1
|
Lee HY, Min KJ. Dietary Restriction and Lipid Metabolism: Unveiling Pathways to Extended Healthspan. Nutrients 2024; 16:4424. [PMID: 39771045 PMCID: PMC11678862 DOI: 10.3390/nu16244424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 12/18/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
Dietary restriction (DR) has been reported to be a significant intervention that influences lipid metabolism and potentially modulates the aging process in a wide range of organisms. Lipid metabolism plays a pivotal role in the regulation of aging and longevity. In this review, we summarize studies on the significant role of lipid metabolism in aging in relation to DR. As a potent intervention to slow down aging, DR has demonstrated promising effects on lipid metabolism, influencing the aging processes across various species. The current review focuses on the relationships among DR-related molecular signaling proteins such as the sirtuins, signaling pathways such as the target of rapamycin and the insulin/insulin-like growth factor (IGF)-1, lipid metabolism, and aging. Furthermore, the review presents research results on diet-associated changes in cell membrane lipids and alterations in lipid metabolism caused by commensal bacteria, highlighting the importance of lipid metabolism in aging. Overall, the review explores the interplay between diet, lipid metabolism, and aging, while presenting untapped areas for further understanding of the aging process.
Collapse
Affiliation(s)
| | - Kyung-Jin Min
- Department of Biological Sciences and Bioengineering, Inha University, Incheon 22212, Republic of Korea;
| |
Collapse
|
2
|
Catalfamo VJ, Tutor AW, Koos A, Vyas A, Lavie CJ, Carbone S. Obesity, Metabolic Syndrome, and Obesity Paradox in Heart Failure: A Critical Evaluation. Curr Heart Fail Rep 2024; 22:1. [PMID: 39546123 DOI: 10.1007/s11897-024-00690-w] [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] [Accepted: 11/04/2024] [Indexed: 11/17/2024]
Abstract
PURPOSE OF REVIEW Since the turn of the millennium, obesity has been on the rise worldwide, and particularly so throughout the United States. Even more concerning is the rate at which persons with severe obesity continue to trend upwards. Given the detrimental effects of obesity on cardiac structure and function, it is not surprising that obesity stands as one of the leading risk factors for developing heart failure (HF). This state-of-the-art article aims to review the updated literature on the obesity paradox to help further understand its relationship to body composition, weight loss, fitness, and exercise. RECENT FINDINGS An intriguing phenomenon appears to exist in which obesity is associated with a better prognosis in those with HF, compared to patients with lesser body mass. Recent studies suggest, however, that weight loss induced by pharmacologic strategies might be beneficial in patients with HF with preserved ejection fraction. Despite the presence of an obesity paradox, recent data suggest that obesity could be targeted in HF, however, long-term data are currently lacking. Consequently, definitive guidelines for managing obesity, and specifically the body composition of these patients, remain amiss.
Collapse
Affiliation(s)
- Vince J Catalfamo
- Department of Internal Medicine, Ochsner Health Foundation, 1514 Jefferson Highway, New Orleans, LA, 70121, USA
| | - Austin W Tutor
- Department of Cardiovascular Health, Ochsner Health Foundation, 1514 Jefferson Highway, New Orleans, LA, 70121, USA
| | - Adrienne Koos
- Department of Internal Medicine, Ochsner Health Foundation, 1514 Jefferson Highway, New Orleans, LA, 70121, USA
| | - Ankit Vyas
- Department of Cardiovascular Health, Ochsner Health Foundation, 1514 Jefferson Highway, New Orleans, LA, 70121, USA
| | - Carl J Lavie
- Department of Cardiovascular Health, Ochsner Health Foundation, 1514 Jefferson Highway, New Orleans, LA, 70121, USA.
- The University of Queensland Faculty of Medicine, 20 Weightman St, Herston, QLD, 4006, Australia.
- Department of Cardiology, Ochsner Medical Center, 3rd Floor Atrium Tower, New Orleans, LA, 70121, USA.
| | - Salvatore Carbone
- Department of Kinesiology & Health Sciences, College of Humanities & Sciences, Virginia Commonwealth University, 817 W. Franklin St, Richmond, VA, USA
| |
Collapse
|
3
|
He S, Wang J, Zhou L, Mao Z, Zhang X, Cai J, Huang P. Enhanced hepatic metabolic perturbation of polystyrene nanoplastics by UV irradiation-induced hydroxyl radical generation. J Environ Sci (China) 2024; 142:259-268. [PMID: 38527891 DOI: 10.1016/j.jes.2023.06.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 06/24/2023] [Accepted: 06/25/2023] [Indexed: 03/27/2024]
Abstract
The environmental behavior of and risks associated with nanoplastics (NPs) have attracted considerable attention. However, compared to pristine NPs, environmental factors such as ultraviolet (UV) irradiation that lead to changes in the toxicity of NPs have rarely been studied. We evaluated the changes in morphology and physicochemical properties of polystyrene (PS) NPs before and after UV irradiation, and compared their hepatotoxicity in mice. The results showed that UV irradiation caused particle size reduction and increased the carbonyl index (CI) and negative charge on the particle surface. UV-aged PS NPs (aPS NPs) could induce the generation of hydroxyl radicals (·OH), but also further promoted the generation of ·OH in the Fenton reaction system. Hepatic pathological damage was more severe in mice exposed to aPS NPs, accompanied by a large number of vacuoles and hepatocyte balloon-like changes and more marked perturbations in blood glucose and serum lipoprotein, alanine aminotransferase and aspartate aminotransferase levels. In addition, exposure to PS NPs and aPS NPs, especially aPS NPs, triggered oxidative stress and significantly damaged the antioxidant capacity of mice liver. Compared with PS NPs, exposure to aPS NPs increased the number of altered metabolites in hepatic and corresponding metabolic pathways, especially glutathione metabolism. Our research suggests that UV irradiation can disrupt the redox balance in organisms by promoting the production of ·OH, enhancing PS NPs-induced liver damage and metabolic disorders. This study will help us understand the health risks of NPs and to avoid underestimation of the risks of NPs in nature.
Collapse
Affiliation(s)
- Shiyu He
- School of Public Health and Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Jingran Wang
- School of Public Health and Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Lihong Zhou
- School of Public Health and Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Zhen Mao
- School of Public Health and Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Xiaodan Zhang
- School of Public Health and Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Jin Cai
- School of Public Health and Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Peili Huang
- School of Public Health and Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
4
|
Abel ED, Gloyn AL, Evans-Molina C, Joseph JJ, Misra S, Pajvani UB, Simcox J, Susztak K, Drucker DJ. Diabetes mellitus-Progress and opportunities in the evolving epidemic. Cell 2024; 187:3789-3820. [PMID: 39059357 PMCID: PMC11299851 DOI: 10.1016/j.cell.2024.06.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 06/21/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024]
Abstract
Diabetes, a complex multisystem metabolic disorder characterized by hyperglycemia, leads to complications that reduce quality of life and increase mortality. Diabetes pathophysiology includes dysfunction of beta cells, adipose tissue, skeletal muscle, and liver. Type 1 diabetes (T1D) results from immune-mediated beta cell destruction. The more prevalent type 2 diabetes (T2D) is a heterogeneous disorder characterized by varying degrees of beta cell dysfunction in concert with insulin resistance. The strong association between obesity and T2D involves pathways regulated by the central nervous system governing food intake and energy expenditure, integrating inputs from peripheral organs and the environment. The risk of developing diabetes or its complications represents interactions between genetic susceptibility and environmental factors, including the availability of nutritious food and other social determinants of health. This perspective reviews recent advances in understanding the pathophysiology and treatment of diabetes and its complications, which could alter the course of this prevalent disorder.
Collapse
Affiliation(s)
- E Dale Abel
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.
| | - Anna L Gloyn
- Department of Pediatrics, Division of Endocrinology & Diabetes, Department of Genetics, Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA, USA
| | - Carmella Evans-Molina
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Joshua J Joseph
- Division of Endocrinology, Diabetes and Metabolism, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Shivani Misra
- Department of Metabolism, Digestion and Reproduction, Imperial College London, and Imperial College NHS Trust, London, UK
| | - Utpal B Pajvani
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Judith Simcox
- Howard Hughes Medical Institute, Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Katalin Susztak
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Daniel J Drucker
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada; Department of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
5
|
Zhou X, Xu J, Dai H. The ratio of alanine aminotransferase to high-density lipoprotein cholesterol is positively correlated with the insulin resistance in American adults: a population-based cohort study. Front Med (Lausanne) 2024; 11:1418364. [PMID: 38962742 PMCID: PMC11220187 DOI: 10.3389/fmed.2024.1418364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 06/05/2024] [Indexed: 07/05/2024] Open
Abstract
Introduction Previous studies have demonstrated a correlation between the ratio of alanine aminotransferase to high-density lipoprotein cholesterol (ALT/HDL-C) in the serum and the risk of diabetes. However, no existing study has investigated the association between insulin resistance (IR) and ALT/HDL-C. Therefore, this study aims to explore the association between ALT/HDL-C and IR in American adults. Methods A total of 7,599 adults selected from the National Health and Nutrition Examination Survey (NHANES) in 2013 to 2020 were studied. IR was assessed based on the homeostatic model assessment of insulin resistance (HOMA-IR). And the association between IR and ALT/HDL-C was assessed through multiple logistic regression, generalized smooth curve fitting and subgroup analyses. Results Multiple logistic regression analysis indicated a significant correlation between IR and ALT/HDL-C, with odds ratios (OR) of 1.04 (95% CI = 1.02-1.05) in males and 1.04 (95% CI = 1.02-1.07) in females. A non-linear association and saturation effect between ALT/HDL-C and IR risk were identified, with an inverted L shaped curve and an inflection point at 33.62. The area under the ROC curve (AUC) of ALT/HDL-C was significantly larger (AUC = 0.725 for males and 0.696 for females, all p < 0.01) compared with the use of ALT, HDL-C, AST and AST/ALT. Subgroup analysis showed a significantly higher independent association in obese individuals and individuals aged ≥50 years (All P interaction <0.05). Conclusion Elevated ALT/HDL-C demonstrates a significant correlation with IR, which can be used as a potential indicator of IR in American adults.
Collapse
Affiliation(s)
| | | | - Huifang Dai
- Department of Endocrinology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
6
|
Zhou X, Xu J. Association between serum uric acid-to-high-density lipoprotein cholesterol ratio and insulin resistance in an American population: A population-based analysis. J Diabetes Investig 2024; 15:762-771. [PMID: 38407574 PMCID: PMC11143423 DOI: 10.1111/jdi.14170] [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: 09/30/2023] [Revised: 01/31/2024] [Accepted: 02/13/2024] [Indexed: 02/27/2024] Open
Abstract
INTRODUCTION Previous studies have demonstrated a correlation between the serum uric acid-to-high-density lipoprotein cholesterol ratio (UHR) and insulin resistance (IR) in individuals with type 2 diabetes mellitus. However, no existing studies have investigated the relationship between IR and UHR in the general population. Therefore, the primary objective of this study was to investigate the correlation between UHR and IR in the general American population. METHODS A sample of 8,817 participants was selected from the 2013 to 2020 National Health and Nutrition Examination Survey (NHANES). Homeostatic model assessment of insulin resistance (HOMA-IR) was used to assess insulin resistance. Multiple logistic regression, generalized smooth curve fitting, and subgroup analysis were used to assess the association between IR and UHR. RESULTS Multiple logistic regression analysis indicated a significant correlation between insulin resistance and UHR, with odds ratios (OR) of 1.07 (95% CI = 1.03-1.11) in males and 1.18 (95% CI = 1.13-1.25) in females. A non-linear relationship and saturation effect between IR risk and UHR were observed, characterized by an inverted L-shaped curve and a critical inflection point at 8.82. It was found that the area under the ROC curve (AUC) of UHR was significantly larger (AUC = 0.703 for males and 0.747 for females, all P < 0.01) compared with the use of UA or HDL-C alone. Subgroup analysis showed that this independent association remain consistent regardless of race, age, BMI, diabetes, moderate activities, education level, alcohol drinking, and gender. CONCLUSION Elevated UHR demonstrates a significant correlation with insulin resistance, so it can be used as a potential indicator of insulin resistance within the American population.
Collapse
Affiliation(s)
- Xiaohai Zhou
- Department of Hematology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jing Xu
- Department of Endocrinology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
7
|
Aizenshtadt A, Wang C, Abadpour S, Menezes PD, Wilhelmsen I, Dalmao‐Fernandez A, Stokowiec J, Golovin A, Johnsen M, Combriat TMD, Røberg‐Larsen H, Gadegaard N, Scholz H, Busek M, Krauss SJK. Pump-Less, Recirculating Organ-on-Chip (rOoC) Platform to Model the Metabolic Crosstalk between Islets and Liver. Adv Healthc Mater 2024; 13:e2303785. [PMID: 38221504 PMCID: PMC11468483 DOI: 10.1002/adhm.202303785] [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: 10/31/2023] [Revised: 12/05/2023] [Indexed: 01/16/2024]
Abstract
Type 2 diabetes mellitus (T2DM), obesity, and metabolic dysfunction-associated steatotic liver disease (MASLD) are epidemiologically correlated disorders with a worldwide growing prevalence. While the mechanisms leading to the onset and development of these conditions are not fully understood, predictive tissue representations for studying the coordinated interactions between central organs that regulate energy metabolism, particularly the liver and pancreatic islets, are needed. Here, a dual pump-less recirculating organ-on-chip platform that combines human pluripotent stem cell (sc)-derived sc-liver and sc-islet organoids is presented. The platform reproduces key aspects of the metabolic cross-talk between both organs, including glucose levels and selected hormones, and supports the viability and functionality of both sc-islet and sc-liver organoids while preserving a reduced release of pro-inflammatory cytokines. In a model of metabolic disruption in response to treatment with high lipids and fructose, sc-liver organoids exhibit hallmarks of steatosis and insulin resistance, while sc-islets produce pro-inflammatory cytokines on-chip. Finally, the platform reproduces known effects of anti-diabetic drugs on-chip. Taken together, the platform provides a basis for functional studies of obesity, T2DM, and MASLD on-chip, as well as for testing potential therapeutic interventions.
