jir.s436147] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 11/17/2023] [Indexed: 12/01/2023] Open
Abstract
Metformin is a classical drug used to treat type 2 diabetes. With the development of research on metformin, it has been found that metformin also has several advantages aside from its hypoglycemic effect, such as anti-inflammatory, anti-aging, anti-cancer, improving intestinal flora, and other effects. The prevention of inflammation is critical because chronic inflammation is associated with numerous diseases of considerable public health. Therefore, there has been growing interest in the role of metformin in treating various inflammatory conditions. However, the precise anti-inflammatory mechanisms of metformin were inconsistent in the reported studies. Thus, this review aims to summarize various currently known possible mechanisms of metformin involved in inflammatory diseases and provide references for the clinical application of metformin.
Collapse
Affiliation(s)
- Huan Lin
- Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital of Nanchang University, Nanchang, People’s Republic of China
| | - Haiyong Ao
- Jiangxi Key Laboratory of Nanobiomaterials & School of Materials Science and Engineering, East China Jiaotong University, Nanchang, Jiangxi, People’s Republic of China
| | - Guanghua Guo
- Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital of Nanchang University, Nanchang, People’s Republic of China
| | - Mingzhuo Liu
- Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital of Nanchang University, Nanchang, People’s Republic of China
| |
Collapse
|
14
|
He W, Wang X, Chen M, Li C, Chen W, Pan L, Cui Y, Yu Z, Wu G, Yang Y, Xu M, Dong Z, Ma K, Wang J, He Z. Metformin reduces hepatocarcinogenesis by inducing downregulation of Cyp26a1 and CD8 + T cells. Clin Transl Med 2023; 13:e1465. [PMID: 37997519 PMCID: PMC10668005 DOI: 10.1002/ctm2.1465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 10/12/2023] [Accepted: 10/19/2023] [Indexed: 11/25/2023] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a highly heterogeneous cancer with major challenges in both prevention and therapy. Metformin, adenosine monophosphate-activated protein kinase (AMPK) activator, has been suggested to reduce the incidence of HCC when used for patients with diabetes in preclinical and clinical studies. However, the possible effects of metformin and their mechanisms of action in non-diabetic HCC have not been adequately investigated. METHODS Fah-/- mice were used to construct a liver-injury-induced non-diabetic HCC model for exploring hepatocarcinogenesis and therapeutic potential of metformin. Changes in relevant tumour and biochemical indicators were measured. Bulk and single-cell RNA-sequencing analyses were performed to validate the crucial role of proinflammatory/pro-tumour CD8+ T cells. In vitro and in vivo experiments were performed to confirm Cyp26a1-related antitumour mechanisms of metformin. RESULTS RNA-sequencing analysis showed that chronic liver injury led to significant changes in AMPK-, glucose- and retinol metabolism-related pathways in Fah-/- mice. Metformin prevented the formation of non-diabetic HCC in Fah-/- mice with chronic liver injury. Cyp26a1 ddexpression in hepatocytes was significantly suppressed after metformin treatment. Moreover, downregulation of Cyp26a1 occurred in conjunction with increased levels of all-trans-retinoic acid (atRA), which is involved in the activation of metformin-suppressed hepatocarcinogenesis in Fah-/- mice. In contrast, both CD8+ T-cell infiltration and proinflammatory/pro-tumour cytokines in the liver were significantly upregulated in Fah-/- mice during chronic liver injury, which was notably reversed by either metformin or atRA treatment. Regarding mechanisms, metformin regulated the decrease in Cyp26a1 enzyme expression and increased atRA expression via the AMPK/STAT3/Gadd45β/JNK/c-Jun pathway. CONCLUSIONS Metformin inhibits non-diabetic HCC by upregulating atRA levels and downregulating CD8+ T cells. This is the first reporting that the traditional drug metformin regulates the metabolite atRA via the Cyp26a1-involved pathway. The present study provides a potential application of metformin and atRA in non-diabetic HCC.
Collapse
Affiliation(s)
- Weizhi He
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East HospitalSchool of Life Sciences and TechnologyTongji University School of MedicineShanghaiChina
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghaiChina
- Fudan University Shanghai Cancer Center, International Co‐Laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Shanghai Medical College of Fudan University, Institutes of Biomedical SciencesShanghai Key Laboratory of Medical EpigeneticsShanghaiChina
| | - Xicheng Wang
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East HospitalSchool of Life Sciences and TechnologyTongji University School of MedicineShanghaiChina
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghaiChina
| | - Miaomiao Chen
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East HospitalSchool of Life Sciences and TechnologyTongji University School of MedicineShanghaiChina
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghaiChina
| | - Chong Li
- Zhoupu Community Health Service Center of Pudong New AreaShanghaiChina
| | - Wenjian Chen
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East HospitalSchool of Life Sciences and TechnologyTongji University School of MedicineShanghaiChina
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghaiChina
| | - Lili Pan
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East HospitalSchool of Life Sciences and TechnologyTongji University School of MedicineShanghaiChina
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghaiChina
| | - Yangyang Cui
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East HospitalSchool of Life Sciences and TechnologyTongji University School of MedicineShanghaiChina
- Postgraduate Training Base of Shanghai East HospitalJinzhou Medical UniversityJinzhouLiaoningChina
| | - Zhao Yu
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East HospitalSchool of Life Sciences and TechnologyTongji University School of MedicineShanghaiChina
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghaiChina
| | - Guoxiu Wu
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East HospitalSchool of Life Sciences and TechnologyTongji University School of MedicineShanghaiChina
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghaiChina
| | - Yang Yang
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East HospitalSchool of Life Sciences and TechnologyTongji University School of MedicineShanghaiChina
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghaiChina
| | - Mingyang Xu
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East HospitalSchool of Life Sciences and TechnologyTongji University School of MedicineShanghaiChina
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghaiChina
| | - Zhaoxuan Dong
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East HospitalSchool of Life Sciences and TechnologyTongji University School of MedicineShanghaiChina
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghaiChina
| | - Keming Ma
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East HospitalSchool of Life Sciences and TechnologyTongji University School of MedicineShanghaiChina
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghaiChina
| | - Jinghan Wang
- Department of Hepatobiliary and Pancreatic SurgeryShanghai East Hospital, Tongji UniversityShanghaiChina
| | - Zhiying He
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East HospitalSchool of Life Sciences and TechnologyTongji University School of MedicineShanghaiChina
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghaiChina
| |
Collapse
|
15
|
Osorio-Llanes E, Villamizar-Villamizar W, Ospino Guerra MC, Díaz-Ariza LA, Castiblanco-Arroyave SC, Medrano L, Mengual D, Belón R, Castellar-López J, Sepúlveda Y, Vásquez-Trincado C, Chang AY, Bolívar S, Mendoza-Torres E. Effects of Metformin on Ischemia/Reperfusion Injury: New Evidence and Mechanisms. Pharmaceuticals (Basel) 2023; 16:1121. [PMID: 37631036 PMCID: PMC10459572 DOI: 10.3390/ph16081121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/02/2023] [Accepted: 08/03/2023] [Indexed: 08/27/2023] Open
Abstract
The search for new drugs with the potential to ensure therapeutic success in the treatment of cardiovascular diseases has become an essential pathway to follow for health organizations and committees around the world. In June 2021, the World Health Organization listed cardiovascular diseases as one of the main causes of death worldwide, representing 32% of them. The most common is coronary artery disease, which causes the death of cardiomyocytes, the cells responsible for cardiac contractility, through ischemia and subsequent reperfusion, which leads to heart failure in the medium and short term. Metformin is one of the most-used drugs for the control of diabetes, which has shown effects beyond the control of hyperglycemia. Some of these effects are mediated by the regulation of cellular energy metabolism, inhibiting apoptosis, reduction of cell death through regulation of autophagy and reduction of mitochondrial dysfunction with further reduction of oxidative stress. This suggests that metformin may attenuate left ventricular dysfunction induced by myocardial ischemia; preclinical and clinical trials have shown promising results, particularly in the setting of acute myocardial infarction. This is a review of the molecular and pharmacological mechanisms of the cardioprotective effects of metformin during myocardial ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Estefanie Osorio-Llanes
- Advanced Biomedicine Research Group, Faculty of Health Sciences, Universidad Libre de Colombia, Seccional Barranquilla, Barranquilla 081001, Colombia; (E.O.-L.); (W.V.-V.); (M.C.O.G.); (L.A.D.-A.); (S.C.C.-A.); (R.B.); (J.C.-L.)
- Allied Research Society S.A.S., Barranquilla 080001, Colombia;
- Global Disease Research Colombia, Barranquilla 080001, Colombia
| | - Wendy Villamizar-Villamizar
- Advanced Biomedicine Research Group, Faculty of Health Sciences, Universidad Libre de Colombia, Seccional Barranquilla, Barranquilla 081001, Colombia; (E.O.-L.); (W.V.-V.); (M.C.O.G.); (L.A.D.-A.); (S.C.C.-A.); (R.B.); (J.C.-L.)
| | - María Clara Ospino Guerra
- Advanced Biomedicine Research Group, Faculty of Health Sciences, Universidad Libre de Colombia, Seccional Barranquilla, Barranquilla 081001, Colombia; (E.O.-L.); (W.V.-V.); (M.C.O.G.); (L.A.D.-A.); (S.C.C.-A.); (R.B.); (J.C.-L.)
| | - Luis Antonio Díaz-Ariza
- Advanced Biomedicine Research Group, Faculty of Health Sciences, Universidad Libre de Colombia, Seccional Barranquilla, Barranquilla 081001, Colombia; (E.O.-L.); (W.V.-V.); (M.C.O.G.); (L.A.D.-A.); (S.C.C.-A.); (R.B.); (J.C.-L.)
| | - Sara Camila Castiblanco-Arroyave
- Advanced Biomedicine Research Group, Faculty of Health Sciences, Universidad Libre de Colombia, Seccional Barranquilla, Barranquilla 081001, Colombia; (E.O.-L.); (W.V.-V.); (M.C.O.G.); (L.A.D.-A.); (S.C.C.-A.); (R.B.); (J.C.-L.)
| | - Luz Medrano
- Healthcare Pharmacy and Pharmacology Research Group, Faculty of Chemistry and Pharmacy, Universidad del Atlántico, Barranquilla 081007, Colombia; (L.M.); (D.M.); (S.B.)
| | - Daniela Mengual
- Healthcare Pharmacy and Pharmacology Research Group, Faculty of Chemistry and Pharmacy, Universidad del Atlántico, Barranquilla 081007, Colombia; (L.M.); (D.M.); (S.B.)
| | - Ricardo Belón
- Advanced Biomedicine Research Group, Faculty of Health Sciences, Universidad Libre de Colombia, Seccional Barranquilla, Barranquilla 081001, Colombia; (E.O.-L.); (W.V.-V.); (M.C.O.G.); (L.A.D.-A.); (S.C.C.-A.); (R.B.); (J.C.-L.)
| | - Jairo Castellar-López
- Advanced Biomedicine Research Group, Faculty of Health Sciences, Universidad Libre de Colombia, Seccional Barranquilla, Barranquilla 081001, Colombia; (E.O.-L.); (W.V.-V.); (M.C.O.G.); (L.A.D.-A.); (S.C.C.-A.); (R.B.); (J.C.-L.)