Collapse
Affiliation(s)
- Aleksandra Aizenshtadt
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
- Dep. of Immunology and Transfusion MedicineOslo University HospitalP.O. Box 4950Oslo0424Norway
| | - Chencheng Wang
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
- Dep. of Transplantation MedicineExperimental Cell Transplantation Research GroupOslo University HospitalP.O. Box 4950Oslo0424Norway
| | - Shadab Abadpour
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
- Dep. of Transplantation MedicineExperimental Cell Transplantation Research GroupOslo University HospitalP.O. Box 4950Oslo0424Norway
- Institute for Surgical ResearchOslo University HospitalOsloNorway
| | - Pedro Duarte Menezes
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
- James Watt School of EngineeringUniversity of GlasgowRankine BuildingGlasgowG12 8LTUK
| | - Ingrid Wilhelmsen
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
- Dep. of Immunology and Transfusion MedicineOslo University HospitalP.O. Box 4950Oslo0424Norway
| | - Andrea Dalmao‐Fernandez
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
- Department of PharmacyFaculty of Mathematics and Natural SciencesUniversity of OsloP.O. Box 1083Oslo0316Norway
| | - Justyna Stokowiec
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
- Dep. of Immunology and Transfusion MedicineOslo University HospitalP.O. Box 4950Oslo0424Norway
| | - Alexey Golovin
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
- Dep. of Immunology and Transfusion MedicineOslo University HospitalP.O. Box 4950Oslo0424Norway
| | - Mads Johnsen
- Section for Chemical Life SciencesDepartment of ChemistryUniversity of OsloP.O. Box 1033Oslo0315Norway
| | - Thomas M. D. Combriat
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
| | - Hanne Røberg‐Larsen
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
- Section for Chemical Life SciencesDepartment of ChemistryUniversity of OsloP.O. Box 1033Oslo0315Norway
| | - Nikolaj Gadegaard
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
- James Watt School of EngineeringUniversity of GlasgowRankine BuildingGlasgowG12 8LTUK
| | - Hanne Scholz
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
- Dep. of Transplantation MedicineExperimental Cell Transplantation Research GroupOslo University HospitalP.O. Box 4950Oslo0424Norway
| | - Mathias Busek
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
- Dep. of Immunology and Transfusion MedicineOslo University HospitalP.O. Box 4950Oslo0424Norway
| | - Stefan J. K. Krauss
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
- Dep. of Immunology and Transfusion MedicineOslo University HospitalP.O. Box 4950Oslo0424Norway
| |
Collapse
|
8
|
Silva RCMC, Ramos IB, Travassos LH, Mendez APG, Gomes FM. Evolution of innate immunity: lessons from mammalian models shaping our current view of insect immunity. J Comp Physiol B 2024; 194:105-119. [PMID: 38573502 DOI: 10.1007/s00360-024-01549-1] [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: 10/07/2023] [Revised: 01/23/2024] [Accepted: 03/09/2024] [Indexed: 04/05/2024]
Abstract
The innate immune system, a cornerstone for organismal resilience against environmental and microbial insults, is highly conserved across the evolutionary spectrum, underpinning its pivotal role in maintaining homeostasis and ensuring survival. This review explores the evolutionary parallels between mammalian and insect innate immune systems, illuminating how investigations into these disparate immune landscapes have been reciprocally enlightening. We further delve into how advancements in mammalian immunology have enriched our understanding of insect immune responses, highlighting the intertwined evolutionary narratives and the shared molecular lexicon of immunity across these organisms. Therefore, this review posits a holistic understanding of innate immune mechanisms, including immunometabolism, autophagy and cell death. The examination of how emerging insights into mammalian and vertebrate immunity inform our understanding of insect immune responses and their implications for vector-borne disease transmission showcases the imperative for a nuanced comprehension of innate immunity's evolutionary tale. This understanding is quintessential for harnessing innate immune mechanisms' potential in devising innovative disease mitigation strategies and promoting organismal health across the animal kingdom.
Collapse
Affiliation(s)
- Rafael Cardoso M C Silva
- Laboratory of Immunoreceptors and Signaling, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Isabela B Ramos
- Laboratório de Ovogênese Molecular de Vetores, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Entomologia Molecular, Rio de Janeiro, Brazil
| | - Leonardo H Travassos
- Laboratory of Immunoreceptors and Signaling, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana Paula Guzman Mendez
- Laboratório de Ultraestrutura Celular Hertha Meyer, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fabio M Gomes
- Instituto Nacional de Entomologia Molecular, Rio de Janeiro, Brazil.
- Laboratório de Ultraestrutura Celular Hertha Meyer, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
9
|
Agius R, Pace NP, Fava S. Phenotyping obesity: A focus on metabolically healthy obesity and metabolically unhealthy normal weight. Diabetes Metab Res Rev 2024; 40:e3725. [PMID: 37792999 DOI: 10.1002/dmrr.3725] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 07/23/2023] [Accepted: 08/11/2023] [Indexed: 10/06/2023]
Abstract
Over the past 4 decades, research has shown that having a normal body weight does not automatically imply preserved metabolic health and a considerable number of lean individuals harbour metabolic abnormalities typically associated with obesity. Conversely, excess adiposity does not always equate with an abnormal metabolic profile. In fact, evidence exists for the presence of a metabolically unhealthy normal weight (MUHNW) and a metabolically healthy obese (MHO) phenotype. It has become increasingly recognised that different fat depots exert different effects on the metabolic profile of each individual by virtue of their location, structure and function, giving rise to these different body composition phenotypes. Furthermore, other factors have been implicated in the aetiopathogenesis of the body composition phenotypes, including genetics, ethnicity, age and lifestyle/behavioural factors. Even though to date both MHO and MUHNW have been widely investigated and documented in the literature, studies report different outcomes on long-term cardiometabolic morbidity and mortality. Future large-scale, observational and population-based studies are required for better profiling of these phenotypes as well as to further elucidate the pathophysiological role of the adipocyte in the onset of metabolic disorders to allow for better risk stratification and a personalised treatment paradigm.
Collapse
Affiliation(s)
- Rachel Agius
- University of Malta Medical School, Msida, Malta
- Mater Dei Hospital, Msida, Malta
| | | | - Stephen Fava
- University of Malta Medical School, Msida, Malta
- Mater Dei Hospital, Msida, Malta
| |
Collapse
|
10
|
Zhou X, Xu J. Association between serum uric acid-to-high-density lipoprotein cholesterol ratio and insulin resistance in patients with type 2 diabetes mellitus. J Diabetes Investig 2024; 15:113-120. [PMID: 37737515 PMCID: PMC10759725 DOI: 10.1111/jdi.14086] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 08/28/2023] [Accepted: 09/01/2023] [Indexed: 09/23/2023] Open
Abstract
INTRODUCTION Previous studies have shown that the serum uric acid-to-high-density lipoprotein cholesterol ratio (UHR) is related to metabolic syndrome. However, no existing study has examined the relationship between UHR and insulin resistance (IR). Therefore, this study aims to explore the association between the UHR and IR in patients with type 2 diabetes mellitus (T2DM). METHODS Patients with type 2 diabetes mellitus (1,532 males and 1,013 females) were enrolled. Insulin resistance was measured by homeostatic model assessment of insulin resistance (HOMA-IR) and was defined as HOMI-IR ≥ 2.69. Pearson correlation, multiple logistic regression, ROC analysis, and subgroup analysis were used to evaluate the association between UHR and IR. RESULTS UHR was associated with HOMA-IR in patients with type 2 diabetes mellitus (pearson's correlation coefficient = 0.274 in males and 0.337 in females, P < 0.001). Multiple logistic regression analysis showed that UHR was significantly correlated with insulin resistance (OR = 1.06, 95%CI = 1.03-1.08 in males and OR = 1.11, 95%CI = 1.08-1.15 in females). The area under the ROC curve (AUC) of UHR (AUC = 0.665 for males and 0.717 for females, all P < 0.01) was the largest compared with that of UA and HDL-C in insulin resistance. Subgroup analysis showed that there was a more significantly positive correlation among subjects with BMI ≥ 24 kg/m2 , age < 60 years old, HbA1c < 7%, non-hypertension, or in female subjects. CONCLUSION Elevated UHR is significantly correlated with insulin resistance, which can be used as an indicator of insulin resistance in patients with type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Xinhe Zhou
- Department of EndocrinologyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Jing Xu
- Department of EndocrinologyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouChina
| |
Collapse
|
11
|
Peng J, Yu L, Huang L, Paschoal VA, Chu H, de Souza CO, Varre JV, Oh DY, Kohler JJ, Xiao X, Xu L, Holland WL, Shaul PW, Mineo C. Hepatic sialic acid synthesis modulates glucose homeostasis in both liver and skeletal muscle. Mol Metab 2023; 78:101812. [PMID: 37777009 PMCID: PMC10583174 DOI: 10.1016/j.molmet.2023.101812] [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: 08/01/2023] [Revised: 09/22/2023] [Accepted: 09/26/2023] [Indexed: 10/02/2023] Open
Abstract
OBJECTIVE Sialic acid is a terminal monosaccharide of glycans in glycoproteins and glycolipids, and its derivation from glucose is regulated by the rate-limiting enzyme UDP-GlcNAc 2-epimerase/ManNAc kinase (GNE). Although the glycans on key endogenous hepatic proteins governing glucose metabolism are sialylated, how sialic acid synthesis and sialylation in the liver influence glucose homeostasis is unknown. Studies were designed to fill this knowledge gap. METHODS To decrease the production of sialic acid and sialylation in hepatocytes, a hepatocyte-specific GNE knockdown mouse model was generated, and systemic glucose metabolism, hepatic insulin signaling and glucagon signaling were evaluated in vivo or in primary hepatocytes. Peripheral insulin sensitivity was also assessed. Furthermore, the mechanisms by which sialylation in the liver influences hepatic insulin signaling and glucagon signaling and peripheral insulin sensitivity were identified. RESULTS Liver GNE deletion in mice caused an impairment of insulin suppression of hepatic glucose production. This was due to a decrease in the sialylation of hepatic insulin receptors (IR) and a decline in IR abundance due to exaggerated degradation through the Eph receptor B4. Hepatic GNE deficiency also caused a blunting of hepatic glucagon receptor (GCGR) function which was related to a decline in its sialylation and affinity for glucagon. An accompanying upregulation of hepatic FGF21 production caused an enhancement of skeletal muscle glucose disposal that led to an overall increase in glucose tolerance and insulin sensitivity. CONCLUSION These collective observations reveal that hepatic sialic acid synthesis and sialylation modulate glucose homeostasis in both the liver and skeletal muscle. By interrogating how hepatic sialic acid synthesis influences glucose control mechanisms in the liver, a new metabolic cycle has been identified in which a key constituent of glycans generated from glucose modulates the systemic control of its precursor.
Collapse
Affiliation(s)
- Jun Peng
- Center for Pulmonary and Vascular Biology, Dept. of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA.
| | - Liming Yu
- Center for Pulmonary and Vascular Biology, Dept. of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Linzhang Huang
- Center for Pulmonary and Vascular Biology, Dept. of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Vivian A Paschoal
- Dept. of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Haiyan Chu
- Center for Pulmonary and Vascular Biology, Dept. of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Camila O de Souza
- Dept. of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Joseph V Varre
- Dept. of Nutrition & Integrative Physiology, University of Utah College of Health, 250 1850 E, Salt Lake City, UT, 84112, USA
| | - Da Young Oh
- Dept. of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Jennifer J Kohler
- Dept. of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Xue Xiao
- Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Lin Xu
- Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - William L Holland
- Dept. of Nutrition & Integrative Physiology, University of Utah College of Health, 250 1850 E, Salt Lake City, UT, 84112, USA
| | - Philip W Shaul
- Center for Pulmonary and Vascular Biology, Dept. of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA.
| | - Chieko Mineo
- Center for Pulmonary and Vascular Biology, Dept. of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA; Dept. of Cell Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA.
| |
Collapse
|
12
|
Zhang Y, Xu J, Zhou D, Ye T, Zhou P, Liu Z, Liu X, Wang Z, Hua T, Zhang Z, Sun Q. Swimming exercise ameliorates insulin resistance and nonalcoholic fatty liver by negatively regulating PPARγ transcriptional network in mice fed high fat diet. Mol Med 2023; 29:150. [PMID: 37907845 PMCID: PMC10617119 DOI: 10.1186/s10020-023-00740-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 10/16/2023] [Indexed: 11/02/2023] Open
Abstract
BACKGROUND Recent findings elucidated hepatic PPARγ functions as a steatogenic-inducer gene that activates de novo lipogenesis, and is involved in regulation of glucose homeostasis, lipid accumulation, and inflammation response. This study delved into a comprehensive analysis of how PPARγ signaling affects the exercise-induced improvement of insulin resistance (IR) and non-alcoholic fatty liver disease (NAFLD), along with its underlying mechanism. METHODS Chronic and acute swimming exercise intervention were conducted in each group mice. IR status was assessed by GTT and ITT assays. Serum inflammatory cytokines were detected by Elisa assays. PPARγ and its target genes expression were detected by qPCR assay. Relative protein levels were quantified via Western blotting. ChIP-qPCR assays were used to detect the enrichment of PPARγ on its target genes promoter. RESULTS Through an exploration of a high-fat diet (HFD)-induced IR and NAFLD model, both chronic and acute swimming exercise training led to significant reductions in body weight and visceral fat mass, as well as hepatic lipid accumulation. The exercise interventions also demonstrated a significant amelioration in IR and the inflammatory response. Meanwhile, swimming exercise significantly inhibited PPARγ and its target genes expression induced by HFD, containing CD36, SCD1 and PLIN2. Furthermore, swimming exercise presented significant modulation on regulatory factors of PPARγ expression and transcriptional activity. CONCLUSION The findings suggest that swimming exercise can improve lipid metabolism in IR and NAFLD, possibly through PPARγ signaling in the liver of mice.