| | - Yanireth Sepúlveda
- Allied Research Society S.A.S., Barranquilla 080001, Colombia;
- Global Disease Research Colombia, Barranquilla 080001, Colombia
| | - César Vásquez-Trincado
- Escuela de Química y Farmacia, Facultad de Medicina, Universidad Andres Bello, Santiago 8370134, Chile;
| | - Aileen Y. Chang
- Department of Medicine, Faculty of Medicine, Foggy Bottom Campus, George Washington University, Washington, DC 20052, USA;
| | - Samir Bolívar
- Healthcare Pharmacy and Pharmacology Research Group, Faculty of Chemistry and Pharmacy, Universidad del Atlántico, Barranquilla 081007, Colombia; (L.M.); (D.M.); (S.B.)
| | - Evelyn Mendoza-Torres
- Advanced Biomedicine Research Group, Faculty of Health Sciences, Universidad Libre de Colombia, Seccional Barranquilla, Barranquilla 081001, Colombia; (E.O.-L.); (W.V.-V.); (M.C.O.G.); (L.A.D.-A.); (S.C.C.-A.); (R.B.); (J.C.-L.)
| |
Collapse
|
16
|
Sarkar K, Bank S, Chatterjee A, Dutta K, Das A, Chakraborty S, Paul N, Sarkar J, De S, Ghosh S, Acharyya K, Chattopadhyay D, Das M. Hyaluronic acid-graphene oxide quantum dots nanoconjugate as dual purpose drug delivery and therapeutic agent in meta-inflammation. J Nanobiotechnology 2023; 21:246. [PMID: 37528408 PMCID: PMC10394801 DOI: 10.1186/s12951-023-02015-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 07/21/2023] [Indexed: 08/03/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) predominantly considered a metabolic disease is now being considered an inflammatory disease as well due to the involvement of meta-inflammation. Obesity-induced adipose tissue inflammation (ATI) is one of the earliest phenomena in the case of meta-inflammation, leading to the advent of insulin resistance (IR) and T2DM. The key events of ATI are orchestrated by macrophages, which aggravate the inflammatory state in the tissue upon activation, ultimately leading to systemic chronic low-grade inflammation and Non-Alcoholic Steatohepatitis (NASH) through the involvement of proinflammatory cytokines. The CD44 receptor on macrophages is overexpressed in ATI, NASH, and IR. Therefore, we developed a CD44 targeted Hyaluronic Acid functionalized Graphene Oxide Quantum Dots (GOQD-HA) nanocomposite for tissue-specific delivery of metformin. Metformin-loaded GOQD-HA (GOQD-HA-Met) successfully downregulated the expression of proinflammatory cytokines and restored antioxidant status at lower doses than free metformin in both palmitic acid-induced RAW264.7 cells and diet induced obese mice. Our study revealed that the GOQD-HA nanocarrier enhanced the efficacy of Metformin primarily by acting as a therapeutic agent apart from being a drug delivery platform. The therapeutic properties of GOQD-HA stem from both HA and GOQD having anti-inflammatory and antioxidant properties respectively. This study unravels the function of GOQD-HA as a targeted drug delivery option for metformin in meta-inflammation where the nanocarrier itself acts as a therapeutic agent.
Collapse
Affiliation(s)
- Kunal Sarkar
- Department of Zoology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata, 700019, India
| | - Sarbashri Bank
- Department of Zoology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata, 700019, India
| | - Arindam Chatterjee
- Department of Zoology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata, 700019, India
| | - Koushik Dutta
- Department of Polymer Science and Technology, University of Calcutta, 92 A.P.C. Road, Kolkata, 700009, India
| | - Anwesha Das
- Department of Zoology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata, 700019, India
| | - Santanu Chakraborty
- Department of Zoology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata, 700019, India
| | - Nirvika Paul
- Department of Zoology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata, 700019, India
| | - Jit Sarkar
- Molecular and Applied Mycology and Plant Pathology Laboratory, Department of Botany, University of Calcutta, Kolkata, 700019, India
| | - Sriparna De
- Department of Allied Health Sciences, Brainware University, Kolkata, 700129, India
| | - Sudakshina Ghosh
- Department of Zoology, Vidyasagar College for Women, Kolkata, 700006, India
| | - Krishnendu Acharyya
- Molecular and Applied Mycology and Plant Pathology Laboratory, Department of Botany, University of Calcutta, Kolkata, 700019, India
| | - Dipankar Chattopadhyay
- Department of Polymer Science and Technology, University of Calcutta, 92 A.P.C. Road, Kolkata, 700009, India
| | - Madhusudan Das
- Department of Zoology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata, 700019, India.
| |
Collapse
|
17
|
Matsumori A. Nuclear Factor-κB is a Prime Candidate for the Diagnosis and Control of Inflammatory Cardiovascular Disease. Eur Cardiol 2023; 18:e40. [PMID: 37456770 PMCID: PMC10345985 DOI: 10.15420/ecr.2023.10] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 03/17/2023] [Indexed: 07/18/2023] Open
Abstract
Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) is responsible for the regulation of genes involved in inflammation and immune responses. NF-κB may play an important role in cardiovascular diseases (CVDs), atherosclerosis and diabetes. Several therapeutic agents used for the treatment of CVDs and diabetes, such as pimobendan and sodium-glucose cotransporter 2 inhibitors, exert anti-inflammatory effects by inhibiting NF-κB activation; anti-inflammatory therapy may have beneficial effects in CVDs and diabetes. Several pharmacological agents and natural compounds may inhibit NF-κB, and these agents alone or in combination may be used to treat various inflammatory diseases. Immunoglobulin-free light chains could be surrogate biomarkers of NF-κB activation and may be useful for evaluating the efficacy of these agents. This review discusses recent advances in our understanding of how the NF-κB signalling pathway controls inflammation, metabolism and immunity, and how improved knowledge of these pathways may lead to better diagnostics and therapeutics for various human diseases.
Collapse
Affiliation(s)
- Akira Matsumori
- Clinical Research Institute, National Hospital Organization, Kyoto Medical Center Kyoto, Japan
| |
Collapse
|
18
|
Dong Y, Qi Y, Jiang H, Mi T, Zhang Y, Peng C, Li W, Zhang Y, Zhou Y, Zang Y, Li J. The development and benefits of metformin in various diseases. Front Med 2023; 17:388-431. [PMID: 37402952 DOI: 10.1007/s11684-023-0998-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 04/01/2023] [Indexed: 07/06/2023]
Abstract
Metformin has been used for the treatment of type II diabetes mellitus for decades due to its safety, low cost, and outstanding hypoglycemic effect clinically. The mechanisms underlying these benefits are complex and still not fully understood. Inhibition of mitochondrial respiratory-chain complex I is the most described downstream mechanism of metformin, leading to reduced ATP production and activation of AMP-activated protein kinase (AMPK). Meanwhile, many novel targets of metformin have been gradually discovered. In recent years, multiple pre-clinical and clinical studies are committed to extend the indications of metformin in addition to diabetes. Herein, we summarized the benefits of metformin in four types of diseases, including metabolic associated diseases, cancer, aging and age-related diseases, neurological disorders. We comprehensively discussed the pharmacokinetic properties and the mechanisms of action, treatment strategies, the clinical application, the potential risk of metformin in various diseases. This review provides a brief summary of the benefits and concerns of metformin, aiming to interest scientists to consider and explore the common and specific mechanisms and guiding for the further research. Although there have been countless studies of metformin, longitudinal research in each field is still much warranted.
Collapse
Affiliation(s)
- Ying Dong
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yingbei Qi
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Haowen Jiang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Tian Mi
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yunkai Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chang Peng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Wanchen Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yongmei Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Yubo Zhou
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528400, China.
| | - Yi Zang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- Lingang Laboratory, Shanghai, 201203, China.
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.
| | - Jia Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Open Studio for Druggability Research of Marine Natural Products, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, 266237, China.
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China.
| |
Collapse
|
19
|
Li H, Guo X, Aquino E, Wu C. Mini review: STING activation during non-alcoholic fatty liver disease. Front Nutr 2023; 10:1139339. [PMID: 36937350 PMCID: PMC10014842 DOI: 10.3389/fnut.2023.1139339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 02/14/2023] [Indexed: 03/05/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is one of the most common chronic diseases serving as a major threat to human health. While the pathogenesis of NAFLD is multi-factorial, inflammation is considered a critical factor driving the development and progression of NAFLD phenotype, including liver fibrosis. As an essential mediator of innate immunity, stimulator of interferon genes (STING) functions to promote anti-viral immunity. Accumulating evidence also indicates that STING functions to promote the proinflammatory activation of several types of liver cells, especially macrophages/Kupffer cells, in a manner independent of interferon production. Over the past several years, a significant body of literature has validated a detrimental role for STING in regulating the pathogenesis of hepatic steatosis and inflammation. In particular, the STING in macrophages/Kupffer cells has attracted much attention due to its importance in not only enhancing macrophage proinflammatory activation, but also generating macrophage-derived mediators to increase hepatocyte fat deposition and proinflammatory responses, and to activate hepatic stellate cell fibrogenic activation. Both intracellular and extracellular signals are participating in STING activation in macrophages, thereby critically contributing to NAFLD phenotype. This mini review summarizes recent advances on how STING is activated in macrophages in the context of NAFLD pathophysiology.
Collapse
Affiliation(s)
| | | | | | - Chaodong Wu
- Department of Nutrition, Texas A&M University, College Station, TX, United States
| |
Collapse
|
20
|
Arefin A, Gage MC. Metformin, Empagliflozin, and Their Combination Modulate Ex-Vivo Macrophage Inflammatory Gene Expression. Int J Mol Sci 2023; 24:ijms24054785. [PMID: 36902218 PMCID: PMC10003317 DOI: 10.3390/ijms24054785] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 02/09/2023] [Accepted: 02/23/2023] [Indexed: 03/06/2023] Open
Abstract
Type-2 Diabetes Mellitus is a complex, chronic illness characterized by persistent high blood glucose levels. Patients can be prescribed anti-diabetes drugs as single agents or in combination depending on the severity of their condition. Metformin and empagliflozin are two commonly prescribed anti-diabetes drugs which reduce hyperglycemia, however their direct effects on macrophage inflammatory responses alone or in combination are unreported. Here, we show that metformin and empagliflozin elicit proinflammatory responses on mouse bone-marrow-derived macrophages with single agent challenge, which are modulated when added in combination. In silico docking experiments suggested that empagliflozin can interact with both TLR2 and DECTIN1 receptors, and we observed that both empagliflozin and metformin increase expression of Tlr2 and Clec7a. Thus, findings from this study suggest that metformin and empagliflozin as single agents or in combination can directly modulate inflammatory gene expression in macrophages and upregulate the expression of their receptors.