Collapse
Affiliation(s)
- Yong Zhang
- Physiology laboratory of College of Life Sciences, Anhui Normal University, Wuhu, China
- the State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Jie Xu
- Department of Hepatology, Affiliated Hospital of Panzhihua University, Panzhihua, China
| | - Di Zhou
- Physiology laboratory of College of Life Sciences, Anhui Normal University, Wuhu, China
| | - Tingting Ye
- Physiology laboratory of College of Life Sciences, Anhui Normal University, Wuhu, China
| | - Puqing Zhou
- Physiology laboratory of College of Life Sciences, Anhui Normal University, Wuhu, China
| | - Zuofeng Liu
- Department of Hepatology, Affiliated Hospital of Panzhihua University, Panzhihua, China
| | - Xinyuan Liu
- the State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Zinan Wang
- the State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Tianmiao Hua
- Physiology laboratory of College of Life Sciences, Anhui Normal University, Wuhu, China
| | - Zhenghao Zhang
- Department of Hematology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China.
| | - Qingyan Sun
- Physiology laboratory of College of Life Sciences, Anhui Normal University, Wuhu, China.
| |
Collapse
|
13
|
Louca P, Štambuk T, Frkatović-Hodžić A, Nogal A, Mangino M, Berry SE, Deriš H, Hadjigeorgiou G, Wolf J, Vinicki M, Franks PW, Valdes AM, Spector TD, Lauc G, Menni C. Plasma protein N-glycome composition associates with postprandial lipaemic response. BMC Med 2023; 21:231. [PMID: 37400796 PMCID: PMC10318725 DOI: 10.1186/s12916-023-02938-z] [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: 01/27/2023] [Accepted: 06/13/2023] [Indexed: 07/05/2023] Open
Abstract
BACKGROUND A dysregulated postprandial metabolic response is a risk factor for chronic diseases, including type 2 diabetes mellitus (T2DM). The plasma protein N-glycome is implicated in both lipid metabolism and T2DM risk. Hence, we first investigate the relationship between the N-glycome and postprandial metabolism and then explore the mediatory role of the plasma N-glycome in the relationship between postprandial lipaemia and T2DM. METHODS We included 995 individuals from the ZOE-PREDICT 1 study with plasma N-glycans measured by ultra-performance liquid chromatography at fasting and triglyceride, insulin, and glucose levels measured at fasting and following a mixed-meal challenge. Linear mixed models were used to investigate the associations between plasma protein N-glycosylation and metabolic response (fasting, postprandial (Cmax), or change from fasting). A mediation analysis was used to further explore the relationship of the N-glycome in the prediabetes (HbA1c = 39-47 mmol/mol (5.7-6.5%))-postprandial lipaemia association. RESULTS We identified 36 out of 55 glycans significantly associated with postprandial triglycerides (Cmax β ranging from -0.28 for low-branched glycans to 0.30 for GP26) after adjusting for covariates and multiple testing (padjusted < 0.05). N-glycome composition explained 12.6% of the variance in postprandial triglycerides not already explained by traditional risk factors. Twenty-seven glycans were also associated with postprandial glucose and 12 with postprandial insulin. Additionally, 3 of the postprandial triglyceride-associated glycans (GP9, GP11, and GP32) also correlate with prediabetes and partially mediate the relationship between prediabetes and postprandial triglycerides. CONCLUSIONS This study provides a comprehensive overview of the interconnections between plasma protein N-glycosylation and postprandial responses, demonstrating the incremental predictive benefit of N-glycans. We also suggest a considerable proportion of the effect of prediabetes on postprandial triglycerides is mediated by some plasma N-glycans.
Collapse
Affiliation(s)
- Panayiotis Louca
- Department of Twin Research and Genetic Epidemiology, King's College London, St Thomas' Hospital Campus, Westminster Bridge Road, London, SE1 7EH, UK
| | | | | | - Ana Nogal
- Department of Twin Research and Genetic Epidemiology, King's College London, St Thomas' Hospital Campus, Westminster Bridge Road, London, SE1 7EH, UK
| | - Massimo Mangino
- Department of Twin Research and Genetic Epidemiology, King's College London, St Thomas' Hospital Campus, Westminster Bridge Road, London, SE1 7EH, UK
- NIHR Biomedical Research Centre at Guy's and St Thomas' Foundation Trust, London, SE1 9RT, UK
| | - Sarah E Berry
- Department of Nutritional Sciences, King's College London, Franklin Wilkins Building, London, SE1 9NH, UK
| | - Helena Deriš
- Genos Glycoscience Research Laboratory, Zagreb, Croatia
| | | | | | | | - Paul W Franks
- Lund University Diabetes Center, Lund University, Malmö, Sweden
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Ana M Valdes
- Academic Rheumatology Clinical Sciences Building, Nottingham City Hospital, University of Nottingham, Nottingham, UK
| | - Tim D Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, St Thomas' Hospital Campus, Westminster Bridge Road, London, SE1 7EH, UK
| | - Gordan Lauc
- Genos Glycoscience Research Laboratory, Zagreb, Croatia
- University of Zagreb Faculty of Pharmacy and Biochemistry, Zagreb, Croatia
| | - Cristina Menni
- Department of Twin Research and Genetic Epidemiology, King's College London, St Thomas' Hospital Campus, Westminster Bridge Road, London, SE1 7EH, UK.
| |
Collapse
|
14
|
Tian Y, Mehta K, Jellinek MJ, Sun H, Lu W, Shi R, Ingram K, Friedline RH, Kim JK, Kemper JK, Ford DA, Zhang K, Wang B. Hepatic Phospholipid Remodeling Modulates Insulin Sensitivity and Systemic Metabolism. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300416. [PMID: 37088778 PMCID: PMC10288282 DOI: 10.1002/advs.202300416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 04/13/2023] [Indexed: 05/03/2023]
Abstract
The liver plays a central role in regulating glucose and lipid metabolism. Aberrant insulin action in the liver is a major driver of selective insulin resistance, in which insulin fails to suppress glucose production but continues to activate lipogenesis in the liver, resulting in hyperglycemia and hypertriglyceridemia. The underlying mechanisms of selective insulin resistance are not fully understood. Here It is shown that hepatic membrane phospholipid composition controlled by lysophosphatidylcholine acyltransferase 3 (LPCAT3) regulates insulin signaling and systemic glucose and lipid metabolism. Hyperinsulinemia induced by high-fat diet (HFD) feeding augments hepatic Lpcat3 expression and membrane unsaturation. Loss of Lpcat3 in the liver improves insulin resistance and blunts lipogenesis in both HFD-fed and genetic ob/ob mouse models. Mechanistically, Lpcat3 deficiency directly facilitates insulin receptor endocytosis, signal transduction, and hepatic glucose production suppression and indirectly enhances fibroblast growth factor 21 (FGF21) secretion, energy expenditure, and glucose uptake in adipose tissue. These findings identify hepatic LPCAT3 and membrane phospholipid composition as a novel regulator of insulin sensitivity and provide insights into the pathogenesis of selective insulin resistance.
Collapse
Affiliation(s)
- Ye Tian
- Department of Comparative BiosciencesCollege of Veterinary MedicineUniversity of Illinois at Urbana‐ChampaignUrbanaIL61802USA
| | - Kritika Mehta
- Department of BiochemistrySchool of Molecular and Cellular BiologyUniversity of Illinois at Urbana‐ChampaignUrbanaIL61801USA
| | - Matthew J. Jellinek
- Department of Biochemistry and Molecular Biologyand Center for Cardiovascular ResearchSaint Louis UniversitySt. LouisMO63104USA
| | - Hao Sun
- Department of Molecular and Integrative PhysiologySchool of Molecular and Cellular BiologyUniversity of Illinois at Urbana‐ChampaignUrbanaIL61801USA
| | - Wei Lu
- Department of Comparative BiosciencesCollege of Veterinary MedicineUniversity of Illinois at Urbana‐ChampaignUrbanaIL61802USA
| | - Ruicheng Shi
- Department of Comparative BiosciencesCollege of Veterinary MedicineUniversity of Illinois at Urbana‐ChampaignUrbanaIL61802USA
| | - Kevin Ingram
- Department of BiochemistrySchool of Molecular and Cellular BiologyUniversity of Illinois at Urbana‐ChampaignUrbanaIL61801USA
| | - Randall H. Friedline
- Program in Molecular Medicine and Division of EndocrinologyMetabolism and DiabetesDepartment of MedicineUniversity of Massachusetts Medical SchoolWorcesterMA01655USA
| | - Jason K. Kim
- Program in Molecular Medicine and Division of EndocrinologyMetabolism and DiabetesDepartment of MedicineUniversity of Massachusetts Medical SchoolWorcesterMA01655USA
| | - Jongsook Kim Kemper
- Department of Molecular and Integrative PhysiologySchool of Molecular and Cellular BiologyUniversity of Illinois at Urbana‐ChampaignUrbanaIL61801USA
- Cancer Center at IllinoisUniversity of Illinois at Urbana‐ChampaignUrbanaIL61801USA
| | - David A. Ford
- Department of Biochemistry and Molecular Biologyand Center for Cardiovascular ResearchSaint Louis UniversitySt. LouisMO63104USA
| | - Kai Zhang
- Department of BiochemistrySchool of Molecular and Cellular BiologyUniversity of Illinois at Urbana‐ChampaignUrbanaIL61801USA
| | - Bo Wang
- Department of Comparative BiosciencesCollege of Veterinary MedicineUniversity of Illinois at Urbana‐ChampaignUrbanaIL61802USA
- Cancer Center at IllinoisUniversity of Illinois at Urbana‐ChampaignUrbanaIL61801USA
- Division of Nutritional SciencesCollege of AgriculturalConsumer and Environmental SciencesUniversity of Illinois at Urbana‐ChampaignUrbanaIL61801USA
| |
Collapse
|
15
|
Delangre E, Pommier G, Tolu S, Uzan B, Bailbé D, Movassat J. Lithium treatment mitigates the diabetogenic effects of chronic cortico-therapy. Biomed Pharmacother 2023; 164:114895. [PMID: 37224758 DOI: 10.1016/j.biopha.2023.114895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/09/2023] [Accepted: 05/13/2023] [Indexed: 05/26/2023] Open
Abstract
BACKGROUND AND PURPOSE Glucocorticoids (GCs) are the main treatment for autoimmune and inflammatory disorders and are also used as immunosuppressive therapy for patients with organ transplantation. However, these treatments have several side effects, including metabolic disorders. Indeed, cortico-therapy may induce insulin resistance, glucose intolerance, disrupted insulin and glucagon secretion, excessive gluconeogenesis, leading to diabetes in susceptible individuals. Recently, lithium has been shown to alleviate deleterious effects of GCs in various diseased conditions. EXPERIMENTAL APPROACH In this study, using two rat models of GC-induced metabolic disorders, we investigated the effects of Lithium Chloride (LiCl) in the mitigation of deleterious effects of GCs. Rats were treated either with corticosterone or dexamethasone, and with or without LiCl. Animals were then assessed for glucose tolerance, insulin sensitivity, in vivo and ex vivo glucose-induced insulin secretion and hepatic gluconeogenesis. KEY RESULTS We showed that in rats chronically treated with corticosterone, lithium treatment markedly reduced insulin resistance. In addition, in rats treated with dexamethasone, lithium administration improved glucose tolerance, associated with enhanced insulin secretion in vivo. Moreover, liver gluconeogenesis was reduced upon LiCl treatment. The improvement of insulin secretion in vivo appeared to be due to an indirect regulation of β cell function, since the ex vivo assessment of insulin secretion and β cell mass in islets from animals treated with LiCl revealed no difference compared to untreated animals. CONCLUSION AND IMPLICATIONS Collectively, our data provide evidences for the beneficial effects of lithium to mitigate the adverse metabolic effects of chronic cortico-therapy.
Collapse
Affiliation(s)
- Etienne Delangre
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, F-75013 Paris, France
| | - Gaëlle Pommier
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, F-75013 Paris, France; Université Paris Cité, UFR Sciences du Vivant, F-75013 Paris, France
| | - Stefania Tolu
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, F-75013 Paris, France
| | - Benjamin Uzan
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, F-75013 Paris, France
| | - Danielle Bailbé
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, F-75013 Paris, France
| | - Jamileh Movassat
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, F-75013 Paris, France.
| |
Collapse
|
16
|
Wei M, Zhang T, Ouyang H, Huang Z, Lu B, Li J, Xu H, Wang Z, Ji L. Erianin alleviated liver steatosis by enhancing Nrf2-mediated VE-cadherin expression in vascular endothelium. Eur J Pharmacol 2023; 950:175744. [PMID: 37094711 DOI: 10.1016/j.ejphar.2023.175744] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 04/20/2023] [Accepted: 04/21/2023] [Indexed: 04/26/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is emerging as the most common chronic liver disease and is closely associated with metabolic syndrome. Endothelial dysfunction was involved in many metabolic diseases, but the concrete participation of hepatic vascular endothelial dysfunction in liver steatosis that is an early stage of NAFLD is still unclear. In this study, the formation of liver steatosis and the elevation of serum insulin content were observed accompanying with the decreased vascular endothelial cadherin (VE-cadherin) expression in hepatic vessels from db/db mice, Goto-Kakizaki (GK) and high-fat diet (HFD)-fed rats. Liver steatosis was obviously enhanced in mice after the application of VE-cadherin neutralizing antibody. In vitro results showed that insulin decreased VE-cadherin expression and caused endothelial barrier breakdown. Furthermore, the alteration of VE-cadherin expression was found to be positively related with the transcriptional activation of nuclear erythroid 2-related factor 2 (Nrf2), and chromatin immunoprecipitation (ChIP) assay displayed that Nrf2 could directly regulate VE-cadherin expression. Insulin reduced Nrf2 activation by decreasing sequestosome-1 (p62/SQSTM1) expression downstream of insulin receptor. Moreover, the p300-mediated Nrf2 acetylation was weakened by enhancing the competitive binding of transcription factor GATA-binding protein 4 (GATA4) to p300. Finally, we found that erianin, a natural compound, could promote VE-cadherin expression by inducing Nrf2 activation, thereby alleviating liver steatosis in GK rats. Our results suggest that hepatic vascular endothelial dysfunction owing to the VE-cadherin deficiency dependent on the reduced Nrf2 activation promoted liver steatosis, and erianin alleviated liver steatosis through enhancing Nrf2-mediated VE-cadherin expression.
Collapse
Affiliation(s)
- Mengjuan Wei
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China.
| | - Tianyu Zhang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China.
| | - Hao Ouyang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China.
| | - Zhenlin Huang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China.
| | - Bin Lu
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China.
| | - Jian Li
- The Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China; Jinling Pharmaceutical Co., Ltd., Nanjing, 210009, China.
| | - Hong Xu
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China.
| | - Zhengtao Wang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China.
| | - Lili Ji
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China.
| |
Collapse
|
17
|
Mohan J, Ghazi T, Sibiya T, Chuturgoon AA. Antiretrovirals Promote Metabolic Syndrome through Mitochondrial Stress and Dysfunction: An In Vitro Study. BIOLOGY 2023; 12:biology12040580. [PMID: 37106780 PMCID: PMC10135454 DOI: 10.3390/biology12040580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/28/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023]
Abstract
The prevalence of metabolic syndrome MetS in HIV-infected patients on chronic antiretroviral (ARV) therapy continues to rise rapidly, with an estimated 21% experiencing insulin resistance. The progression of insulin resistance is strongly related to mitochondrial stress and dysfunction. This study aimed to draw links between the singular and combinational use of Tenofovir disoproxil fumarate (TDF), Lamivudine (3TC), and Dolutegravir (DTG) on mitochondrial stress and dysfunction as an underlying mechanism for insulin resistance following a 120 h treatment period using an in vitro system of human liver cells (HepG2). The relative protein expressions of pNrf2, SOD2, CAT, PINK1, p62, SIRT3, and UCP2, were determined using Western blot. Transcript levels of PINK1 and p62 were assessed using quantitative PCR (qPCR). ATP concentrations were quantified using luminometry, and oxidative damage (malondialdehyde (MDA) concentration) was measured using spectrophotometry. The findings suggest that despite the activation of antioxidant responses (pNrf2, SOD2, CAT) and mitochondrial maintenance systems (PINK1 and p62) in selected singular and combinational treatments with ARVs, oxidative damage and reduced ATP production persisted. This was attributed to a significant suppression in mitochondrial stress responses SIRT3 and UCP2 for all treatments. Notable results were observed for combinational treatments with significant increases in pNrf2 (p = 0.0090), SOD2 (p = 0.0005), CAT (p = 0.0002), PINK1 (p = 0.0064), and p62 (p = 0.0228); followed by significant decreases in SIRT3 (p = 0.0003) and UCP2 (p = 0.0119) protein expression. Overall there were elevated levels of MDA (p = 0.0066) and decreased ATP production (p = 0.0017). In conclusion, ARVs induce mitochondrial stress and dysfunction, which may be closely associated with the progression of insulin resistance.
Collapse
Affiliation(s)
- Jivanka Mohan
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban 4041, South Africa
| | - Terisha Ghazi
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban 4041, South Africa
| | - Thabani Sibiya
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban 4041, South Africa
| | - Anil A Chuturgoon
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban 4041, South Africa
| |
Collapse
|
18
|
Immune response gene 1 deficiency aggravates high fat diet-induced nonalcoholic fatty liver disease via promotion of redox-sensitive AKT suppression. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166656. [PMID: 36706797 DOI: 10.1016/j.bbadis.2023.166656] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 01/18/2023] [Accepted: 01/19/2023] [Indexed: 01/26/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disorder worldwide. Immune response gene 1 (IRG1) catalyzes the production of bio-active itaconate, which is actively involved in the regulation of signal transduction. A recent study has found that the expression of IRG1 was significantly down-regulated in obesity-associated fatty liver, but the potential roles of IRG1 in the development NAFLD remain unclear. The present study found that genetic deletion of IRG1 aggravated high fat diet (HFD)-induced metabolic disturbance, including obesity, dyslipidemia and insulin resistance. In addition, HFD induced more severe liver steatosis and higher serum ALT and AST level in IRG1 KO mice, which were accompanied with altered expression of genes involved in lipid uptake, synthesis and catabolism. RNA-seq and immunoblot analysis indicated that deficiency of IRG1 is associated with suppressed activation of AKT, a master metabolic regulator. Mechanistically, IRG1/itaconate enhanced the antioxidative NRF2 pathway and prevented redox-sensitive suppression of AKT. Interestingly, supplementation with 4-octyl itaconate (4-OI), a cell-permeable derivate of itaconate, alleviated HFD-induced oxidative stress, AKT suppression and liver steatosis. Therefore, IRG1 probably functions as a protective regulator in the development of NAFLD and the cell-permeable 4-OI might have potential value for the pharmacological intervention of NAFLD.
Collapse
|
19
|
Mohan J, Ghazi T, Mazibuko MS, Chuturgoon AA. Antiretrovirals Promote Insulin Resistance in HepG2 Liver Cells through miRNA Regulation and Transcriptional Activation of the NLRP3 Inflammasome. Int J Mol Sci 2023; 24:ijms24076267. [PMID: 37047241 PMCID: PMC10094183 DOI: 10.3390/ijms24076267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/23/2023] [Accepted: 03/24/2023] [Indexed: 03/29/2023] Open
Abstract
Metabolic syndrome (MetS) is a non-communicable disease characterized by a cluster of metabolic irregularities. Alarmingly, the prevalence of MetS in people living with Human Immunodeficiency Virus (HIV) and antiretroviral (ARV) usage is increasing rapidly. Insulin resistance is a common characteristic of MetS that leads to the development of Type 2 diabetes mellitus (T2DM). The progression of insulin resistance is strongly linked to inflammasome activation. This study aimed to draw links between the combinational use of Tenofovir disoproxil fumarate (TDF), Lamivudine (3TC), and Dolutegravir (DTG), and inflammasome activation and subsequent promotion of insulin resistance following a 120 h treatment period in HepG2 liver in vitro cell model. Furthermore, we assess microRNA (miR-128a) expression as a negative regulator of the IRS1/AKT signaling pathway. The relative expression of phosphorylated IRS1 was determined by Western blot. Transcript levels of NLRP3, IL-1β, JNK, IRS1, AKT, PI3K, and miR-128a were assessed using quantitative PCR (qPCR). Caspase-1 activity was measured using luminometry. Following exposure to ARVs for 120 h, NLRP3 mRNA expression (p = 0.0500) and caspase-1 activity (p < 0.0001) significantly increased. This was followed by a significant elevation in IL-1β in mRNA expression (p = 0.0015). Additionally, JNK expression (p = 0.0093) was upregulated with coinciding increases in p-IRS1 protein expression (p < 0.0001) and decreased IRS1 mRNA expression (p = 0.0004). Consequently, decreased AKT (p = 0.0005) and PI3K expressions (p = 0.0007) were observed. Interestingly miR-128a expression was significantly upregulated. The results indicate that combinational use of ARVs upregulates inflammasome activation and promotes insulin resistance through dysregulation of the IRS1/PI3K/AKT insulin signaling pathway.
Collapse
|
20
|
Banerjee S, Tiwari A, Kar A, Chanda J, Biswas S, Ulrich-Merzenich G, Mukherjee PK. Combining LC-MS/MS profiles with network pharmacology to predict molecular mechanisms of the hyperlipidemic activity of Lagenaria siceraria stand. JOURNAL OF ETHNOPHARMACOLOGY 2023; 300:115633. [PMID: 36031104 DOI: 10.1016/j.jep.2022.115633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 07/24/2022] [Accepted: 08/06/2022] [Indexed: 06/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Lagenaria siceraria Stand. (Family: Cucurbitaceae), popularly known as bottle gourd, is traditionally used in Ayurvedic medicine as a food plant, especially in hypertension and obesity. AIM OF THE STUDY Investigations were undertaken to assign novel lead combinations from this common food plant to multi-molecular modes of actions in the complex disease networks of obesity and hypertension. LC-MS/MS based metabolite screening, in-vivo high fat diet induced hyperlipidemia animal study and network pharmacology explorations of the mechanism of action for lipid lowering effects including a neighbourhood community approach for molecular combinations were performed. MATERIAL AND METHODS Major chemical constituents of the fruits of LS (LSFE) were analysed by HPLC-DAD-MS/MS-QTOF. Wistar albino rats (n = 36), divided into 6 groups (n = 6) received either no treatment or a high-fat diet along with LSFE or Atorvastatin. Lipid profiles and biochemical parameters were evaluated. In silico cross-validated network analyses using different databases and Cytospace were applied. RESULTS Profiling of LSFE revealed 18 major constituents: phenolic acids like p-Coumaric acid and Ferulic acid, the monolignolconferyl alcohol, the flavonoid glycosides hesperidin and apigenin-7-glucoside. Hyperlipidemic animals treated with LSFE (200 mg/kg, 400 mg/kg, 600 mg/kg) showed a significant improvement of their lipid profiles after 30 days of treatment. Network pharmacology analyses for the major 18 compounds revealed enrichment of the insulin and the ErbB signalling pathway. Novel target node combinations (e.g. AKR1C1, AGXT) including their connection to different pathways were identified in silico. CONCLUSIONS The combined in vivo and bioinformatics analyses propose that lead compounds of LSFE act in combination on relevant targets of hyperlipidemia. Perturbations of the IRS→Akt→Foxo1 cascade are predicted which suggest further clinical investigation towards development of safe natural alternative to manage hyperlipidemia.
Collapse
Affiliation(s)
- Subhadip Banerjee
- School of Natural Product Studies, Dept. of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India.
| | - Amrendra Tiwari
- School of Natural Product Studies, Dept. of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India.
| | - Amit Kar
- Institute of Bioresources and Sustainable Development, Department of Biotechnology, Government of India, Takyelpat, Imphal, 795001, India.
| | - Joydeb Chanda
- School of Natural Product Studies, Dept. of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India.
| | - Sayan Biswas
- Institute of Bioresources and Sustainable Development, Department of Biotechnology, Government of India, Takyelpat, Imphal, 795001, India.
| | - Gudrun Ulrich-Merzenich
- University Hospital Bonn (UKB), Medical Clinic III, AG Synergy Research and Experimental, Medicine, D 53127, Bonn, Germany.
| | - Pulok K Mukherjee
- School of Natural Product Studies, Dept. of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India; Institute of Bioresources and Sustainable Development, Department of Biotechnology, Government of India, Takyelpat, Imphal, 795001, India.
| |
Collapse
|
21
|
Hao Y, Lü S, Li W, Long M, Cui Y. Biphasic flow dynamics and polarized mass transportation in branched hepatic sinusoids. BIOMICROFLUIDICS 2022; 16:054110. [PMID: 36313188 PMCID: PMC9616607 DOI: 10.1063/5.0100911] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 09/28/2022] [Indexed: 06/16/2023]
Abstract
In fatty liver diseases, such as liver fibrosis and liver cirrhosis, blood flow in hepatic sinusoids, an elementary building block of the liver lobule, tends to bypass through collateral vessels inside sinusoids and presents distinct sinusoidal flows compared to normal physiological flows. It remains unclear in those flow characteristics in branched sinusoids and the correlation of pathological flows with liver lesions, mainly due to the difficulty of direct hemodynamics measurements in the sinusoids. Here, we developed a dual-branched theoretical model of hepatic sinusoidal flow to elucidate the relevant flow dynamics and mass transport. Numerical simulations, based on the lattice Boltzmann method, indicated that the flow velocity distribution in hepatic sinusoids is mainly dominated by endothelium permeability and presents a non-monotonic variation with the permeability at the fusion segment of these branched sinusoids. Flow-induced shear stress on the endothelium at the side of the Disse space exhibited a biphasic pattern, yielding a low shear stress region at the junctional site. Meanwhile, a highly polarized distribution of lipoproteins concentration was also presented at the low shear stress region, indicating a localized accumulation of typical hepatic serum proteins. Thus, this work provides the basic understanding of blood flow features and mass transport regulations in branched hepatic sinusoids.
Collapse
Affiliation(s)
- Yinjing Hao
- Department of Mechanics, Tianjin University, Tianjin 300072, China
| | | | | | - Mian Long
- Authors to whom correspondence should be addressed:, Tel.: +86 10 8254 4131, Fax:+86 10 8254 4131 and , Tel.: +86 22 27404934, Fax:+86 22 27404934
| | - Yuhong Cui
- Department of Mechanics, Tianjin University, Tianjin 300072, China
| |
Collapse
|
22
|
Bathish B, Robertson H, Dillon JF, Dinkova-Kostova AT, Hayes JD. Nonalcoholic steatohepatitis and mechanisms by which it is ameliorated by activation of the CNC-bZIP transcription factor Nrf2. Free Radic Biol Med 2022; 188:221-261. [PMID: 35728768 DOI: 10.1016/j.freeradbiomed.2022.06.226] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 06/13/2022] [Indexed: 12/11/2022]
Abstract
Non-alcoholic steatohepatitis (NASH) represents a global health concern. It is characterised by fatty liver, hepatocyte cell death and inflammation, which are associated with lipotoxicity, endoplasmic reticulum (ER) stress, mitochondrial dysfunction, iron overload and oxidative stress. NF-E2 p45-related factor 2 (Nrf2) is a transcription factor that combats oxidative stress. Remarkably, Nrf2 is downregulated during the development of NASH, which probably accelerates disease, whereas in pre-clinical studies the upregulation of Nrf2 inhibits NASH. We now review the scientific literature that proposes Nrf2 downregulation during NASH involves its increased ubiquitylation and proteasomal degradation, mediated by Kelch-like ECH-associated protein 1 (Keap1) and/or β-transducin repeat-containing protein (β-TrCP) and/or HMG-CoA reductase degradation protein 1 (Hrd1, also called synoviolin (SYVN1)). Additionally, downregulation of Nrf2-mediated transcription during NASH may involve diminished recruitment of coactivators by Nrf2, due to increased levels of activating transcription factor 3 (ATF3) and nuclear factor-kappaB (NF-κB) p65, or competition for promoter binding due to upregulation of BTB and CNC homology 1 (Bach1). Many processes that downregulate Nrf2 are triggered by transforming growth factor-beta (TGF-β), with oxidative stress amplifying its signalling. Oxidative stress may also increase suppression of Nrf2 by β-TrCP through facilitating formation of the DSGIS-containing phosphodegron in Nrf2 by glycogen synthase kinase-3. In animal models, knockout of Nrf2 increases susceptibility to NASH, while pharmacological activation of Nrf2 by inducing agents that target Keap1 inhibits development of NASH. These inducing agents probably counter Nrf2 downregulation affected by β-TrCP, Hrd1/SYVN1, ATF3, NF-κB p65 and Bach1, by suppressing oxidative stress. Activation of Nrf2 is also likely to inhibit NASH by ameliorating lipotoxicity, inflammation, ER stress and iron overload. Crucially, pharmacological activation of Nrf2 in mice in which NASH has already been established supresses liver steatosis and inflammation. There is therefore compelling evidence that pharmacological activation of Nrf2 provides a comprehensive multipronged strategy to treat NASH.