Collapse
Affiliation(s)
- Adittya Arefin
- Wolfson Institute for Biomedical Research, Division of Medicine, University College London, Gower Street, London WC1E 6BT, UK
| | - Matthew C. Gage
- Department of Comparative Biomedical Sciences, Royal Veterinary College, 4 Royal College Street, London NW1 0TU, UK
- Correspondence:
| |
Collapse
|
21
|
The Synergistic Action of Metformin and Glycyrrhiza uralensis Fischer Extract Alleviates Metabolic Disorders in Mice with Diet-Induced Obesity. Int J Mol Sci 2023; 24:ijms24020936. [PMID: 36674447 PMCID: PMC9862386 DOI: 10.3390/ijms24020936] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/31/2022] [Accepted: 01/02/2023] [Indexed: 01/06/2023] Open
Abstract
Metformin, an antidiabetic drug, and Glycyrrhiza uralensis Fischer (GU), an oriental medicinal herb, have been reported to exert anti-obesity effects. This study investigated the synergistic action of metformin and GU in improving diet-induced obesity. Mice were fed a normal diet, a high-fat diet (HFD), or HFD + 0.015% GU water extract for 8 weeks. The HFD and GU groups were then randomly divided into two groups and fed the following diets for the next 8 weeks: HFD with 50 mg/kg metformin (HFDM) and GU with 50 mg/kg metformin (GUM). GUM prevented hepatic steatosis and adiposity by suppressing expression of mRNAs and enzyme activities related to lipogenesis in the liver and upregulating the expression of adipocyte mRNAs associated with fatty acid oxidation and lipolysis, and as a result, improved dyslipidemia. Moreover, GUM improved glucose homeostasis by inducing glucose uptake in tissues and upregulating mRNA expressions associated with glycolysis in the liver and muscle through AMP-activated protein kinase activation. GUM also improved inflammation by increasing antioxidant activity in the liver and erythrocytes and decreasing inflammatory cytokine productions. Here, we demonstrate that GU and metformin exert synergistic action in the prevention of obesity and its complications.
Collapse
|
22
|
Ciric D, Kravic-Stevovic T, Bumbasirevic V, Petricevic S, Jovanovic S, Trajkovic V, Martinovic T. Effects of metformin and simvastatin treatment on ultrastructural features of liver macrophages in HFD mice. Ultrastruct Pathol 2023; 47:1-11. [PMID: 36520527 DOI: 10.1080/01913123.2022.2156639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Type 2 diabetes is a major health burden to the society. Macrophages and liver inflammation emerged as important factors in its development. We investigated ultrastructural changes in the liver, with a special emphasis on macrophages in high fat diet (HFD) fed C57BL/6 J mice treated with metformin or simvastatin, two drugs that are used frequently in diabetes. Both metformin and simvastatin reduced the liver damage in HFD fed animals, manifested as the prevention of nonalcoholic steatohepatitis development and reduced activation and number of macrophages in the liver, as well as the percentage of these cells with lipid droplets in the cytoplasm compared to untreated HFD animals. In contrast with untreated HFD-fed animals, lipid droplets were not observed in lysosomes of macrophages in HFD animals treated with metformin and simvastatin. These findings provide new insight into the effects of metformin and simvastatin on the liver in this experimental model of type 2 diabetes and provide further rationale for implementation of statins in the therapeutic regimens in this disease.
Collapse
Affiliation(s)
- Darko Ciric
- Institute of Histology and Embryology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Tamara Kravic-Stevovic
- Institute of Histology and Embryology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Vladimir Bumbasirevic
- Institute of Histology and Embryology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia.,Department of Medical Science Serbian Academy of Sciences and Arts, Belgrade, Serbia
| | - Sasa Petricevic
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Sofija Jovanovic
- Institute of Histology and Embryology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Vladimir Trajkovic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Tamara Martinovic
- Institute of Histology and Embryology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
23
|
Bao Y, Han X, Liu D, Tan Z, Deng Y. Gut microbiota: The key to the treatment of metabolic syndrome in traditional Chinese medicine - a case study of diabetes and nonalcoholic fatty liver disease. Front Immunol 2022; 13:1072376. [PMID: 36618372 PMCID: PMC9816483 DOI: 10.3389/fimmu.2022.1072376] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 12/09/2022] [Indexed: 12/25/2022] Open
Abstract
Metabolic syndrome mainly includes obesity, type 2 diabetes (T2DM), alcoholic fatty liver (NAFLD) and cardiovascular diseases. According to the ancient experience philosophy of Yin-Yang, monarch-minister compatibility of traditional Chinese medicine, prescription is given to treat diseases, which has the advantages of small toxic and side effects and quick effect. However, due to the diversity of traditional Chinese medicine ingredients and doubts about the treatment theory of traditional Chinese medicine, the mechanism of traditional Chinese medicine is still in doubt. Gastrointestinal tract is an important part of human environment, and participates in the occurrence and development of diseases. In recent years, more and more TCM researches have made intestinal microbiome a new frontier for understanding and treating diseases. Clinically, nonalcoholic fatty liver disease (NAFLD) and diabetes mellitus (DM) often co-occur. Our aim is to explain the mechanism of interaction between gastrointestinal microbiome and traditional Chinese medicine (TCM) or traditional Chinese medicine formula to treat DM and NAFLD. Traditional Chinese medicine may treat these two diseases by influencing the composition of intestinal microorganisms, regulating the metabolism of intestinal microorganisms and transforming Chinese medicinal compounds.
Collapse
Affiliation(s)
- Yang Bao
- Department of Endosecretory Metabolic Diseases, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Xiao Han
- Department of Endosecretory Metabolic Diseases, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Da Liu
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, Jilin, China,*Correspondence: Yongzhi Deng, ; Zhaolin Tan, ; Da Liu,
| | - Zhaolin Tan
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, Jilin, China,*Correspondence: Yongzhi Deng, ; Zhaolin Tan, ; Da Liu,
| | - Yongzhi Deng
- Department of Acupuncture and Massage, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin, China,*Correspondence: Yongzhi Deng, ; Zhaolin Tan, ; Da Liu,
| |
Collapse
|
24
|
Baiges-Gaya G, Rodríguez-Tomàs E, Castañé H, Jiménez-Franco A, Amigó N, Camps J, Joven J. Combining Dietary Intervention with Metformin Treatment Enhances Non-Alcoholic Steatohepatitis Remission in Mice Fed a High-Fat High-Sucrose Diet. Biomolecules 2022; 12:biom12121787. [PMID: 36551216 PMCID: PMC9775246 DOI: 10.3390/biom12121787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 11/23/2022] [Accepted: 11/25/2022] [Indexed: 12/05/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH) are serious health concerns for which lifestyle interventions are the only effective first-line treatment. Dietary interventions are effective in body weight reduction, but not in improving insulin sensitivity and hepatic lipid mobilization. Conversely, metformin increases insulin sensitivity and promotes the inhibition of de novo hepatic lipogenesis. In this study, we evaluated the metformin effectiveness in NASH prevention and treatment, when combined with dietary intervention in male mice fed a high-fat high-sucrose diet (HFHSD). Eighty 5-week-old C57BL/6J male mice were fed a chow or HFHSD diet and sacrificed at 20 or 40 weeks. The HFHSD-fed mice developed NASH after 20 weeks. Lipoprotein and lipidomic analyses showed that the changes associated with diet were not prevented by metformin administration. HFHSD-fed mice subject to dietary intervention combined with metformin showed a 19.6% body weight reduction compared to 9.8% in those mice subjected to dietary intervention alone. Lower hepatic steatosis scores were induced. We conclude that metformin should not be considered a preventive option for NAFLD, but it is effective in the treatment of this disorder when combined with dietary intervention.
Collapse
Affiliation(s)
- Gerard Baiges-Gaya
- Department of Medicine and Surgery, Rovira i Virgili University (URV), 43201 Reus, Spain
- Unitat de Recerca Biomèdica (URB-CRB), Hospital Universitari de Sant Joan, Institut d’Investigació Santiària Pere i Virgili (IISPV), 43201 Reus, Spain
| | - Elisabet Rodríguez-Tomàs
- Department of Medicine and Surgery, Rovira i Virgili University (URV), 43201 Reus, Spain
- Unitat de Recerca Biomèdica (URB-CRB), Hospital Universitari de Sant Joan, Institut d’Investigació Santiària Pere i Virgili (IISPV), 43201 Reus, Spain
| | - Helena Castañé
- Department of Medicine and Surgery, Rovira i Virgili University (URV), 43201 Reus, Spain
- Unitat de Recerca Biomèdica (URB-CRB), Hospital Universitari de Sant Joan, Institut d’Investigació Santiària Pere i Virgili (IISPV), 43201 Reus, Spain
| | - Andrea Jiménez-Franco
- Department of Medicine and Surgery, Rovira i Virgili University (URV), 43201 Reus, Spain
| | - Núria Amigó
- Department of Medicine and Surgery, Rovira i Virgili University (URV), 43201 Reus, Spain
- CIBER of Diabetes and Associated Metabolic Disease (CIBERDEM), ISCIII, 28029 Madrid, Spain
- Biosfer Teslab, 43201 Reus, Spain
| | - Jordi Camps
- Department of Medicine and Surgery, Rovira i Virgili University (URV), 43201 Reus, Spain
- Unitat de Recerca Biomèdica (URB-CRB), Hospital Universitari de Sant Joan, Institut d’Investigació Santiària Pere i Virgili (IISPV), 43201 Reus, Spain
- Correspondence: (J.C.); (J.J.)
| | - Jorge Joven
- Department of Medicine and Surgery, Rovira i Virgili University (URV), 43201 Reus, Spain
- Unitat de Recerca Biomèdica (URB-CRB), Hospital Universitari de Sant Joan, Institut d’Investigació Santiària Pere i Virgili (IISPV), 43201 Reus, Spain
- Campus of International Excellence Southern Catalonia, 43003 Tarragona, Spain
- Correspondence: (J.C.); (J.J.)
| |
Collapse
|
25
|
Huang KH, Lee CH, Cheng YD, Gau SY, Tsai TH, Chung NJ, Lee CY. Correlation between long-term use of metformin and incidence of NAFLD among patients with type 2 diabetes mellitus: A real-world cohort study. Front Endocrinol (Lausanne) 2022; 13:1027484. [PMID: 36531446 PMCID: PMC9748475 DOI: 10.3389/fendo.2022.1027484] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 11/09/2022] [Indexed: 12/05/2022] Open
Abstract
BACKGROUND AND AIMS Studies have demonstrated that the short-term use of metformin benefits liver function among patients with type 2 diabetes mellitus (T2DM). However, few studies have reported on the effects of long-term metformin treatment on liver function or liver histology. This study investigated the correlation between metformin use and the incidence of nonalcoholic fatty liver disease (NAFLD) among patients with T2DM. METHODS This population-based study investigated the risk of NAFLD among patients with T2DM who received metformin treatment between 2001-2018. Metformin users and metformin nonusers were enrolled and matched to compare the risk of NAFLD. RESULTS After 3 years, the patients who received <300 cDDD of metformin and those with metformin use intensity of <10 and 10-25 DDD/month had odds ratios (ORs) of 1.11 (95% confidence interval [CI] = 1.06-1.16), 1.08 (95% CI = 1.02-1.13), and 1.18 (95% CI = 1.11-1.26) for NAFLD, respectively. Moreover, metformin users who scored high on the Diabetes Complications and Severity Index (DCSI) were at high risk of NAFLD. Patients with comorbid hyperlipidemia, hyperuricemia, obesity, and hepatitis C were also at high risk of NAFLD. CONCLUSION Patients with T2DM who received metformin of <300 cDDD or used metformin at an intensity of <10 and 10-25 DDD/month were at a high risk of developing NAFLD. The results of this study also indicated that patients with T2DM receiving metformin and with high scores on the DCSI were at a high risk of developing NAFLD.
Collapse
Affiliation(s)
- Kuang-Hua Huang
- Department of Health Services Administration, China Medical University, Taichung, Taiwan
| | - Chiu-Hsiang Lee
- School of Nursing, Chung Shan Medical University, Taichung, Taiwan
- Department of Nursing, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Yih-Dih Cheng
- School of Pharmacy, China Medical University, Taichung, Taiwan
- Department of Pharmacy, China Medical University Hospital, Taichung, Taiwan
| | - Shuo-Yan Gau
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Tung-Han Tsai
- Department of Health Services Administration, China Medical University, Taichung, Taiwan
| | - Ning-Jen Chung
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Chien-Ying Lee
- Department of Pharmacology, Chung Shan Medical University, Taichung, Taiwan
- Department of Pharmacy, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
26
|
Guru B, Tamrakar AK, Manjula S, Prashantha Kumar B. Novel dual PPARα/γ agonists protect against liver steatosis and improve insulin sensitivity while avoiding side effects. Eur J Pharmacol 2022; 935:175322. [PMID: 36228743 DOI: 10.1016/j.ejphar.2022.175322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 10/04/2022] [Accepted: 10/05/2022] [Indexed: 11/03/2022]
|
27
|
Abdalla MA, Shah N, Deshmukh H, Sahebkar A, Östlundh L, Al-Rifai RH, Atkin SL, Sathyapalan T. Impact of metformin on the clinical and metabolic parameters of women with polycystic ovary syndrome: a systematic review and meta-analysis of randomised controlled trials. Ther Adv Endocrinol Metab 2022; 13:20420188221127142. [PMID: 36225721 PMCID: PMC9548689 DOI: 10.1177/20420188221127142] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 09/01/2022] [Indexed: 11/17/2022] Open
Abstract
Context Polycystic ovary syndrome (PCOS) is one of the commonest endocrine disorders affecting women of reproductive age, and metformin is a widely used medication in managing this condition. Aim To review the available literature comprehensively on the therapeutic impact of metformin on the clinical and metabolic parameters of women with PCOS. Data source We searched PubMed, MEDLINE, Scopus, Embase, Cochrane Library and the Web of Science and selected sources for grey literature from their inception to April 2020. An updated search in PubMed was performed in June 2022. Data synthesis Two reviewers selected eligible studies and extracted data, and the review is reported following the 2020 Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA). Results In 24 eligible randomised controlled trials (RCTs) involving 564 participants who received metformin therapy, metformin was associated with significant reduction in body weight by 3.13 kg (95% CI: -5.33, -0.93), body mass index (BMI) by 0.82 kg/m² (95% CI: -1.22, -0.41), fasting blood glucose [standardised mean difference (SMD): -0.23; 95% CI: -0.40, -0.06], low-density lipoprotein cholesterol (LDL-C) (SMD: -0.41; 95% CI: -0.85, 0.03), total testosterone (SMD: -0.33; 95% CI: -0.49, -0.17), androstenedione (SMD: -0.45; 95% CI: -0.70, -0.20), 17-hydroxyprogesterone (17-OHP) (SMD: -0.58; 95% CI: -1.16, 0.00) and increase the likelihood of clinical pregnancy rate [odds ratio (OR): 3.00; 95% CI: 1.95, 4.59] compared with placebo. Conclusion In women with PCOS, metformin use has shown a positive impact in reducing body weight, BMI, total testosterone, androstenedione, 17-OHP, LDL-C, fasting blood glucose and increasing the likelihood of pregnancy in women with PCOS.
Collapse
Affiliation(s)
- Mohammed Altigani Abdalla
- Allam Diabetes Centre, Academic Diabetes, Endocrinology and Metabolism, Hull York Medical School (HYMS), University of Hull, Hull, UK
| | - Najeeb Shah
- Allam Diabetes Centre, Academic Diabetes, Endocrinology and Metabolism, Hull York Medical School (HYMS), University of Hull, Hull, UK
| | - Harshal Deshmukh
- Allam Diabetes Centre, Academic Diabetes, Endocrinology and Metabolism, Hull York Medical School (HYMS), University of Hull, Hull, UK
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Medicine, The University of Western Australia, Perth, WA, Australia
| | - Linda Östlundh
- National Medical Library, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Rami H. Al-Rifai
- Institute of Public Health, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Stephen L. Atkin
- School of Postgraduate Studies and Research, RCSI Medical University of Bahrain, Busaiteen, Kingdom of Bahrain
| | - Thozhukat Sathyapalan
- Academic Diabetes, Endocrinology and Metabolism, Allam Diabetes Centre Hull Royal Infirmary Anlaby Road HU3 2JZ, Hull, UK
| |
Collapse
|
28
|
Molecular mechanisms of metabolic associated fatty liver disease (MAFLD): functional analysis of lipid metabolism pathways. Clin Sci (Lond) 2022; 136:1347-1366. [PMID: 36148775 PMCID: PMC9508552 DOI: 10.1042/cs20220572] [Citation(s) in RCA: 162] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/12/2022] [Accepted: 09/14/2022] [Indexed: 01/30/2023]
Abstract
The metabolic-associated fatty liver disease (MAFLD) is a condition of fat accumulation in the liver in combination with metabolic dysfunction in the form of overweight or obesity and insulin resistance. It is also associated with an increased cardiovascular disease risk, including hypertension and atherosclerosis. Hepatic lipid metabolism is regulated by a combination of the uptake and export of fatty acids, de novo lipogenesis, and fat utilization by β-oxidation. When the balance between these pathways is altered, hepatic lipid accumulation commences, and long-term activation of inflammatory and fibrotic pathways can progress to worsen the liver disease. This review discusses the details of the molecular mechanisms regulating hepatic lipids and the emerging therapies targeting these pathways as potential future treatments for MAFLD.
Collapse
|
29
|
Zhu B, Li H, Lu B, Guo X, Wu C, Wang F, Li Q, Xie L, Glaser S, Francis H, Alpini G, Wu C. Indole supplementation ameliorates MCD-induced NASH in mice. J Nutr Biochem 2022; 107:109041. [PMID: 35568098 DOI: 10.1016/j.jnutbio.2022.109041] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 02/27/2022] [Accepted: 03/20/2022] [Indexed: 10/18/2022]
Abstract
Indole is a microbiota metabolite that functions to protect against obesity-associated non-alcoholic fatty liver disease. The present study examined the extent to which indole supplementation alleviates the severity of non-alcoholic steatohepatitis (NASH), which is the advanced form of non-alcoholic fatty liver disease. In C57BL/6J mice, feeding a methionine- and choline-deficient diet (MCD) resulted in significant weight loss, overt hepatic steatosis, and massive aggregations of macrophages in the liver compared with control diet-fed mice. Upon indole supplementation, the severity of MCD-induced hepatic steatosis and inflammation, as well as liver fibrosis, was significantly decreased compared with that of MCD-fed and control-treated mice. In vitro, indole treatment caused significant decreases in lipopolysaccharide-induced proinflammatory responses in hepatocytes incubated with either basal or MCD-mimicking media. However, indole treatment only significantly decreased lipopolysaccharide-induced proinflammatory responses in bone marrow-derived macrophages incubated with basal, but not MCD-mimicking media. These differential effects suggest that, relative to the responses of macrophages to indole, the responses of hepatocytes to indole appeared to make a greater contribution to indole alleviation of NASH, in particular liver inflammation. While indole supplementation decreased liver expression of desmin in MCD-fed mice, treatment of LX2 cells (a line of hepatic stellate cells) with indole also decreased the expression of various markers of hepatic stellate cell fibrogenic activation. Lastly, indole supplementation decreased intestinal inflammation in MCD-fed mice, suggesting that decreased intestinal inflammation also was involved in indole alleviation of NASH. Collectively, these results demonstrate that indole supplementation alleviates MCD-induced NASH, which is attributable to, in large part, indole suppression of hepatocyte proinflammatory responses and hepatic stellate cell fibrogenic activation, as well as intestinal proinflammatory responses.
Collapse
Affiliation(s)
- Bilian Zhu
- Department of Nutrition, Texas A&M University, College Station, Texas, USA
| | - Honggui Li
- Department of Nutrition, Texas A&M University, College Station, Texas, USA
| | - Bangchao Lu
- Department of Nutrition, Texas A&M University, College Station, Texas, USA
| | - Xinlei Guo
- Department of Nutrition, Texas A&M University, College Station, Texas, USA
| | - Chiashan Wu
- Department of Nutrition, Texas A&M University, College Station, Texas, USA
| | - Fen Wang
- Institute of Biosciences and Technology, Texas A&M Health Science Center, Department of Translational Medical Sciences, College of Medicine, Texas A&M University, Houston, Texas, USA
| | - Qingsheng Li
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Linglin Xie
- Department of Nutrition, Texas A&M University, College Station, Texas, USA
| | - Shannon Glaser
- Medical Physiology, Texas A&M University College of Medicine, Bryan, Texas, USA
| | - Heather Francis
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA; Research, Richard L. Roudebush VA Medical Center, Indianapolis, Indiana, USA
| | - Gianfranco Alpini
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA; Research, Richard L. Roudebush VA Medical Center, Indianapolis, Indiana, USA
| | - Chaodong Wu
- Department of Nutrition, Texas A&M University, College Station, Texas, USA.
| |
Collapse
|
30
|
The melatonergic agonist agomelatine ameliorates high fat diet-induced obesity in mice through the modulation of the gut microbiome. Biomed Pharmacother 2022; 153:113445. [PMID: 36076560 DOI: 10.1016/j.biopha.2022.113445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 07/11/2022] [Accepted: 07/18/2022] [Indexed: 11/21/2022] Open
|
31
|
Wu QL, Zeng SX, Peng JY, Yuan Y, Zhu Z, Xie ZC, Huang ZH, Huang JS, Lai JM, Chen JA, Lin MH. Advances in metformin for the treatment of non-alcoholic fatty liver disease in children. Expert Rev Gastroenterol Hepatol 2022; 16:863-877. [PMID: 36039840 DOI: 10.1080/17474124.2022.2118112] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
INTRODUCTION The increased economic and social burdens for NAFLD worldwide make treating such a disease a significant public health issue. Metformin, a kind of insulin sensitizer generally used to treat type 2 diabetes, has been recently found to have efficacy on children's NAFLD in various areas such as glucolipid metabolism, intestinal bacterial metabolism, oxidative stress, and anti-inflammatory response. This article aims to provide an overview of the possible mechanisms of NAFLD in children and the potential therapeutic application of metformin. AREAS COVERED The Cochrane Library, PubMed, Scopus, and EMBASE database was systematically searched on 12 April 2022, using the keywords metformin; non-alcoholic fatty liver disease; and children to identify similar studies. An additional search for recently published research was performed in June 2020. EXPERT OPINION Although metformin has been proved to have an excellent therapeutic effect on children's NAFLD; we can still explore its potential impacts and mechanisms from different angles, such as combined medication. At the same time, we should also pay attention to its side effects.