Collapse
Affiliation(s)
- Boushra Bathish
- Jacqui Wood Cancer Centre, Division of Cellular Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - Holly Robertson
- Jacqui Wood Cancer Centre, Division of Cellular Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, UK; Wellcome Trust Sanger Institute, Wellcome Genome Campus, Cambridge, CB10 1SA, UK
| | - John F Dillon
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, UK
| | - Albena T Dinkova-Kostova
- Jacqui Wood Cancer Centre, Division of Cellular Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - John D Hayes
- Jacqui Wood Cancer Centre, Division of Cellular Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, UK.
| |
Collapse
|
23
|
Sabir U, Irfan HM, Alamgeer, Umer I, Niazi ZR, Asjad HMM. Phytochemicals targeting NAFLD through modulating the dual function of forkhead box O1 (FOXO1) transcription factor signaling pathways. Naunyn Schmiedebergs Arch Pharmacol 2022; 395:741-755. [PMID: 35357518 DOI: 10.1007/s00210-022-02234-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 03/18/2022] [Indexed: 02/06/2023]
Abstract
Literature evidence reveals that natural compounds are potential candidates for ameliorating obesity-associated non-alcoholic fatty liver disease (NAFLD) by targeting forkhead box O1 (FOXO1) transcription factor. FOXO1 has a dual and complex role in regulating both increase and decrease in lipid accumulation in hepatocytes and adipose tissues (AT) at different stages of NAFLD. In insulin resistance (IR), it is constitutively expressed, resulting in increased hepatic glucose output and lipid metabolism irregularity. The studies on different phytochemicals indicate that dysregulation of FOXO1 causes disturbance in cellular nutrients homeostasis, and the natural entities have an enduring impact on the mitigation of these abnormalities. The current review communicates and evaluates certain phytochemicals through different search engines, targeting FOXO1 and its downstream cellular pathways to find lead compounds as potential therapeutic agents for treating NAFLD and related metabolic disorders. The findings of this review confirm that polyphenols, flavonoids, alkaloids, terpenoids, and anthocyanins are capable of modulating FOXO1 and associated signaling pathways, and they are potential therapeutic agents for NAFLD and related complications. HIGHLIGHTS: • FOXO1 has the potential to be targeted by novel drugs from natural sources for the treatment of NAFLD and obesity. • FOXO1 regulates cellular autophagy, inflammation, oxidative stress, and lipogenesis through alternative mechanisms. • Phytochemicals treat NAFLD by acting on FOXO1 or SREBP1c and PPARγ transcription factor signaling pathways.
Collapse
Affiliation(s)
- Usman Sabir
- Department of Pharmacology, College of Pharmacy, University of Sargodha, Sargodha, Pakistan
| | - Hafiz Muhammad Irfan
- Department of Pharmacology, College of Pharmacy, University of Sargodha, Sargodha, Pakistan.
| | - Alamgeer
- Punjab University College of Pharmacy, University of the Punjab Lahore, Lahore, Pakistan
| | - Ihtisham Umer
- Pharmacy Department, Comsat International University Lahore Campus, Lahore, Pakistan
| | | | | |
Collapse
|
24
|
Liu L, Chen C, Dong Y, Cheng Y, You C, Wang S, Ma H, Li Y. Insulin activates LC-PUFA biosynthesis of hepatocytes by regulating the PI3K/Akt/mTOR/Srebp1 pathway in teleost Siganus canaliculatus. Comp Biochem Physiol B Biochem Mol Biol 2022; 260:110734. [PMID: 35321854 DOI: 10.1016/j.cbpb.2022.110734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 03/02/2022] [Accepted: 03/17/2022] [Indexed: 11/30/2022]
Abstract
Insulin is well known an important metabolic regulator in glucose and lipid metabolism. It has been proved to activate long-chain (≥ C20) polyunsaturated fatty acids (LC-PUFA) biosynthesis in mammals, but little is known about such a role in fish. To explore the effects and molecular mechanisms of insulin in fish LC-PUFA biosynthesis, we treated the rabbitfish S. canaliculatus hepatocyte line (SCHL) cells with 65 nM insulin for 12 h, and the results showed that the mRNA levels of genes encoding the key enzymes and transcription factor involved in rabbitfish LC-PUFA biosynthesis such as Δ6Δ5 fads2, elovl5 and srebp1, as well as those of PI3K pathway genes including pdk1, akt2 and mtor increased significantly. Moreover, SCHL cells treated with different PI3K/Akt pathway inhibitors (LY294002, Wortmannin, AKTi-1/2) alone or combined with insulin decreased the mRNA levels of PI3K/Akt/mTOR downstream signaling genes, including Δ6Δ5 fads2, Δ4 fads2, elovl5, elovl4 and srebp1. While PI3K/Akt agonists (740 Y-P, IGF-1, SC-79) had the opposite results. The results of fatty acid composition analysis of hepatocytes showed that insulin stimulation increased the Δ6Δ5 Fads2-dependent PUFA desaturation indexes, while Elovl5-dependent PUFA elongation indexes had upward trends, and consequently LC-PUFA contents increased. Taken together, these results indicated that insulin activated LC-PUFA biosynthesis probably through PI3K/Akt/mTOR/Srebp1 pathway in S. canaliculatus hepatocytes.
Collapse
Affiliation(s)
- Lijie Liu
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou 515063, China
| | - Cuiying Chen
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou 515063, China
| | - Yewei Dong
- College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong 510225, China
| | - Yu Cheng
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou 515063, China
| | - Cuihong You
- College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong 510225, China
| | - Shuqi Wang
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou 515063, China
| | - Hongyu Ma
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou 515063, China.
| | - Yuanyou Li
- School of Marine Sciences, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
25
|
Xie W, Liu B, Tang Y, Yang T, Song Z. Gamma-glutamyl transferase to high-density lipoprotein cholesterol ratio: A valuable predictor of type 2 diabetes mellitus incidence. Front Endocrinol (Lausanne) 2022; 13:1026791. [PMID: 36246883 PMCID: PMC9557082 DOI: 10.3389/fendo.2022.1026791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 09/15/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Gamma-glutamyl transferase (GGT) and high-density lipoprotein cholesterol (HDL-C) have been proven to be valuable predictors of type 2 diabetes mellitus (T2DM). The aim of this study was to investigate the association between GGT/HDL-C ratio and incident T2DM. METHODS The study retrospectively analyzed 15453 participants from 2004 to 2015. Cox proportional hazards regression models and Kaplan-Meier curves were used to elucidate the effect of GGT/HDL-C ratio on T2DM. Restricted cubic spline (RCS) analysis was performed to explore any non-linear correlation between GGT/HDL-C ratio and the risk of T2DM. The predictive performance of GGT, HDL-C and GGT/HDL-C ratio for T2DM was evaluated utilizing receiver-operating-characteristic (ROC) curves. RESULTS During a median follow-up of 5.39 years, 373 cases of incident T2DM were observed. Kaplan-Meier curves showed that the cumulative probabilities of T2DM increased in the participants with higher GGT/HDL-C ratio significantly (P < 0.001). Cox models further clarified that high GGT/HDL-C ratio was an independent risk factor for T2DM (HR = 1.01, 95% CI = 1.00-1.01, P = 0.011). Linear positive correlation between GGT/HDL-C ratio and the risk of T2DM was demonstrated through RCS analysis. In the ROC analysis, GGT/HDL-C ratio (AUC = 0.75, 95% CI = 0.73-0.77) showed competitive role in the prediction of T2DM compared with single GGT and HDL-C. CONCLUSIONS The GGT/HDL-C ratio could serve as a valuable predictor of T2DM, and the risk of T2DM increases in the condition of higher GGT/HDL-C ratio.
Collapse
Affiliation(s)
- Wangcheng Xie
- Department of General Surgery, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Bin Liu
- Department of Gastrointestinal Surgery, Anqing First People’s Hospital, Anhui Medical University, Anqing, China
| | - Yansong Tang
- Department of Cardiology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Tingsong Yang
- Department of General Surgery, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
- *Correspondence: Zhenshun Song, ; Tingsong Yang,
| | - Zhenshun Song
- Department of General Surgery, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
- *Correspondence: Zhenshun Song, ; Tingsong Yang,
| |
Collapse
|
26
|
Song X, Dong H, Zang Z, Wu W, Zhu W, Zhang H, Guan Y. Kudzu Resistant Starch: An Effective Regulator of Type 2 Diabetes Mellitus. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:4448048. [PMID: 34691353 PMCID: PMC8528595 DOI: 10.1155/2021/4448048] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/30/2021] [Accepted: 09/16/2021] [Indexed: 12/23/2022]
Abstract
Kudzu is a traditional medicinal dietary supplement, and recent research has shown its significant benefits in the prevention/treatment of type 2 diabetes mellitus (T2DM). Starch is one of the main substances in Kudzu that contribute decisively to the treatment of T2DM. However, the underlying mechanism of the hypoglycemic activity is not clear. In this study, the effect of Kudzu resistant starch supplementation on the insulin resistance, gut physical barrier, and gut microbiota was investigated in T2DM mice. The result showed that Kudzu resistant starch could significantly decrease the value of fasting blood glucose and the levels of total cholesterol, total triglyceride, and high-density lipoprotein, as well as low-density lipoprotein, in the blood of T2DM mice. The insulin signaling sensitivity in liver tissue was analyzed; the result indicated that intake of different doses of Kudzu resistant starch can help restore the expression of IRS-1, p-PI3K, p-Akt, and Glut4 and thus enhance the efficiency of insulin synthesis. Furthermore, the intestinal microorganism changes before and after ingestion of Kudzu resistant starch were also analyzed; the result revealed that supplementation of KRS helps to alleviate and improve the dysbiosis of the gut microbiota caused by T2DM. These results validated that Kudzu resistant starch could improve the glucose sensitivity of T2DM mice by modulating IRS-1/PI3K/AKT/Glut4 signaling transduction. Kudzu resistant starch can be used as a promising prebiotic, and it also has beneficial effects on the gut microbiota structure of T2DM mice.
Collapse
Affiliation(s)
- Xinqi Song
- Key Laboratory of Modern Preparation of Traditional Chinese Medicines, Ministry of Education, Jiangxi University of Chinese Medicine, 330004 Nanchang, China
| | - Huanhuan Dong
- School of Pharmacy, Jiangxi University of Chinese Medicine, 330004 Nanchang, China
| | - Zhenzhong Zang
- Key Laboratory of Modern Preparation of Traditional Chinese Medicines, Ministry of Education, Jiangxi University of Chinese Medicine, 330004 Nanchang, China
| | - Wenting Wu
- School of Pharmacy, Jiangxi University of Chinese Medicine, 330004 Nanchang, China
| | - Weifeng Zhu
- Key Laboratory of Modern Preparation of Traditional Chinese Medicines, Ministry of Education, Jiangxi University of Chinese Medicine, 330004 Nanchang, China
| | - Hua Zhang
- School of Pharmacy, Jiangxi University of Chinese Medicine, 330004 Nanchang, China
| | - Yongmei Guan
- Key Laboratory of Modern Preparation of Traditional Chinese Medicines, Ministry of Education, Jiangxi University of Chinese Medicine, 330004 Nanchang, China
| |
Collapse
|
27
|
Abstract
The molecular mechanisms of cellular insulin action have been the focus of much investigation since the discovery of the hormone 100 years ago. Insulin action is impaired in metabolic syndrome, a condition known as insulin resistance. The actions of the hormone are initiated by binding to its receptor on the surface of target cells. The receptor is an α2β2 heterodimer that binds to insulin with high affinity, resulting in the activation of its tyrosine kinase activity. Once activated, the receptor can phosphorylate a number of intracellular substrates that initiate discrete signaling pathways. The tyrosine phosphorylation of some substrates activates phosphatidylinositol-3-kinase (PI3K), which produces polyphosphoinositides that interact with protein kinases, leading to activation of the kinase Akt. Phosphorylation of Shc leads to activation of the Ras/MAP kinase pathway. Phosphorylation of SH2B2 and of Cbl initiates activation of G proteins such as TC10. Activation of Akt and other protein kinases produces phosphorylation of a variety of substrates, including transcription factors, GTPase-activating proteins, and other kinases that control key metabolic events. Among the cellular processes controlled by insulin are vesicle trafficking, activities of metabolic enzymes, transcriptional factors, and degradation of insulin itself. Together these complex processes are coordinated to ensure glucose homeostasis.
Collapse
|
28
|
Ren X, Wang L, Chen Z, Hou D, Xue Y, Diao X, Shen Q. Foxtail Millet Improves Blood Glucose Metabolism in Diabetic Rats through PI3K/AKT and NF-κB Signaling Pathways Mediated by Gut Microbiota. Nutrients 2021; 13:nu13061837. [PMID: 34072141 PMCID: PMC8228963 DOI: 10.3390/nu13061837] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/22/2021] [Accepted: 05/25/2021] [Indexed: 12/11/2022] Open
Abstract
Foxtail millet (FM) is receiving ongoing increased attention due to its beneficial health effects, including the hypoglycemic effect. However, the underlying mechanisms of the hypoglycemic effect have been underexplored. In the present study, the hypoglycemic effect of FM supplementation was confirmed again in high-fat diet and streptozotocin-induced diabetic rats with significantly decreased fasting glucose (FG), glycated serum protein, and areas under the glucose tolerance test (p < 0.05). We employed 16S rRNA and liver RNA sequencing technologies to identify the target gut microbes and signaling pathways involved in the hypoglycemic effect of FM supplementation. The results showed that FM supplementation significantly increased the relative abundance of Lactobacillus and Ruminococcus_2, which were significantly negatively correlated with FG and 2-h glucose. FM supplementation significantly reversed the trends of gene expression in diabetic rats. Specifically, FM supplementation inhibited gluconeogenesis, stimulated glycolysis, and restored fatty acid synthesis through activation of the PI3K/AKT signaling pathway. FM also reduced inflammation through inhibition of the NF-κB signaling pathway. Spearman’s correlation analysis indicated a complicated set of interdependencies among the gut microbiota, signaling pathways, and metabolic parameters. Collectively, the above results suggest that the hypoglycemic effect of FM was at least partially mediated by the increased relative abundance of Lactobacillus, activation of the PI3K/AKT signaling pathway, and inhibition of the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Xin Ren
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University, Beijing 100048, China; (X.R.); (L.W.)