Collapse
Affiliation(s)
- Qian-Long Wu
- Guangzhou Medical University, Guangzhou, Guangdong province, China
| | - Shu-Xin Zeng
- Guangzhou Medical University, Guangzhou, Guangdong province, China
| | | | | | | | - Zi-Chun Xie
- Guangzhou Medical University, Guangzhou, Guangdong province, China
| | - Ze-Hong Huang
- Guangzhou Medical University, Guangzhou, Guangdong province, China
| | - Jia-Shuan Huang
- Guangzhou Medical University, Guangzhou, Guangdong province, China
| | - Jian-Mei Lai
- Guangzhou Medical University, Guangzhou, Guangdong province, China
| | - Jin-An Chen
- Guangzhou Medical University, Guangzhou, Guangdong province, China
| | - Min-Hua Lin
- Guangzhou Medical University, Guangzhou, Guangdong province, China
| |
Collapse
|
32
|
Bautista-Olivier CD, Elizondo G. PXR as the tipping point between innate immune response, microbial infections, and drug metabolism. Biochem Pharmacol 2022; 202:115147. [PMID: 35714683 DOI: 10.1016/j.bcp.2022.115147] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/08/2022] [Accepted: 06/09/2022] [Indexed: 11/30/2022]
Abstract
Pregnane X receptor (PXR) is a xenosensor that acts as a transcription factor in the cell nucleus to protect cells from toxic insults. In response to exposure to several chemical agents, PXR induces the expression of enzymes and drug transporters that biotransform xenobiotic and endobiotic and eliminate metabolites. Recently, PXR has been shown to have immunomodulatory effects that involve cross-communication with molecular pathways in innate immunity cells. Conversely, several inflammatory factors regulate PXR signaling. This review examines the crosstalk between PXR and nuclear factor kappa B (NFkB), Toll-like receptors (TLRs), and inflammasome components. Discussions of the consequences of these interactions on immune responses to infections caused by viruses, bacteria, fungi, and parasites are included together with a review of the effects of microorganisms on PXR-associated drug metabolism. This paper aims to encourage researchers to pursue studies that will better elucidate the relationship between PXR and the immune system and thus inform treatment development.
Collapse
Affiliation(s)
| | - Guillermo Elizondo
- Departamento de Biología Celular, CINVESTAV-IPN, Av. IPN 2508, C.P. 07360, Ciudad de México, Mexico.
| |
Collapse
|
33
|
Hasanvand A. The role of AMPK-dependent pathways in cellular and molecular mechanisms of metformin: a new perspective for treatment and prevention of diseases. Inflammopharmacology 2022; 30:775-788. [PMID: 35419709 PMCID: PMC9007580 DOI: 10.1007/s10787-022-00980-6] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 03/20/2022] [Indexed: 02/07/2023]
Abstract
Metformin can suppress gluconeogenesis and reduce blood sugar by activating adenosine monophosphate-activated protein kinase (AMPK) and inducing small heterodimer partner (SHP) expression in the liver cells. The main mechanism of metformin's action is related to its activation of the AMPK enzyme and regulation of the energy balance. AMPK is a heterothermic serine/threonine kinase made of a catalytic alpha subunit and two subunits of beta and a gamma regulator. This enzyme can measure the intracellular ratio of AMP/ATP. If this ratio is high, the amino acid threonine 172 available in its alpha chain would be activated by the phosphorylated liver kinase B1 (LKB1), leading to AMPK activation. Several studies have indicated that apart from its significant role in the reduction of blood glucose level, metformin activates the AMPK enzyme that in turn has various efficient impacts on the regulation of various processes, including controlling inflammatory conditions, altering the differentiation pathway of immune and non-immune cell pathways, and the amelioration of various cancers, liver diseases, inflammatory bowel disease (IBD), kidney diseases, neurological disorders, etc. Metformin's activation of AMPK enables it to control inflammatory conditions, improve oxidative status, regulate the differentiation pathways of various cells, change the pathological process in various diseases, and finally have positive therapeutic effects on them. Due to the activation of AMPK and its role in regulating several subcellular signalling pathways, metformin can be effective in altering the cells' proliferation and differentiation pathways and eventually in the prevention and treatment of certain diseases.
Collapse
Affiliation(s)
- Amin Hasanvand
- Department of Physiology and Pharmacology, Faculty of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran.
| |
Collapse
|
34
|
Beheshti F, Hosseini M, Arab Z, Asghari A, Anaeigoudari A. Ameliorative role of metformin on lipopolysaccharide-mediated liver malfunction through suppression of inflammation and oxidative stress in rats. TOXIN REV 2022; 41:55-63. [DOI: 10.1080/15569543.2020.1833037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 10/02/2020] [Indexed: 10/23/2022]
Affiliation(s)
- Farimah Beheshti
- Neuroscience Research Center, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
- Department of Physiology, School of Paramedical Sciences, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Mahmoud Hosseini
- Division of Neurocognitive Sciences, Psychiatry and Behavioral Sciences Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zohreh Arab
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Asghari
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Akbar Anaeigoudari
- Department of Physiology, School of Medicine, Jiroft University of Medical Sciences, Jiroft, Iran
| |
Collapse
|
35
|
18:0 Lyso PC Derived by Bioactivity-Based Molecular Networking from Lentil Mutant Lines and Its Effects on High-Fat Diet-Induced Obese Mice. Molecules 2021; 26:molecules26247547. [PMID: 34946633 PMCID: PMC8707596 DOI: 10.3390/molecules26247547] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 01/12/2023] Open
Abstract
Lentil (Lens culinaris; Fabaceae), one of the major pulse crops in the world, is an important source of proteins, prebiotics, lipids, and essential minerals as well as functional components such as flavonoids, polyphenols, and phenolic acids. To improve crop nutritional and medicinal traits, hybridization and mutation are widely used in plant breeding research. In this study, mutant lentil populations were generated by γ-irradiation for the development of new cultivars by inducing genetic diversity. Molecular networking via Global Natural Product Social Molecular Networking web platform and dipeptidyl peptide-IV inhibitor screening assay were utilized as tools for structure-based discovery of active components in active mutant lines selected among the lentil population. The bioactivity-based molecular networking analysis resulted in the annotation of the molecular class of phosphatidylcholine (PC) from the most active mutant line. Among PCs, 1-stearoyl-2-hydroxy-sn-glycero-3-phosphocholine (18:0 Lyso PC) was selected for further in vivo study of anti-obesity effect in a high-fat diet (HFD)-induced obese mouse model. The administration of 18:0 Lyso PC not only prevented body weight gain and decreased relative gonadal adipose tissue weight, but also attenuated the levels of total cholesterol, triglycerides, low-density lipoprotein cholesterol, and leptin in the sera of HFD-induced obese mice. Additionally, 18:0 Lyso PC treatment inhibited the increase of adipocyte area and crown-like structures in adipose tissue. Therefore, these results suggest that 18:0 Lyso PC is a potential compound to have protective effects against obesity, improving obese phenotype induced by HFD.
Collapse
|
36
|
Li Y, Yang L, Wang Y, Deng Z, Xu S, Xie H, Zhang Y, Li J. Exploring metformin as a candidate drug for rosacea through network pharmacology and experimental validation. Pharmacol Res 2021; 174:105971. [PMID: 34763093 DOI: 10.1016/j.phrs.2021.105971] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/28/2021] [Accepted: 11/02/2021] [Indexed: 12/12/2022]
Abstract
Rosacea is a common chronic inflammatory disease that affects the middle of the face. Due to the unclear pathogenesis, the effective treatment options for rosacea remain limited. In this study, weighted gene co-expression network analyses (WGCNA) identified three rosacea-related hub modules, which were involved in immune-, metabolic- and development- related signaling pathways. Next, the key genes from green and brown modules were submitted to CMap database for drug prediction and metformin was identified as a candidate drug for rosacea. Moreover, network pharmacology analysis identified pharmacological targets of metformin and demonstrated that metformin could help in treating rosacea partly by modulating inflammatory and angiogenesis signaling pathways. Finally, we verified the therapeutic role and mechanism of metformin on rosacea in vivo and vitro. We found that metformin treatment significantly improved rosacea-like skin lesions including immune cells infiltration, cytokines/chemokines expression and angiogenesis. Moreover, metformin suppressed LL37- and TNF-α-induced the ROS production and MAPK-NF-κB signal activation in keratinocytes cells. In conclusion, our findings identified and verified metformin as a novel therapeutic candidate for rosacea, and it alleviates the pathological symptoms, possibly by suppressing inflammatory responses, angiogenesis in rosacea.
Collapse
Affiliation(s)
- Yangfan Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan key laboratory of aging biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders,Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Li Yang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan key laboratory of aging biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders,Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Yaling Wang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan key laboratory of aging biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders,Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Zhili Deng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan key laboratory of aging biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders,Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - San Xu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan key laboratory of aging biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders,Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Hongfu Xie
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan key laboratory of aging biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders,Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Yiya Zhang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan key laboratory of aging biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders,Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China.
| | - Ji Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan key laboratory of aging biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders,Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China; Department of Dermatology, The Second Affiliated Hospital of Xinjiang Medical University, Urumqi, China.
| |
Collapse
|
37
|
Metformin treatment reverses high fat diet- induced non-alcoholic fatty liver diseases and dyslipidemia by stimulating multiple antioxidant and anti-inflammatory pathways. Biochem Biophys Rep 2021; 28:101168. [PMID: 34825068 PMCID: PMC8605070 DOI: 10.1016/j.bbrep.2021.101168] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 10/18/2021] [Accepted: 11/01/2021] [Indexed: 01/06/2023] Open
Abstract
Purpose This current study investigated the effect of metformin treatment on hepatic oxidative stress and inflammation associated with nonalcoholic fatty liver disease (NADLD) in high fat diet (HFD) fed rats. Method Wistar rats were fed with a HFD or laboratory chow diet for 8 weeks. Metformin was administered orally at a dose of 200 mg/kg. Body weight, food and water intake were recorded on daily basis. Oral glucose tolerance test (OGTT), biochemical analysis and histological examinations were conducted on plasma and tissue samples. Antioxidant and anti-inflammatory mRNA expression was analyzed using reverse transcription polymeric chain reaction (RT-PCR). Results Metformin treatment for 8 weeks prevented HFD-induced weight gain and decreased fat deposition in HFD fed rats. Biochemical analysis revealed that metformin treatment significantly attenuated nitro-oxidative stress markers malondialdehyde (MDA), advanced protein oxidation product (APOP), and excessive nitric oxide (NO) levels in the liver of HFD fed rats. Gene expression analysis demonestrated that metformin treatment was associated with an enhanced expression of antioxidant genes such as Nrf-2, HO-1, SOD and catalase in liver of HFD fed rats. Metformin treatment also found to modulate the expression of fat metabolizing and anti-inflammatory genes including PPAR--γ, C/EBP-α, SREBP1c, FAS, AMPK and GLUT-4. Consistent with the biochemical and gene expression data, the histopathological examination unveiled that metformin treatment attenuated inflammatory cells infiltration, steatosis, hepatocyte necrosis, collagen deposition, and fibrosis in the liver of HFD fed rats. Conclusion In conclusion, this study suggests that metformin might be effective in the prevention and treatment of HFD-induced steatosis by reducing hepatic oxidative stress and inflammation in the liver.
Collapse
Key Words
- ALP, alkaline phosphatase
- ALT, alanine aminotransferase
- AMPK, AMP-activated protein kinase
- APOP, advanced protein oxidation product
- AST, aspartate aminotransferase
- ATP, Adinosine triphosphate
- AUC, area under the curve
- CAT, catalase
- FAS, Fatty acid synthase
- HDL, high density lipoprotein
- HF, High fat
- HSCs, Hepatic stellate cells
- IACUC, Institutional Animal Care and Use Committee
- IL-6, interleukin-6
- Inflammation
- LDL, low density lipoprotein
- Lipid peroxidation
- MDA, Malondialdehyde
- MPO, Myeloperoxidase
- Met, Metformin
- Metformin
- NAFLD, nonalcoholic fatty liver disease
- NO, nitric oxide
- Non-alcoholic fatty liver disease
- OGTT, Oral glucose tolerance test
- Obesity
- PBS, Phosphate buffer saline
- PGC-1α, peroxisome proliferator-activated receptor γ coactivator 1
- PPAR-γ, peroxisome proliferator-activated receptor γ
- ROS, reactive oxygen species
- SOD, Superoxide dismutase
- SREBP1c, sterol regulatory element-binding protein 1c
- TBA, Thiobarbituric acid
- TBARS, Thiobarbituric acid reactive substances
Collapse
|
38
|
N-butylidenephthalide ameliorates high-fat diet-induced obesity in mice and promotes browning through adrenergic response/AMPK activation in mouse beige adipocytes. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:159033. [PMID: 34487913 DOI: 10.1016/j.bbalip.2021.159033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 08/04/2021] [Accepted: 08/17/2021] [Indexed: 11/20/2022]
Abstract
Thermogenesis (non-exercise activity) in brown adipose tissue (BAT) promotes energy expenditure because of its higher number of mitochondria than white adipose tissue (WAT). The main function of thermogenesis in BAT can counteract obesity through the dissipation of calories as heat. N-butylidenephthalide (BP) is a natural derivative from Angelica sinensis, a Chinese herb that has been used for thousands of years. In this report, we demonstrated that BP improved the metabolic profiles of mice with high fat diet-induced obesity (DIO) by preventing weight gain, improving serum blood parameters, enhancing energy expenditure, stimulating white fat browning, and reversing hepatic steatosis. Further investigations demonstrated that BP administration upregulated the mRNA expression of beige (CD137, TMEM26) and brown fat selected genes (UCP1, PRDM16, PGC-1α, PPARγ) in white adipose tissues. In vitro studies, BP treatment increased multilocular lipid droplet levels, induced β-adrenergic receptor (cAMP/PKA) and AMP-activated protein kinase (AMPK) signaling (AMPK/acetyl-CoA carboxylase/SIRT1), and increased oxygen consumption in murine differentiated beige adipocytes, and the effects of BP were blocked by an AMPK inhibitor. BP promoted the interaction of AMPK with PGC-1α in beige adipocytes. Our findings provide novel insights into the application of BP in regulating energy metabolism and suggest its utility for clinical use in the treatment of obesity and related diseases.
Collapse
|
39
|
Metformin Affects Cardiac Arachidonic Acid Metabolism and Cardiac Lipid Metabolite Storage in a Prediabetic Rat Model. Int J Mol Sci 2021; 22:ijms22147680. [PMID: 34299301 PMCID: PMC8305829 DOI: 10.3390/ijms22147680] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 06/30/2021] [Accepted: 07/12/2021] [Indexed: 02/07/2023] Open
Abstract
Metformin can reduce cardiovascular risk independent of glycemic control. The mechanisms behind its non-glycemic benefits, which include decreased energy intake, lower blood pressure and improved lipid and fatty acid metabolism, are not fully understood. In our study, metformin treatment reduced myocardial accumulation of neutral lipids—triglycerides, cholesteryl esters and the lipotoxic intermediates—diacylglycerols and lysophosphatidylcholines in a prediabetic rat model (p < 0.001). We observed an association between decreased gene expression and SCD-1 activity (p < 0.05). In addition, metformin markedly improved phospholipid fatty acid composition in the myocardium, represented by decreased SFA profiles and increased n3-PUFA profiles. Known for its cardioprotective and anti-inflammatory properties, metformin also had positive effects on arachidonic acid metabolism and CYP-derived arachidonic acid metabolites. We also found an association between increased gene expression of the cardiac isoform CYP2c with increased 14,15-EET (p < 0.05) and markedly reduced 20-HETE (p < 0.001) in the myocardium. Based on these results, we conclude that metformin treatment reduces the lipogenic enzyme SCD-1 and the accumulation of the lipotoxic intermediates diacylglycerols and lysophosphatidylcholine. Increased CYP2c gene expression and beneficial effects on CYP-derived arachidonic acid metabolites in the myocardium can also be involved in cardioprotective effect of metformin.
Collapse
|
40
|
Matthews DR, Li H, Zhou J, Li Q, Glaser S, Francis H, Alpini G, Wu C. Methionine- and Choline-Deficient Diet-Induced Nonalcoholic Steatohepatitis Is Associated with Increased Intestinal Inflammation. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:1743-1753. [PMID: 34242656 DOI: 10.1016/j.ajpath.2021.06.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 06/14/2021] [Accepted: 06/24/2021] [Indexed: 12/16/2022]
Abstract
Inflammation drives the development and progression of nonalcoholic steatohepatitis (NASH). The current study examined changes in intestinal inflammation during NASH. In male C57BL/6J mice, feeding a methionine- and choline-deficient diet (MCD) resulted in severe hepatic steatosis and inflammation relative to feeding a chow diet (CD). Also, MCD-fed mice exhibited characteristics of mucosal and submucosal inflammatory responses and increased CD68+ cells compared with mice fed a CD. Moreover, intestinal phosphorylation states of c-Jun N-terminal protein kinase p46 and mRNA levels of IL-1B, IL-6, tumor necrosis factor alpha, and monocyte chemoattractant protein-1 were significantly higher and intestinal mRNA levels of IL-4 and IL-13 significantly lower in MCD-fed mice compared with their respective levels in CD mice. Surprisingly, upon treatment with MCD-mimicking media, the proinflammatory responses in cultured intestinal epithelial cells (CMT-93 cells, a transformed epithelial cell line) did not differ significantly from those in intestinal epithelial cells treated with control media. In contrast, in RAW264.7 cells (transformed macrophages), MCD-mimicking media significantly increased the phosphorylation states of c-Jun N-terminal protein kinase p46 and mitogen-activated protein kinases p38 and mRNA levels of IL-1B, IL-6, IL-10, and tumor necrosis factor alpha under either basal or lipopolysaccharide-stimulated conditions. Collectively, these results suggest that increased intestinal inflammation is associated with NASH phenotype. In addition, elevated proinflammatory responses in macrophages likely contribute to, in large part, increased intestinal inflammation in NASH.
Collapse
Affiliation(s)
| | - Honggui Li
- Department of Nutrition, Texas A&M University, College Station, Texas
| | - Jing Zhou
- Department of Nutrition, Texas A&M University, College Station, Texas
| | - Qingsheng Li
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska
| | - Shannon Glaser
- Medical Physiology, Texas A&M University College of Medicine, Bryan, Texas
| | - Heather Francis
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana; Research, Richard L. Roudebush VA Medical Center, Indianapolis, Indiana
| | - Gianfranco Alpini
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana; Research, Richard L. Roudebush VA Medical Center, Indianapolis, Indiana
| | - Chaodong Wu
- Department of Nutrition, Texas A&M University, College Station, Texas.
| |
Collapse
|
41
|
Feng X, Chen W, Ni X, Little PJ, Xu S, Tang L, Weng J. Metformin, Macrophage Dysfunction and Atherosclerosis. Front Immunol 2021; 12:682853. [PMID: 34163481 PMCID: PMC8215340 DOI: 10.3389/fimmu.2021.682853] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 05/07/2021] [Indexed: 12/17/2022] Open
Abstract
Metformin is one of the most widely prescribed hypoglycemic drugs and has the potential to treat many diseases. More and more evidence shows that metformin can regulate the function of macrophages in atherosclerosis, including reducing the differentiation of monocytes and inhibiting the inflammation, oxidative stress, polarization, foam cell formation and apoptosis of macrophages. The mechanisms by which metformin regulates the function of macrophages include AMPK, AMPK independent targets, NF-κB, ABCG5/8, Sirt1, FOXO1/FABP4 and HMGB1. On the basis of summarizing these studies, we further discussed the future research directions of metformin: single-cell RNA sequencing, neutrophil extracellular traps (NETs), epigenetic modification, and metformin-based combination drugs. In short, macrophages play an important role in a variety of diseases, and improving macrophage dysfunction may be an important mechanism for metformin to expand its pleiotropic pharmacological profile. In addition, the combination of metformin with other drugs that improve the function of macrophages (such as SGLT2 inhibitors, statins and IL-1β inhibitors/monoclonal antibodies) may further enhance the pleiotropic therapeutic potential of metformin in conditions such as atherosclerosis, obesity, cancer, dementia and aging.
Collapse
Affiliation(s)
- Xiaojun Feng
- Department of Pharmacy, the First Affiliated Hospital of University of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, China
| | - Wenxu Chen
- Department of Pharmacy, the First Affiliated Hospital of University of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, China
| | - Xiayun Ni
- Department of Pharmacy, the First Affiliated Hospital of University of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, China
| | - Peter J. Little
- Sunshine Coast Health Institute, University of the Sunshine Coast, Birtinya, QLD, Australia
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, QLD, Australia
| | - Suowen Xu
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China( USTC), Hefei, China
| | - Liqin Tang
- Department of Pharmacy, the First Affiliated Hospital of University of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, China
| | - Jianping Weng
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China( USTC), Hefei, China
| |
Collapse
|
42
|
Sato Y, Qiu J, Hirose T, Miura T, Sato Y, Kohzuki M, Ito O. Metformin slows liver cyst formation and fibrosis in experimental model of polycystic liver disease. Am J Physiol Gastrointest Liver Physiol 2021; 320:G464-G473. [PMID: 33439105 DOI: 10.1152/ajpgi.00120.2020] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Polycystic liver disease (PLD) is a hereditary liver disease in which the number of cysts increases over time, causing various abdominal symptoms and poor quality of life. Although effective treatment for PLD has not been established, we recently reported that long-term exercise ameliorated liver cyst formation and fibrosis with the activation of AMP-activated protein kinase (AMPK) in polycystic kidney (PCK) rats, a PLD model. Therefore, the aim of this study was to investigate whether metformin, an indirect AMPK activator, was effective in PCK rats. PCK rats were randomly divided into a control (Con) group and a metformin-treated (Met) group. The Met group was treated orally with metformin in drinking water. After 12 wk, liver function, histology, and signaling cascades of PLD were examined in the groups. Metformin did not affect the body weight or liver weight, but it reduced liver cyst formation, cholangiocyte proliferation, and fibrosis around the cyst. Metformin increased the phosphorylation of AMPK and tuberous sclerosis complex 2 and decreased the phosphorylation of mammalian target of rapamycin, S6, and extracellular signal-regulated kinase and the expression of cystic fibrosis transmembrane conductance regulator, aquaporin I, transforming growth factor-β, and type 1 collagen without changes in apoptosis or collagen degradation factors in the liver. Metformin slows the development of cyst formation and fibrosis with the activation of AMPK and inhibition of signaling cascades responsible for cellular proliferation and fibrosis in the liver of PCK rats.NEW & NOTEWORTHY This study indicates that metformin, an indirect AMPK activator slows liver cyst formation and fibrosis in PLD rat model. Metformin attenuates excessive cell proliferation in the liver with the inactivation of mTOR and ERK pathways. Metformin also reduces the expression of proteins responsible for cystic fluid secretion and liver fibrosis. Metformin and AMPK activators may be potent drugs for polycystic liver disease.