- Key Laboratory of Plant Protein and Grain Processing, National Engineering and Technology Research Center for Fruits and Vegetables, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (D.H.); (Y.X.)
| | - Linxuan Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University, Beijing 100048, China; (X.R.); (L.W.)
| | - Zenglong Chen
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China;
| | - Dianzhi Hou
- Key Laboratory of Plant Protein and Grain Processing, National Engineering and Technology Research Center for Fruits and Vegetables, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (D.H.); (Y.X.)
| | - Yong Xue
- Key Laboratory of Plant Protein and Grain Processing, National Engineering and Technology Research Center for Fruits and Vegetables, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (D.H.); (Y.X.)
| | - Xianmin Diao
- Center for Crop Germplasm Resources, Institute of Crop Science, Chinese Academy of Agricultural Sciences, Beijing 100081, China;
| | - Qun Shen
- Key Laboratory of Plant Protein and Grain Processing, National Engineering and Technology Research Center for Fruits and Vegetables, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (D.H.); (Y.X.)
- Correspondence: ; Tel.: +86-10-62737524
| |
Collapse
|
29
|
Remchak MME, Piersol KL, Bhatti S, Spaeth AM, Buckman JF, Malin SK. Considerations for Maximizing the Exercise "Drug" to Combat Insulin Resistance: Role of Nutrition, Sleep, and Alcohol. Nutrients 2021; 13:1708. [PMID: 34069950 PMCID: PMC8157556 DOI: 10.3390/nu13051708] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/24/2021] [Accepted: 05/13/2021] [Indexed: 01/12/2023] Open
Abstract
Insulin resistance is a key etiological factor in promoting not only type 2 diabetes mellitus but also cardiovascular disease (CVD). Exercise is a first-line therapy for combating chronic disease by improving insulin action through, in part, reducing hepatic glucose production and lipolysis as well as increasing skeletal muscle glucose uptake and vasodilation. Just like a pharmaceutical agent, exercise can be viewed as a "drug" such that identifying an optimal prescription requires a determination of mode, intensity, and timing as well as consideration of how much exercise is done relative to sitting for prolonged periods (e.g., desk job at work). Furthermore, proximal nutrition (nutrient timing, carbohydrate intake, etc.), sleep (or lack thereof), as well as alcohol consumption are likely important considerations for enhancing adaptations to exercise. Thus, identifying the maximal exercise "drug" for reducing insulin resistance will require a multi-health behavior approach to optimize type 2 diabetes and CVD care.
Collapse
Affiliation(s)
- Mary-Margaret E. Remchak
- Department of Kinesiology & Health, Rutgers University, New Brunswick, NJ 08901, USA; (M.-M.E.R.); (K.L.P.); (A.M.S.); (J.F.B.)
| | - Kelsey L. Piersol
- Department of Kinesiology & Health, Rutgers University, New Brunswick, NJ 08901, USA; (M.-M.E.R.); (K.L.P.); (A.M.S.); (J.F.B.)
| | - Sabha Bhatti
- Division of Cardiovascular Medicine, Rutgers University, New Brunswick, NJ 08901, USA;
| | - Andrea M. Spaeth
- Department of Kinesiology & Health, Rutgers University, New Brunswick, NJ 08901, USA; (M.-M.E.R.); (K.L.P.); (A.M.S.); (J.F.B.)
| | - Jennifer F. Buckman
- Department of Kinesiology & Health, Rutgers University, New Brunswick, NJ 08901, USA; (M.-M.E.R.); (K.L.P.); (A.M.S.); (J.F.B.)
- Center of Alcohol Studies, Rutgers University, Piscataway, NJ 08854, USA
| | - Steven K. Malin
- Department of Kinesiology & Health, Rutgers University, New Brunswick, NJ 08901, USA; (M.-M.E.R.); (K.L.P.); (A.M.S.); (J.F.B.)
- Division of Endocrinology, Metabolism & Nutrition, Rutgers University, New Brunswick, NJ 08901, USA
- New Jersey Institute for Food, Nutrition and Health, Rutgers University, New Brunswick, NJ 08901, USA
- Institute of Translational Medicine and Science, Rutgers University, New Brunswick, NJ 08901, USA
| |
Collapse
|
30
|
Luo P, Zheng M, Zhang R, Zhang H, Liu Y, Li W, Sun X, Yu Q, Tipoe GL, Xiao J. S-Allylmercaptocysteine improves alcoholic liver disease partly through a direct modulation of insulin receptor signaling. Acta Pharm Sin B 2021; 11:668-679. [PMID: 33777674 PMCID: PMC7982498 DOI: 10.1016/j.apsb.2020.11.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/31/2020] [Accepted: 09/07/2020] [Indexed: 12/18/2022] Open
Abstract
Alcoholic liver disease (ALD) causes insulin resistance, lipid metabolism dysfunction, and inflammation. We investigated the protective effects and direct regulating target of S-allylmercaptocysteine (SAMC) from aged garlic on liver cell injury. A chronic ethanol-fed ALD in vivo model (the NIAAA model) was used to test the protective functions of SAMC. It was observed that SAMC (300 mg/kg, by gavage method) effectively ameliorated ALD-induced body weight reduction, steatosis, insulin resistance, and inflammation without affecting the health status of the control mice, as demonstrated by histological, biochemical, and molecular biology assays. By using biophysical assays and molecular docking, we demonstrated that SAMC directly targeted insulin receptor (INSR) protein on the cell membrane and then restored downstream IRS-1/AKT/GSK3β signaling. Liver-specific knock-down in mice and siRNA-mediated knock-down in AML-12 cells of Insr significantly impaired SAMC (250 μmol/L in cells)-mediated protection. Restoration of the IRS-1/AKT signaling partly recovered hepatic injury and further contributed to SAMC's beneficial effects. Continuous administration of AKT agonist and recombinant IGF-1 in combination with SAMC showed hepato-protection in the mice model. Long-term (90-day) administration of SAMC had no obvious adverse effect on healthy mice. We conclude that SAMC is an effective and safe hepato-protective complimentary agent against ALD partly through the direct binding of INSR and partial regulation of the IRS-1/AKT/GSK3β pathway.
Collapse
Key Words
- ADIPOQ, adiponectin
- AKT
- ALD, alcoholic liver disease
- ALDH2, aldehyde dehydrogenase 2
- ALT, alanine aminotransferase
- AMPK, adenosine 5′-monophosphate (AMP)-activated protein kinase
- AST, aspartate aminotransferase
- ATGL, adipose triglyceride lipase
- Alcoholic liver disease
- CPT1, carnitine palmitoyltransferase I
- CYP2E1, cytochrome P450 2E1
- FDA, U.S. Food and Drug Administration
- FFA, free fatty acids
- GRB14, growth factor receptor-bound protein 14
- GSK3β
- GSK3β, glycogen synthase kinase 3 beta
- GTT, glucose tolerance test
- HSL, hormone sensitive lipase
- IGF-1, insulin-like growth factors-1
- IL, interleukin
- INSR, insulin receptor
- IRS, insulin receptor substrate
- IRS-1
- IRTK, insulin receptor tyrosine kinase
- Insulin receptor
- Insulin resistance
- LDLR, low-density lipoprotein receptor
- LRP6, low-density lipoprotein receptor related protein 6
- MTT, 3-(4,5-dimethyl-thiazol-2-yl)-2,5-diphenyl-tetrazolium bromide
- NAC, N-acetyl-cysteine
- NAFLD, non-alcoholic fatty liver disease
- NAS, NAFLD activity score
- NF-κB, nuclear factor kappa B
- NIAAA, National Institute on Alcohol Abuse and Alcoholism
- NRF2, nuclear factor erythroid 2-related factor 2
- ORF, open reading frame
- PA, palmitate acid
- PPARα, peroxisome proliferator-activated receptor alpha
- RER, respiratory exchange ratio
- S-Allylmercaptocysteine
- SAMC, S-allylmercaptocysteine
- SPR, surface plasmon resonance
- SREBP-1c, sterol regulatory element-binding protein 1c
- Safety
- TC, total cholesterol
- TCF/LEF, T-cell factor/lymphoid enhancer factor
- TG, triglyceride
- TNF, tumor necrosis factor
- TSA, thermal shift assay
- WAT, white adipose tissues
- WT, wild-type
Collapse
|
31
|
Du F, Huang R, Lin D, Wang Y, Yang X, Huang X, Zheng B, Chen Z, Huang Y, Wang X, Chen F. Resveratrol Improves Liver Steatosis and Insulin Resistance in Non-alcoholic Fatty Liver Disease in Association With the Gut Microbiota. Front Microbiol 2021; 12:611323. [PMID: 33708180 PMCID: PMC7942199 DOI: 10.3389/fmicb.2021.611323] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 01/11/2021] [Indexed: 12/12/2022] Open
Abstract
Resveratrol (RSV) is a potential alternative therapy for non-alcoholic fatty liver disease (NAFLD) that has been evaluated in many clinical trials, but the mechanisms of RSV action have not been fully elucidated. Recent studies suggested that the gut microbiota is an important RSV target; therefore, we speculated that the gut microbiota might mediate the beneficial effects of RSV in NAFLD. To verify this hypothesis, we established a high-fat diet (HFD)-induced NAFLD mouse model, which was subjected to RSV gavage to evaluate the therapeutic effects. We observed that RSV reduced liver steatosis and insulin resistance in NAFLD. RSV significantly changed the diversity and composition of the gut microbiota according to 16S rRNA sequencing. Gut microbiota gene function prediction showed that the enrichment of pathways related to lipid and glucose metabolism decreased after RSV treatment. Furthermore, correlation analysis indicated that the improvements in NAFLD metabolic indicators were closely related to the altered gut microbiota. We further fermented RSV with the gut microbiota in vitro to verify that RSV directly affected the gut microbiota. Our data suggested that the gut microbiota might be an important target through which RSV exerts its anti-NAFLD effect.
Collapse
Affiliation(s)
- Fan Du
- Department of Gastroenterology, Fujian Medical University Union Hospital, Fuzhou, China.,Fujian Medical University Cancer Center, Fujian Medical University, Fuzhou, China
| | - Rongfeng Huang
- Department of Gastroenterology, Fujian Medical University Union Hospital, Fuzhou, China.,Fujian Medical University Cancer Center, Fujian Medical University, Fuzhou, China
| | - Dan Lin
- Department of Gastroenterology, Fujian Medical University Union Hospital, Fuzhou, China.,Fujian Medical University Cancer Center, Fujian Medical University, Fuzhou, China
| | - Yuying Wang
- Department of Gastroenterology, Fujian Medical University Union Hospital, Fuzhou, China.,Fujian Medical University Cancer Center, Fujian Medical University, Fuzhou, China
| | - Xiaohuang Yang
- Department of Gastroenterology, Fujian Medical University Union Hospital, Fuzhou, China.,Fujian Medical University Cancer Center, Fujian Medical University, Fuzhou, China
| | - Xiaoyun Huang
- Department of Gastroenterology, Fujian Medical University Union Hospital, Fuzhou, China.,Fujian Medical University Cancer Center, Fujian Medical University, Fuzhou, China
| | - Biyun Zheng
- Department of Gastroenterology, Fujian Medical University Union Hospital, Fuzhou, China.,Fujian Medical University Cancer Center, Fujian Medical University, Fuzhou, China
| | - Zhixin Chen
- Department of Gastroenterology, Fujian Medical University Union Hospital, Fuzhou, China.,Fujian Medical University Cancer Center, Fujian Medical University, Fuzhou, China
| | - Yuehong Huang
- Department of Gastroenterology, Fujian Medical University Union Hospital, Fuzhou, China.,Fujian Medical University Cancer Center, Fujian Medical University, Fuzhou, China
| | - Xiaozhong Wang
- Department of Gastroenterology, Fujian Medical University Union Hospital, Fuzhou, China.,Fujian Medical University Cancer Center, Fujian Medical University, Fuzhou, China
| | - Fenglin Chen
- Department of Gastroenterology, Fujian Medical University Union Hospital, Fuzhou, China.,Fujian Medical University Cancer Center, Fujian Medical University, Fuzhou, China
| |
Collapse
|
32
|
Sekizkardes H, Chung ST, Chacko S, Haymond MW, Startzell M, Walter M, Walter PJ, Lightbourne M, Brown RJ. Free fatty acid processing diverges in human pathologic insulin resistance conditions. J Clin Invest 2021; 130:3592-3602. [PMID: 32191645 DOI: 10.1172/jci135431] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 03/17/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUNDPostreceptor insulin resistance (IR) is associated with hyperglycemia and hepatic steatosis. However, receptor-level IR (e.g., insulin receptor pathogenic variants, INSR) causes hyperglycemia without steatosis. We examined 4 pathologic conditions of IR in humans to examine pathways controlling lipid metabolism and gluconeogenesis.METHODSCross-sectional study of severe receptor IR (INSR, n = 7) versus postreceptor IR that was severe (lipodystrophy, n = 14), moderate (type 2 diabetes, n = 9), or mild (obesity, n = 8). Lipolysis (glycerol turnover), hepatic glucose production (HGP), gluconeogenesis (deuterium incorporation from body water into glucose), hepatic triglyceride (magnetic resonance spectroscopy), and hepatic fat oxidation (plasma β-hydroxybutyrate) were measured.RESULTSLipolysis was 2- to 3-fold higher in INSR versus all other groups, and HGP was 2-fold higher in INSR and lipodystrophy versus type 2 diabetes and obesity (P < 0.001), suggesting severe adipose and hepatic IR. INSR subjects had a higher contribution of gluconeogenesis to HGP, approximately 77%, versus 52% to 59% in other groups (P = 0.0001). Despite high lipolysis, INSR subjects had low hepatic triglycerides (0.5% [interquartile range 0.1%-0.5%]), in contrast to lipodystrophy (10.6% [interquartile range 2.8%-17.1%], P < 0.0001). β-hydroxybutyrate was 2- to 7-fold higher in INSR versus all other groups (P < 0.0001), consistent with higher hepatic fat oxidation.CONCLUSIONThese data support a key pathogenic role of adipose tissue IR to increase glycerol and FFA availability to the liver in both receptor and postreceptor IR. However, the fate of FFA diverges in these populations. In receptor-level IR, FFA oxidation drives gluconeogenesis rather than being reesterified to triglyceride. In contrast, in postreceptor IR, FFA contributes to both gluconeogenesis and hepatic steatosis.TRIAL REGISTRATIONClinicalTrials.gov NCT01778556, NCT00001987, and NCT02457897.FUNDINGNational Institute of Diabetes and Digestive and Kidney Diseases, US Department of Agriculture/Agricultural Research Service 58-3092-5-001.