Collapse
Affiliation(s)
- Yoichi Sato
- Department of Internal Medicine and Rehabilitation Science, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Jiahe Qiu
- Department of Internal Medicine and Rehabilitation Science, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takuo Hirose
- Division of Endocrinology and Applied Medical Science, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takahiro Miura
- Department of Internal Medicine and Rehabilitation Science, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yasunori Sato
- Department of Human Pathology, Kanazawa University Graduate School of Medicine, Kanazawa, Japan
| | - Masahiro Kohzuki
- Department of Internal Medicine and Rehabilitation Science, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Osamu Ito
- Department of Internal Medicine and Rehabilitation Science, Tohoku University Graduate School of Medicine, Sendai, Japan.,Division of General Medicine and Rehabilitation, Tohoku Medical and Pharmaceutical University Faculty of Medicine, Sendai, Japan
| |
Collapse
|
43
|
Seif El-Din SH, Salem MB, El-Lakkany NM, Hammam OA, Nasr SM, Okasha H, Ahmed LA, Saleh S, Botros SS. Early intervention with probiotics and metformin alleviates liver injury in NAFLD rats via targeting gut microbiota dysbiosis and p-AKT/mTOR/LC-3II pathways. Hum Exp Toxicol 2021; 40:1496-1509. [PMID: 33678036 DOI: 10.1177/0960327121999445] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) constitutes a major health problem worldwide and intimately links with obesity and diabetes. This study aimed to explore the therapeutic impact of early treatment with metformin (MTF) alone or in combination with Lactobacillus reuteri DSM 17938 (L. reuteri) + metronidazole (MTZ) in male Sprague Dawley rats with high-fat diet (HFD)-induced NAFLD. Hepatic steatosis was induced by feeding rats HFD for 6 weeks. MTF (150 mg/kg/day) or L. reuteri (2 × 109 colony forming unit/day) were given orally for 4 weeks; meanwhile, MTZ (15 mg/kg/day, p.o.) was administered for 1 week. Administration of L. reuteri + MTZ in combination with MTF produced a superior effect concerning insulin resistance (IR), lipid profile, liver function, oxidative stress, inflammatory and autophagic markers than using each treatment alone. Besides, this combination resulted in disappearance of steatosis, inflammation and vacuolation within hepatic architecture. Moreover, it normalized short chain fatty acids (SCFAs) as well as Firmicutes and Bacteroidetes faecal contents. In conclusion, early treatment with L. reuteri + MTZ in combination with MTF could prevent NAFLD progression and liver injury through targeting gut dysbiosis, inflammation and autophagic pathways.
Collapse
Affiliation(s)
- Sayed H Seif El-Din
- Pharmacology Department, 230796Theodor Bilharz Research Institute, Warak El-Hadar, Imbaba, Giza, Egypt
| | - M B Salem
- Pharmacology Department, 230796Theodor Bilharz Research Institute, Warak El-Hadar, Imbaba, Giza, Egypt
| | - N M El-Lakkany
- Pharmacology Department, 230796Theodor Bilharz Research Institute, Warak El-Hadar, Imbaba, Giza, Egypt
| | - O A Hammam
- Pathology Department, 230796Theodor Bilharz Research Institute, Warak El-Hadar, Imbaba, Giza, Egypt
| | - S M Nasr
- Biochemistry Department, 230796Theodor Bilharz Research Institute, Warak El-Hadar, Imbaba, Giza, Egypt
| | - H Okasha
- Biochemistry Department, 230796Theodor Bilharz Research Institute, Warak El-Hadar, Imbaba, Giza, Egypt
| | - L A Ahmed
- Pharmacology and Toxicology Department, 110154Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - S Saleh
- Pharmacology and Toxicology Department, 110154Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - S S Botros
- Pharmacology Department, 230796Theodor Bilharz Research Institute, Warak El-Hadar, Imbaba, Giza, Egypt
| |
Collapse
|
44
|
Zhu B, Guo X, Xu H, Jiang B, Li H, Wang Y, Yin Q, Zhou T, Cai JJ, Glaser S, Meng F, Francis H, Alpini G, Wu C. Adipose tissue inflammation and systemic insulin resistance in mice with diet-induced obesity is possibly associated with disruption of PFKFB3 in hematopoietic cells. J Transl Med 2021; 101:328-340. [PMID: 33462362 PMCID: PMC7897240 DOI: 10.1038/s41374-020-00523-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 10/13/2020] [Accepted: 11/28/2020] [Indexed: 02/06/2023] Open
Abstract
Obesity-associated inflammation in white adipose tissue (WAT) is a causal factor of systemic insulin resistance; however, precisely how immune cells regulate WAT inflammation in relation to systemic insulin resistance remains to be elucidated. The present study examined a role for 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3) in hematopoietic cells in regulating WAT inflammation and systemic insulin sensitivity. Male C57BL/6J mice were fed a high-fat diet (HFD) or low-fat diet (LFD) for 12 weeks and examined for WAT inducible 6-phosphofructo-2-kinase (iPFK2) content, while additional HFD-fed mice were treated with rosiglitazone and examined for PFKFB3 mRNAs in WAT stromal vascular cells (SVC). Also, chimeric mice in which PFKFB3 was disrupted only in hematopoietic cells and control chimeric mice were also fed an HFD and examined for HFD-induced WAT inflammation and systemic insulin resistance. In vitro, adipocytes were co-cultured with bone marrow-derived macrophages and examined for adipocyte proinflammatory responses and insulin signaling. Compared with their respective levels in controls, WAT iPFK2 amount in HFD-fed mice and WAT SVC PFKFB3 mRNAs in rosiglitazone-treated mice were significantly increased. When the inflammatory responses were analyzed, peritoneal macrophages from PFKFB3-disrputed mice revealed increased proinflammatory activation and decreased anti-inflammatory activation compared with control macrophages. At the whole animal level, hematopoietic cell-specific PFKFB3 disruption enhanced the effects of HFD feeding on promoting WAT inflammation, impairing WAT insulin signaling, and increasing systemic insulin resistance. In vitro, adipocytes co-cultured with PFKFB3-disrupted macrophages revealed increased proinflammatory responses and decreased insulin signaling compared with adipocytes co-cultured with control macrophages. These results suggest that PFKFB3 disruption in hematopoietic cells only exacerbates HFD-induced WAT inflammation and systemic insulin resistance.
Collapse
Affiliation(s)
- Bilian Zhu
- Department of Nutrition, Texas A&M University, College Station, TX, USA
- Department of VIP Medical Service Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xin Guo
- Department of Nutrition, Texas A&M University, College Station, TX, USA
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Shandong, Jinan, China
| | - Hang Xu
- Department of Nutrition, Texas A&M University, College Station, TX, USA
| | - Boxiong Jiang
- Department of VIP Medical Service Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Honggui Li
- Department of Nutrition, Texas A&M University, College Station, TX, USA
| | - Yina Wang
- Department of VIP Medical Service Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Qiongli Yin
- Department of VIP Medical Service Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Tianhao Zhou
- Medical Physiology, Texas A&M University College of Medicine, Bryan, TX, USA
| | - James J Cai
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA
| | - Shannon Glaser
- Medical Physiology, Texas A&M University College of Medicine, Bryan, TX, USA
| | - Fanyin Meng
- Hepatology and Gastroenterology, Medicine, Indiana University, Indianapolis, IN, USA
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, USA
| | - Heather Francis
- Hepatology and Gastroenterology, Medicine, Indiana University, Indianapolis, IN, USA
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, USA
| | - Gianfranco Alpini
- Hepatology and Gastroenterology, Medicine, Indiana University, Indianapolis, IN, USA
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, USA
| | - Chaodong Wu
- Department of Nutrition, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
45
|
AbdelAziz EY, Tadros MG, Menze ET. The effect of metformin on indomethacin-induced gastric ulcer: Involvement of nitric oxide/Rho kinase pathway. Eur J Pharmacol 2021; 892:173812. [PMID: 33345855 DOI: 10.1016/j.ejphar.2020.173812] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 12/11/2020] [Accepted: 12/11/2020] [Indexed: 02/06/2023]
Abstract
Gastric ulcer is a very common disease that represent an economic burden. Non-steroidal anti-inflammatory drugs induce ulcer in old patients and in patients with comorbidities. Indomethacin is widely used to induce gastric ulcer in animal models. Diabetic patients are highly susceptible to develop gastric ulcer. Metformin, the first line medication for the treatment of type II diabetes melilites that have many off label uses in non-diabetic patients, has been recently reported to have anti-inflammatory activities. Therefore, this research was conducted to assess the possible healing effects of metformin on gastric ulcers induced by indomethacin in rats. Indomethacin (48 mg/kg) single dose increased stomach acidity, ulcer index and induced histopathological changes. Indomethacin also decreased mucin levels and increased the activity of tumor necrosis factor-α (TNF-α), nuclear factor kappa-B (NF-κB), Rho-associated protein kinas-1 (ROCK-1) and decreased the levels of the protective nitric oxide (NO). After the induction of ulcer, rats were treated by omeprazole (30 mg/kg) or metformin (50, 100 or 200 mg/kg). Omeprazole and metformin were found to decrease stomach acidity and ulcer index, restored the histological features and increased mucin levels. Both also decreased the levels of NF-κB, TNF-α, ROCK-1 and increased NO. Metformin exerted ulcer healing effects comparable to that of omeprazole. This can be attributed, at least partly, to its anti-inflammatory activity and increasing NO levels.
Collapse
Affiliation(s)
- Eman Y AbdelAziz
- Pharmacology and Toxicology Department, Faculty of Pharmacy Ain Shams University, Cairo, Egypt
| | - Mariane G Tadros
- Pharmacology and Toxicology Department, Faculty of Pharmacy Ain Shams University, Cairo, Egypt
| | - Esther T Menze
- Pharmacology and Toxicology Department, Faculty of Pharmacy Ain Shams University, Cairo, Egypt.
| |
Collapse
|
46
|
Acín S, Muñoz DL, Guillen A, Soscue D, Castaño A, Echeverri F, Balcazar N. Triterpene-enriched fractions from Eucalyptus tereticornis ameliorate metabolic alterations in a mouse model of diet-induced obesity. JOURNAL OF ETHNOPHARMACOLOGY 2021; 265:113298. [PMID: 32860892 DOI: 10.1016/j.jep.2020.113298] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 08/11/2020] [Accepted: 08/16/2020] [Indexed: 06/11/2023]
Abstract
ETNOPHARMACOLOGICAL RELEVANCE Eucalyptus tereticornis Sm. (Eu) is a plant species used in traditional medicine to treat diabetes mellitus. Eu leaf extracts have been shown to regulate immuno-metabolic activities that are associated with obesity and insulin resistance. OBE100 and OBE104 are two natural Eu extracts that are rich in pentacyclic triterpenes. The major compounds identified in OBE100 are ursolic acid (UA), oleanolic acid (OA), and ursolic acid lactone (UAL), and the major compounds identified in OBE104 are UA and OA. AIM OF THE STUDY This study aimed to investigate the effects of two extracts from Eu leaves with different triterpene composition in a nutritional animal model of prediabetes. METHODS A mouse model of diet-induced obesity was used to analyze the effects of the OBE100 and OBE104 treatments on metabolic markers and gene expression in liver and visceral adipose tissue. RESULTS Treating the prediabetic mouse model with OBE100 and OBE104 increased glucose tolerance. However, only the Eu extract that contained three triterpenes reduced mouse body weight, hepatic and adipose fat content, and plasma lipid levels. OBE100 treatment also led to decreased hepatic mRNA levels of PPARA, CPT1A, and SERBP1. In visceral adipose tissue, OBE100 treatment reduced expression of PPARA and ACACA and increased UCP1 expression. CONCLUSIONS These results suggest that developing a new multitargeting bioactive compound from the natural extract from Eu may help combat obesity and diabetes. Treatment with OBE100 had better effects than OBE104 in a diet-induced obesity mouse model, suggesting that the OBE100 extract, which contains three triterpenes, may be beneficial in combating obesity.
Collapse
Affiliation(s)
- Sergio Acín
- Molecular Genetics Group, Universidad de Antioquia, Calle 70, N° 52-21, A.A, 1226, Medellin, Colombia; Department of Physiology and Biochemistry, School of Medicine, Universidad de Antioquia, Calle 70, N° 52-21, A.A, 1226, Medellin, Colombia
| | - Diana Lorena Muñoz
- Department of Physiology and Biochemistry, School of Medicine, Universidad de Antioquia, Calle 70, N° 52-21, A.A, 1226, Medellin, Colombia
| | - Alis Guillen
- Molecular Genetics Group, Universidad de Antioquia, Calle 70, N° 52-21, A.A, 1226, Medellin, Colombia
| | - Duberney Soscue
- Molecular Genetics Group, Universidad de Antioquia, Calle 70, N° 52-21, A.A, 1226, Medellin, Colombia
| | - Adriana Castaño
- Group of Organic Natural Product Chemistry, Faculty of Natural and Exact Sciences, Universidad de Antioquia, Calle 70, N° 52-21, A.A, 1226, Medellin, Colombia
| | - Fernando Echeverri
- Group of Organic Natural Product Chemistry, Faculty of Natural and Exact Sciences, Universidad de Antioquia, Calle 70, N° 52-21, A.A, 1226, Medellin, Colombia
| | - Norman Balcazar
- Molecular Genetics Group, Universidad de Antioquia, Calle 70, N° 52-21, A.A, 1226, Medellin, Colombia; Department of Physiology and Biochemistry, School of Medicine, Universidad de Antioquia, Calle 70, N° 52-21, A.A, 1226, Medellin, Colombia.
| |
Collapse
|
47
|
Amoani B, Sakyi SA, Mantey R, Laing EF, Ephraim RD, Sarfo-Katanka O, Koffie S, Obese E, Afranie BO. Increased metformin dosage suppresses pro-inflammatory cytokine levels in systemic circulation and might contribute to its beneficial effects. J Immunoassay Immunochem 2021; 42:252-264. [PMID: 33444083 DOI: 10.1080/15321819.2020.1862861] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Type 2 Diabetes Mellitus (T2DM) is a metabolic disorder, characterized by persistent elevation of blood glucose either due to insulin resistance or insulin insufficiency. Metformin is the recommended first choice of drug for the management of T2DM and is known to improve insulin sensitivity and prevents hyperglycemia by reducing chronic inflammation. T-helper type 1 (Th1) and type 17 (Th17) cells, are important pro-inflammatory CD4+ T cell subsets secreting TNF-α, and INF-γ (Th1), and interleukin 17 (Th17). These cytokines have been shown to play a crucial role in inflammation, insulin resistance, and the development of T2DM. Here, we explore the effect of different metformin dosages on pro-inflammatory cytokine (TNF-α, INF-γ, GM-CSF and IL-17) levels in systemic circulation among T2DM patients in Ghana, since inflammatory responses and cytokines play significant roles in the pathogenesis and progression of T2DM patients on metformin. Two hundred and nine (209) consenting T2DM patients receiving treatment at the Diabetic unit of the Komfo Anokye Teaching Hospital (KATH) in the Ashanti region of Ghana were recruited in a hospital-based cross-sectional study design. Blood samples were collected and serum obtained from each participant were analyzed for the concentrations of TNF-α, INF-γ, GM-CSF and IL-17 cytokine levels by solid-phase sandwich ELISA. We observed that participants on 3000 mg/day dose of metformin had significantly lower levels of TNF-α (p < .001) and IFN-γ (p = .014) compared to those on other dosages (1000 mg and 2000 mg/day). However, GM-CSF and IL-17 levels were not affected by increased metformin dosages. After adjusting for age, gender, dose and duration of metformin use, we observed that participants who took higher doses of metformin had significantly reduced levels of TNF-α (β = -0.0297, 95% CI = (-0.005 to -0.002) p < .001. Metformin dosage independently predicted reduced TNF-α levels with 14.4% variations in the metformin dosage levels. Increased metformin dosage suppresses TNF-α levels in systemic circulation and hence might contribute to its beneficial effects.
Collapse
Affiliation(s)
- Benjamin Amoani
- Department of Biomedical Sciences, College of Health and Allied Sciences, School of Allied Health Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Samuel A Sakyi
- Department of Molecular Medicine. School of Medical Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Richard Mantey
- Department of Molecular Medicine. School of Medical Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Edwin F Laing
- Department of Molecular Medicine. School of Medical Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Richard D Ephraim
- Department of Medical Laboratory Sciences, School of Allied Health Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Osei Sarfo-Katanka
- Department of Medicine, Diabetes Clinic, School of Medical Sciences, Kwame Nkrumah University of Science and Technology, Komfo Anokye Teaching Hospital (KATH), Kumasi, Ghana
| | - Simon Koffie
- Department of Molecular Medicine. School of Medical Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Ernest Obese
- Department of Pharmacology, School of Medical Sciences, College of Health and Allied Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Bright Oppong Afranie
- Department of Molecular Medicine. School of Medical Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| |
Collapse
|
48
|
Zamani-Garmsiri F, Hashemnia SMR, Shabani M, Bagherieh M, Emamgholipour S, Meshkani R. Combination of metformin and genistein alleviates non-alcoholic fatty liver disease in high-fat diet-fed mice. J Nutr Biochem 2021; 87:108505. [DOI: 10.1016/j.jnutbio.2020.108505] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 07/16/2020] [Accepted: 08/26/2020] [Indexed: 12/12/2022]
|
49
|
Zamani-Garmsiri F, Ghasempour G, Aliabadi M, Hashemnia SMR, Emamgholipour S, Meshkani R. Combination of metformin and chlorogenic acid attenuates hepatic steatosis and inflammation in high-fat diet fed mice. IUBMB Life 2020; 73:252-263. [PMID: 33326684 DOI: 10.1002/iub.2424] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/24/2020] [Accepted: 11/24/2020] [Indexed: 12/15/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) has become an important health problem in the world. Natural products, with anti-inflammatory properties, are potential candidates for alleviating NAFLD. Metformin (MET) and chlorogenic acid (CGA) have been reported to be effective in the improvement of NAFLD. Here, we aimed to evaluate the efficacy of MET and CGA combination in ameliorating NAFLD in high-fat diet (HFD) fed mice. Fifty C57BL/6 male mice were divided into two groups, one fed a standard chow diet (n = 10) and the other was fed an HFD (n = 40) for 10 weeks. Animals in the HFD group were then randomly divided into a four groups (HFD, HFD + MET (0.25%), HFD + CGA (0.02%) and HFD + MET + CGA (0.25 + 0.02%). MET and CGA combination decreases fasting blood glucose and improves glucose intolerance. Decreased hepatic triglyceride level was associated with lower expression levels of fatty acid synthase and sterol regulatory element-binding protein-1c in MET+CGA treated mice. MET and CGA combination treatment resulted in the polarization of macrophages to the M2 phenotype, reduction of the expression of pro-inflammatory cytokines (TNF-α, IL-1β and IL-6), and decreasing protein level of NF-kB p65. It was found that the lowering effect of combined MET and CGA on the expression of gluconeogenic genes was accompanied by increasing phosphorylation of glycogen synthase kinase 3β. Treatment of HFD mice with the combination of MET and CGA was found to be more effective at alleviating inflammation and lipid accumulation by increasing phosphorylation of AMP-activated protein kinase. In conclusion, these findings suggest that the MET + CGA combination might exert therapeutic effects against NAFLD.
Collapse
Affiliation(s)
- Fahimeh Zamani-Garmsiri
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ghasem Ghasempour
- Department of Clinical Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Masoume Aliabadi
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Solaleh Emamgholipour
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Meshkani
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
50
|
Park JE, Jeong YJ, Kim HY, Yoo YH, Lee KS, Yang WT, Kim DH, Kim JM. Hepatic Steatosis Alleviated in Diabetic Mice upon Dietary Exposure to Fibroin via Transgenic Rice: Potential STAMP2 Involvement in Hepatocytes. Dev Reprod 2020; 24:231-239. [PMID: 33110955 PMCID: PMC7576964 DOI: 10.12717/dr.2020.24.3.231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 09/09/2020] [Accepted: 09/20/2020] [Indexed: 11/17/2022]
Abstract
Many benefits of silk protein fibroin (SPF) have been suggested in biomedical
applications; and notably, significant SPF effects have been observed for
metabolic syndromes that are directly linked to insulin resistance, such as type
2 diabetes mellitus (T2DM). Based on our previous findings, we believe that SPF
from spiders exhibits outstanding glucose-lowering effects in diabetic
BKS.Cg-m+/+Leprdb mice. In order to evaluate the
dietary effects of SPF in diabetic animals, we generated several lines of
transgenic rice (TR) that expresses SPF, and the feeding of TR-SPF to diabetic
animals decreased blood glucose levels, but did not change insulin levels.
Western blot analyses of hepatic proteins showed that AMP-activated protein
kinase (AMPK) expression and phosphorylation both decreased in TR-SPF-fed
groups, compared with controls. This finding suggests that the glucose-lowering
effects in this diabetic animal model might be AMPK-independent. In contrast,
six-transmembrane protein of prostate 2 (STAMP2) was upregulated after TR-SPF
exposure. Together with STAMP2, the Akt protein phosphorylation increased after
TR-SPF exposure, which indicates that STAMP2 leads to Akt phosphorylation and
thus increases insulin sensitivity in hepatocytes. Importantly, the hepatic
steatosis that was seen in the liver of diabetic mice was remarkably alleviated
in TR-SPF-fed mice. Hepatocytes that were immunopositive for STAMP2 were
overwhelmingly observed in hepatic tissues from TR-SPF-fed mice compared to the
control. Taken together, these results suggest that feeding diabetic mice with
TR-SPF upregulates STAMP2 expression and increases Akt phosphorylation in
hepatic tissues and thus potentially alleviates insulin resistance and hepatic
steatosis.
Collapse
Affiliation(s)
- Ji-Eun Park
- Dept. of Anatomy and Cell Biology, College of Medicine, Dong-A University, Busan 49201, Korea
| | - Yeon Jae Jeong
- Dept. of Anatomy and Cell Biology, College of Medicine, Dong-A University, Busan 49201, Korea
| | - Hye Young Kim
- Dept. of Anatomy and Cell Biology, College of Medicine, Dong-A University, Busan 49201, Korea
| | - Young Hyun Yoo
- Dept. of Anatomy and Cell Biology, College of Medicine, Dong-A University, Busan 49201, Korea
| | - Kwang Sik Lee
- College of Life Sciences and Natural Resources, Dong-A University, Busan 49315, Korea
| | - Won Tae Yang
- College of Life Sciences and Natural Resources, Dong-A University, Busan 49315, Korea
| | - Doh Hoon Kim
- College of Life Sciences and Natural Resources, Dong-A University, Busan 49315, Korea
| | - Jong-Min Kim
- Dept. of Anatomy and Cell Biology, College of Medicine, Dong-A University, Busan 49201, Korea
| |
Collapse
|