Collapse
Affiliation(s)
| | - Stephanie Therese Chung
- National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Shaji Chacko
- Children's Nutrition Research Center, Department of Pediatrics, US Department of Agriculture/Agricultural Research Service, Baylor College of Medicine, Houston, Texas, USA
| | - Morey W Haymond
- Children's Nutrition Research Center, Department of Pediatrics, US Department of Agriculture/Agricultural Research Service, Baylor College of Medicine, Houston, Texas, USA
| | - Megan Startzell
- National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Mary Walter
- National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Peter J Walter
- National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | | | - Rebecca J Brown
- National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| |
Collapse
|
33
|
Li Y, Liu Y, Yang M, Wang Q, Zheng Y, Xu J, Zheng P, Song H. A Study on the Therapeutic Efficacy of San Zi Yang Qin Decoction for Non-Alcoholic Fatty Liver Disease and the Underlying Mechanism Based on Network Pharmacology. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:8819245. [PMID: 33505505 PMCID: PMC7810527 DOI: 10.1155/2021/8819245] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 12/16/2020] [Accepted: 12/22/2020] [Indexed: 12/03/2022]
Abstract
OBJECTIVE This study aims to explore the therapeutic efficacy of San Zi Yang Qin Decoction (SZ) and its potential mechanism in the treatment of non-alcoholic fatty liver disease (NAFLD) based on network pharmacology and in vivo experiments. METHODS Effective chemicals and targets of SZ were searched in online databases, according to the drug-likeness of compounds and the binomial distribution of targets. A disease-target-chemical network was established using NAFLD-associated genes screened through GeneCards database, Gene Ontology (GO) terms, and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. Furthermore, animal experiments were conducted to verify the efficacy and mechanism of SZ predicted by network pharmacology. The NAFLD mouse model was established with C57BL/6J mice fed with a high-fat diet for 22 weeks. The mice in the control group were fed with a chow diet. From the 23rd week, the NAFLD mice were treated with intragastric SZ or normal saline for 8 weeks. After the glucose tolerance was measured, the mice were sacrificed, followed by the collection of serum and liver tissues. Pathological changes in liver tissues were examined by H&E staining. Additionally, alanine aminotransferase (ALT), aspartate aminotransferase (AST), serum fast blood glucose, and insulin levels were detected. Expression levels of TNF-α of serum and liver tissues were determined by ELISA and qRT-PCR, respectively. Western blot was used to detect the activation of AKT in liver tissues. RESULTS A total of 27 effective compounds and 20 targets of SZ were screened. GO analysis uncovered a significant correlation between the targets of SZ and those of NAFLD. KEGG analysis presented the signaling pathways enriched in SZ and NAFLD, including NAFLD, TNF-α, and apoptosis pathways. The area under the curve of major GO and KEGG pathways indicated the potential role of SZ in improving NAFLD. In vivo experiments demonstrated that SZ significantly alleviated hepatosteatosis and inflammatory cell infiltration in liver tissues, reduced serum transaminases, and improved insulin resistance and glucose tolerance of NAFLD mice. The protein level of phospho-AKT was upregulated by SZ. Additionally, SZ treatment obviously impaired the TNF-α level in the serum and liver tissue of NAFLD mice. CONCLUSIONS According to the network pharmacology analysis and in vivo experiments, SZ could have therapeutic efficacy for NALFD. The mechanism mainly involves pathways relative to insulin resistance, TNF-α, and apoptosis. Our results provide a scientific basis for SZ in the clinical treatment of NAFLD.
Collapse
Affiliation(s)
- Yiping Li
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wanping Road, Shanghai 200032, China
| | - Yang Liu
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wanping Road, Shanghai 200032, China
| | - Ming Yang
- Office of National Drug Clinical Trial, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Qianlei Wang
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wanping Road, Shanghai 200032, China
| | - Yu Zheng
- Department II of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200120, China
| | - Jiaoya Xu
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wanping Road, Shanghai 200032, China
| | - Peiyong Zheng
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wanping Road, Shanghai 200032, China
| | - Haiyan Song
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wanping Road, Shanghai 200032, China
| |
Collapse
|
34
|
Lu Z, Li Y, Syn WK, Li AJ, Ritter WS, Wank SA, Lopes-Virella MF, Huang Y. GPR40 deficiency is associated with hepatic FAT/CD36 upregulation, steatosis, inflammation, and cell injury in C57BL/6 mice. Am J Physiol Endocrinol Metab 2021; 320:E30-E42. [PMID: 33103454 PMCID: PMC8436599 DOI: 10.1152/ajpendo.00257.2020] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 09/21/2020] [Accepted: 10/15/2020] [Indexed: 02/07/2023]
Abstract
G-protein-coupled receptor 40 (GPR40) is highly expressed in pancreatic islets, and its activation increases glucose-stimulated insulin secretion from pancreas. Therefore, GPR40 is considered as a target for type 2 diabetes mellitus (T2DM). Since nonalcoholic fatty liver disease (NAFLD) is associated with T2DM and GPR40 is also expressed by hepatocytes and macrophages, it is important to understand the role of GPR40 in NAFLD. However, the role of GPR40 in NAFLD in animal models has not been well defined. In this study, we fed wild-type or GPR40 knockout C57BL/6 mice a high-fat diet (HFD) for 20 wk and then assessed the effect of GPR40 deficiency on HFD-induced NAFLD. Assays on metabolic parameters showed that an HFD increased body weight, glucose, insulin, insulin resistance, cholesterol, and alanine aminotransferase (ALT), and GPR40 deficiency did not mitigate the HFD-induced metabolic abnormalities. In contrast, we found that GPR40 deficiency was associated with increased body weight, insulin, insulin resistance, cholesterol, and ALT in control mice fed a low-fat diet (LFD). Surprisingly, histology and Oil Red O staining showed that GPR40 deficiency in LFD-fed mice was associated with steatosis. Immunohistochemical analysis showed that GPR40 deficiency also increased F4/80, a macrophage biomarker, in LFD-fed mice. Furthermore, results showed that GPR40 deficiency led to a robust upregulation of hepatic fatty acid translocase (FAT)/CD36 expression. Finally, our in vitro studies showed that GPR40 knockdown by siRNA or a GPR40 antagonist increased palmitic acid-induced FAT/CD36 mRNA in hepatocytes. Taken together, this study indicates that GPR40 plays an important role in homeostasis of hepatic metabolism and inflammation and inhibits nonalcoholic steatohepatitis by possible modulation of FAT/CD36 expression.
Collapse
Affiliation(s)
- Zhongyang Lu
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| | - Yanchun Li
- Division of Endocrinology, Diabetes, and Medical Genetics, Medical University of South Carolina, Charleston, South Carolina
| | - Wing-Kin Syn
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
- Division of Gastroenterology and Hepatology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country, Euskal Herriko Unibertsitatea/Universidad del País Vasco, Leioa, Spain
| | - Ai-Jun Li
- Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| | - W Sue Ritter
- Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| | - Stephen A Wank
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland
| | - Maria F Lopes-Virella
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
- Division of Endocrinology, Diabetes, and Medical Genetics, Medical University of South Carolina, Charleston, South Carolina
| | - Yan Huang
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
- Division of Endocrinology, Diabetes, and Medical Genetics, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
35
|
Gao ST, Girma DD, Bionaz M, Ma L, Bu DP. Hepatic transcriptomic adaptation from prepartum to postpartum in dairy cows. J Dairy Sci 2020; 104:1053-1072. [PMID: 33189277 DOI: 10.3168/jds.2020-19101] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 08/31/2020] [Indexed: 12/25/2022]
Abstract
The transition from pregnancy to lactation is the most challenging period for high-producing dairy cows. The liver plays a key role in biological adaptation during the peripartum. Prior works have demonstrated that hepatic glucose synthesis, cholesterol metabolism, lipogenesis, and inflammatory response are increased or activated during the peripartum in dairy cows; however, those works were limited by a low number of animals used or by the use of microarray technology, or both. To overcome such limitations, an RNA sequencing analysis was performed on liver biopsies from 20 Holstein cows at 7 ± 5d before (Pre-P) and 16 ± 2d after calving (Post-P). We found 1,475 upregulated and 1,199 downregulated differently expressed genes (DEG) with a false discovery rate adjusted P-value < 0.01 between Pre-P and Post-P. Bioinformatic analysis revealed an activation of the metabolism, especially lipid, glucose, and amino acid metabolism, with increased importance of the mitochondria and a key role of several signaling pathways, chiefly peroxisome proliferators-activated receptor (PPAR) and adipocytokines signaling. Fatty acid oxidation and gluconeogenesis, with a likely increase in amino acid utilization to produce glucose, were among the most important functions revealed by the transcriptomic adaptation to lactation in the liver. Although gluconeogenesis was induced, data indicated decrease in expression of glucose transporters. The analysis also revealed high activation of cell proliferation but inhibition of xenobiotic metabolism, likely due to the liver response to inflammatory-like conditions. Co-expression network analysis disclosed a tight connection and coordination among genes driving biological processes associated with protein synthesis, energy and lipid metabolism, and cell proliferation. Our data confirmed the importance of metabolic adaptation to lipid and glucose metabolism in the liver of early Post-P cows, with a pivotal role of PPAR and adipocytokines.
Collapse
Affiliation(s)
- S T Gao
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - D D Girma
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - M Bionaz
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis 97331
| | - L Ma
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.
| | - D P Bu
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.
| |
Collapse
|
36
|
Rosso C, Caviglia GP, Younes R, Ribaldone DG, Fagoonee S, Pellicano R, Bugianesi E. Molecular mechanisms of hepatic fibrosis in chronic liver diseases. MINERVA BIOTECNOL 2020; 32. [DOI: 10.23736/s1120-4826.20.02619-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
37
|
Aleksenko L, Quaye IK. Pregnancy-induced Cardiovascular Pathologies: Importance of Structural Components and Lipids. Am J Med Sci 2020; 360:447-466. [PMID: 32540145 DOI: 10.1016/j.amjms.2020.05.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 03/09/2020] [Accepted: 05/07/2020] [Indexed: 01/22/2023]
Abstract
Pregnancy leads to adaptations for maternal and fetal energy needs. The cardiovascular system bears the brunt of the adaptations as the heart and vessels enable nutrient supply to maternal organs facilitated by the placenta to the fetus. The components of the cardiovascular system are critical in the balance between maternal homeostatic and fetus driven homeorhetic regulation. Since lipids intersect maternal cardiovascular function and fetal needs with growth and in stress, factors affecting lipid deposition and mobilization impact risk outcomes. Here, the cardiovascular components and functional derangements associated with cardiovascular pathology in pregnancy, vis-à-vis lipid deposition, mobilization and maternal and/or cardiac and fetal energy needs are detailed. Most reports on the components and associated pathology in pregnancy, are on derangements affecting the extracellular matrix and epicardial fat, followed by the endothelium, vascular smooth muscle, pericytes and myocytes. Targeted studies on all cardiovascular components and pathological outcomes in pregnancy will enhance targeted interventions.
Collapse
Affiliation(s)
- Larysa Aleksenko
- Division of Obstetrics and Gynecology, Department of Clinical Sciences, Lund University, Lund, Sweden.
| | - Isaac K Quaye
- Regent University College of Science and Technology, Accra, Ghana
| |
Collapse
|
38
|
Nagaraju R, Joshi AKR, Vamadeva SG, Rajini PS. Deregulation of hepatic lipid metabolism associated with insulin resistance in rats subjected to chronic monocrotophos exposure. J Biochem Mol Toxicol 2020; 34:e22506. [PMID: 32267039 DOI: 10.1002/jbt.22506] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 02/24/2020] [Accepted: 03/26/2020] [Indexed: 12/19/2022]
Abstract
In our previous study, we demonstrated the potential of monocrotophos (MCP), an organophosphorus insecticide (OPI), to induce glucose intolerance, insulin resistance (IR), and dyslipidemia with hyperinsulinemia in rats after chronic exposure. As hyperinsulinemia is likely to exert an impact on hepatic lipid metabolism, we carried out this study to establish the effect of chronic MCP exposure (0.9 and 1.8 mg/kg/day for 180 days) on hepatic lipid metabolism in rats. The state of IR induced by MCP in rats was associated with an increase in the liver lipid content (triglyceride and cholesterol) and expression levels of sterol regulatory element-binding proteins, PPARγ, acetyl-CoA carboxylase, and fatty acid synthase in the liver. Similarly, activities of key enzymes (acetyl-COA carboxylase, fatty acid synthase, lipin 1, malic enzyme, glucose-6-phosphate dehydrogenase, and glycerol-3-phosphate dehydrogenase), which regulate lipogenesis, were enhanced in livers of pesticide-treated rats. A strong correlation was observed between insulin levels, hepatic lipid content, and plasma lipid profile in treated rats. Our study suggests that long-term exposure to OPIs not only has a propensity to induce a state of hyperinsulinemic IR, but it is also associated with augmented hepatic lipogenesis, which may explain dyslipidemia induced by chronic exposure to MCP.
Collapse
Affiliation(s)
- Raju Nagaraju
- Food Protectants and Infestation Control Department, CSIR-Central Food Technological Research Institute, Mysore, Karnataka, India
| | - Apurva K R Joshi
- Food Protectants and Infestation Control Department, CSIR-Central Food Technological Research Institute, Mysore, Karnataka, India
| | - Sowmya G Vamadeva
- Food Protectants and Infestation Control Department, CSIR-Central Food Technological Research Institute, Mysore, Karnataka, India
| | - Padmanabhan S Rajini
- Food Protectants and Infestation Control Department, CSIR-Central Food Technological Research Institute, Mysore, Karnataka, India
| |
Collapse
|
39
|
Moriyama K. Associations Between the Triglyceride to High-Density Lipoprotein Cholesterol Ratio and Metabolic Syndrome, Insulin Resistance, and Lifestyle Habits in Healthy Japanese. Metab Syndr Relat Disord 2020; 18:260-266. [PMID: 32191558 DOI: 10.1089/met.2019.0123] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Background: The triglyceride (TG)/high-density lipoprotein cholesterol (HDL-C) ratio is related to insulin resistance (IR). However, little information is available on whether TG/HDL-C is associated with IR and components of metabolic syndrome (MetS) and how lifestyle habits affect TG/HDL-C in the healthy Japanese population. Methods: In total, 1068 Japanese subjects who had undergone an annual health examination and who were not receiving medication were recruited. Determinants for TG/HDL-C ratio were investigated using multiple regression analyses. The subjects were divided into three groups by lifestyle habits (i.e., smoking, exercise, and physical activity), homeostasis model assessment of IR (HOMA-IR), and fasting plasma glucose, four groups by alcohol intake, and five groups by numbers of MetS components to compare TG/HDL-C values. All analyses were done separately by sex. Results: Multiple regression analyses revealed that waist circumference and smoking were positively associated with TG/HDL-C in both men and women, whereas physical activity was negatively associated with TG/HDL-C ratio in women. TG/HDL-C increased with increasing number of MetS in both men and women. TG/HDL-C increased as HOMA-IR increased in both men and women, when subjects were stratified according to HOMA-IR. TG/HDL-C values were lower in both men and women who exercised regularly, had high physical activity, or were nonsmokers. Independent of exercise and physical activity, TG/HDL-C was higher in smokers than in nonsmokers for both men and women. The relationship between TG/HDL-C and alcohol intake was not statistically significant in both men and women. Conclusions: The TG/HDL-C ratio is associated with IR, components of MetS, exercise, physical activity, and smoking, but not alcohol intake, in healthy Japanese subjects.
Collapse
Affiliation(s)
- Kengo Moriyama
- Department of Clinical Health Science, Tokai University School of Medicine, Tokyo, Japan
| |
Collapse
|
40
|
Improving Dissolution and Cytotoxicity by Forming Multidrug Crystals. Molecules 2020; 25:molecules25061343. [PMID: 32188020 PMCID: PMC7144552 DOI: 10.3390/molecules25061343] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 03/11/2020] [Accepted: 03/13/2020] [Indexed: 12/13/2022] Open
Abstract
Both rosiglitazone and metformin have effects on blood glucose regulation and the proliferation of liver cancer cells. Combination therapy with these two drugs is common and effective for the treatment of diabetes in the clinic, however, the application of these two drugs is influenced by the poor dissolution of rosiglitazone and the gastrointestinal side-effect of metformin resulting from a high solubility. The formation of a multidrug crystal form (Rsg-Met) by a solvent evaporation method can solve the solubility issue. Crystal structure data and intramolecular hydrogen bonds were detected by X-ray diffraction and infrared spectroscopy. Surprisingly, Rsg-Met shortens the time spent in solubility equilibrium and multiplies the dissolution rate of Rsg. Finally, we found that a low concentration of Rsg-Met enhanced the proliferation inhibition effect on liver cancer cells (HepG2, SK-hep1) compared with rosiglitazone, without affecting the human normal cell line LO2.
Collapse
|
41
|
Macho-González A, Garcimartín A, López-Oliva M, Celada P, Bastida S, Benedí J, Sánchez-Muniz F. Carob-fruit-extract-enriched meat modulates lipoprotein metabolism and insulin signaling in diabetic rats induced by high-saturated-fat diet. J Funct Foods 2020. [DOI: 10.1016/j.jff.2019.103600] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
42
|
Li H, Park HM, Ji HS, Han J, Kim SK, Park HY, Jeong TS. Phenolic-enriched blueberry-leaf extract attenuates glucose homeostasis, pancreatic β-cell function, and insulin sensitivity in high-fat diet–induced diabetic mice. Nutr Res 2020; 73:83-96. [DOI: 10.1016/j.nutres.2019.09.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 09/11/2019] [Accepted: 09/16/2019] [Indexed: 12/26/2022]
|
43
|
Xiong Q, Wu Y, Yang M, Wu G, Wang Y, Wang H, Feng J, Song L, Tong B, He G, Xu Y. Nr2e1 ablation impairs liver glucolipid metabolism and induces inflammation, high-fat diets amplify the damage. Biomed Pharmacother 2019; 120:109503. [DOI: 10.1016/j.biopha.2019.109503] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Revised: 09/17/2019] [Accepted: 09/26/2019] [Indexed: 02/08/2023] Open
|
44
|
Lu Z, Xu Z, Shen Z, Shen H, Aschenbach JR. Transcriptomic analyses suggest a dominant role of insulin in the coordinated control of energy metabolism and ureagenesis in goat liver. BMC Genomics 2019; 20:854. [PMID: 31726987 PMCID: PMC6854773 DOI: 10.1186/s12864-019-6233-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Accepted: 10/28/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The ureagenesis plays a central role in the homeostatic control of nitrogen metabolism. This process occurs in the liver, the key metabolic organ in the maintenance of energy homeostasis in the body. To date, the understanding of the influencing factors and regulators of ureagenesis in ruminants is still poor. The aim of this study was to investigate the relationship between energy metabolism and ureagenesis and detect the direct regulators of ureagenesis in the liver by using RNA-seq technology. RESULTS Eighteen four-month-old male goats were divided into two groups randomly and received a diet containing 10% (LNFC group, n = 9) or 30% non-fiber carbohydrate (MNFC group, n = 9), respectively, for four weeks. The global gene expression analysis of liver samples showed that, compared with a LNFC diet, the MNFC diet promoted the expression of genes required for synthesis of fatty acid and glycerol, whereas it suppressed those related to fatty acid oxidation, gluconeogenesis from amino acids and ureagenesis. Additionally, gene expression for rate-limiting enzymes of ureagenesis were highly correlated to the gene expression of key enzymes of both fatty acid synthesis and glycerol synthesis (Spearman correlation coefficient > 0.8 and p < 0.05). In the differentially expressed signaling pathways related to the endocrine system, the MNFC diet activated the insulin and PPAR signaling pathway, whereas it suppressed the leptin-JAK/STAT signaling pathway, compared with the LNFC diet. Reverse transcription quantitative PCR analyses of 40 differentially expressed genes confirmed the RNA-seq results (R2 = 0.78). CONCLUSION Our study indicated that a dietary NFC-induced increase of energy supply promoted lipid anabolism and decreased ureagenesis in the caprine liver. By combining our results with previously published reports, insulin signaling can be suggested to play the dominant role in the coordinated control of hepatic energy metabolism and ureagenesis.
Collapse
Affiliation(s)
- Zhongyan Lu
- Key Lab of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Zhihui Xu
- College of Life Science, Nanjing Agricultural University, Nanjing, Jiangsu, China.,Bioinformatics Center, Nanjing Agricultural University, Weigang No.1, Nanjing, 210095, Jiangsu, China
| | - Zanming Shen
- Key Lab of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Hong Shen
- College of Life Science, Nanjing Agricultural University, Nanjing, Jiangsu, China. .,Bioinformatics Center, Nanjing Agricultural University, Weigang No.1, Nanjing, 210095, Jiangsu, China.
| | - Jörg R Aschenbach
- Institute of Veterinary Physiology, Freie Universität Berlin, Berlin, Germany.
| |
Collapse
|
45
|
Patel BM, Goyal RK. Liver and insulin resistance: New wine in old bottle!!! Eur J Pharmacol 2019; 862:172657. [DOI: 10.1016/j.ejphar.2019.172657] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 09/02/2019] [Accepted: 09/05/2019] [Indexed: 12/20/2022]
|
46
|
Dittmann A, Kennedy NJ, Soltero NL, Morshed N, Mana MD, Yilmaz ÖH, Davis RJ, White FM. High-fat diet in a mouse insulin-resistant model induces widespread rewiring of the phosphotyrosine signaling network. Mol Syst Biol 2019; 15:e8849. [PMID: 31464373 PMCID: PMC6674232 DOI: 10.15252/msb.20198849] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 07/08/2019] [Accepted: 07/09/2019] [Indexed: 12/17/2022] Open
Abstract
Obesity-associated type 2 diabetes and accompanying diseases have developed into a leading human health risk across industrialized and developing countries. The complex molecular underpinnings of how lipid overload and lipid metabolites lead to the deregulation of metabolic processes are incompletely understood. We assessed hepatic post-translational alterations in response to treatment of cells with saturated and unsaturated free fatty acids and the consumption of a high-fat diet by mice. These data revealed widespread tyrosine phosphorylation changes affecting a large number of enzymes involved in metabolic processes as well as canonical receptor-mediated signal transduction networks. Targeting two of the most prominently affected molecular features in our data, SRC-family kinase activity and elevated reactive oxygen species, significantly abrogated the effects of saturated fat exposure in vitro and high-fat diet in vivo. In summary, we present a comprehensive view of diet-induced alterations of tyrosine signaling networks, including proteins involved in fundamental metabolic pathways.
Collapse
MESH Headings
- Animals
- Cell Line, Tumor
- Diabetes Mellitus, Type 2/etiology
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Diet, High-Fat/adverse effects
- Disease Models, Animal
- Fatty Acids/pharmacology
- Hepatocytes/drug effects
- Hepatocytes/metabolism
- Hepatocytes/pathology
- Liver/drug effects
- Liver/metabolism
- Liver/pathology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Obesity/etiology
- Obesity/genetics
- Obesity/metabolism
- Obesity/pathology
- Phosphorylation/drug effects
- Phosphotyrosine/metabolism
- Protein Kinase Inhibitors/pharmacology
- Protein Processing, Post-Translational
- Proteomics/methods
- Rats
- Reactive Oxygen Species/agonists
- Reactive Oxygen Species/metabolism
- Signal Transduction
- src-Family Kinases/genetics
- src-Family Kinases/metabolism
Collapse
Affiliation(s)
- Antje Dittmann
- The David H. Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMAUSA
- Center for Precision Cancer MedicineMassachusetts Institute of TechnologyCambridgeMAUSA
| | - Norman J Kennedy
- Program in Molecular MedicineUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Nina L Soltero
- The David H. Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMAUSA
| | - Nader Morshed
- The David H. Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMAUSA
- Center for Precision Cancer MedicineMassachusetts Institute of TechnologyCambridgeMAUSA
| | - Miyeko D Mana
- The David H. Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMAUSA
- Broad Institute of Harvard and MITCambridgeMAUSA
| | - Ömer H Yilmaz
- The David H. Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMAUSA
- Broad Institute of Harvard and MITCambridgeMAUSA
- Department of PathologyMassachusetts General Hospital and Harvard Medical SchoolBostonMAUSA
| | - Roger J Davis
- Program in Molecular MedicineUniversity of Massachusetts Medical SchoolWorcesterMAUSA
- Howard Hughes Medical InstituteWorcesterMAUSA
| | - Forest M White
- The David H. Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMAUSA
- Center for Precision Cancer MedicineMassachusetts Institute of TechnologyCambridgeMAUSA
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMAUSA
| |
Collapse
|
47
|
Gu C, Li P, Liu W, Zhou Y, Tan W. The role of insulin in transdifferentiated hepatocyte proliferation and function in serum-free medium. J Cell Mol Med 2019; 23:4165-4178. [PMID: 30950200 PMCID: PMC6533558 DOI: 10.1111/jcmm.14303] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 02/20/2019] [Accepted: 03/14/2019] [Indexed: 12/22/2022] Open
Abstract
Transdifferentiated hepatocytes are potential seeding cells for bioartificial liver (BAL) treatment, and it is important to obtain a sufficient number of functional hepatocytes in serum-free medium (SFM). Although insulin plays an essential role in promoting cell proliferation and metabolism, the functions of insulin in transdifferentiated cells remain poorly understood. Here, we found that 1.0 mg/L insulin significantly increased human-induced hepatocyte-like cells (hiHeps) proliferation and viability in SFM. The pro-proliferative effect of insulin on these cells occurred via augmented cyclin D1 expression that was mediated by activation of the Akt1/mTOR/p70S6K and Akt1/P53 pathways. Further studies revealed that insulin also enhanced the specific liver function of hiHeps in SFM. Additionally, Western blotting and siHNF1A transfection analysis showed that insulin increased the protein expression of Albumin (ALB) and UDP-glucuronosyltransferase1A1 (UGT1A1 ) in hiHeps via HNF1A. Finally, hiHep proliferation and the expression of specific genes were maintained during long-term passaging in SFM supplemented with 1.0 mg/L insulin. Collectively, our findings show that insulin promotes transdifferentiated hiHep proliferation and specific functional expression. These findings have important implications for the expansion of functional hiHeps prior to clinical applications of BALs.
Collapse
Affiliation(s)
- Ce Gu
- State Key Laboratory of Bioreactor EngineeringEast China University of Science and TechnologyShanghaiP. R. China
| | - Panpan Li
- State Key Laboratory of Bioreactor EngineeringEast China University of Science and TechnologyShanghaiP. R. China
| | - Wei Liu
- State Key Laboratory of Bioreactor EngineeringEast China University of Science and TechnologyShanghaiP. R. China
| | - Yan Zhou
- State Key Laboratory of Bioreactor EngineeringEast China University of Science and TechnologyShanghaiP. R. China
| | - Wen‐Song Tan
- State Key Laboratory of Bioreactor EngineeringEast China University of Science and TechnologyShanghaiP. R. China
| |
Collapse
|
48
|
Sergi D, Naumovski N, Heilbronn LK, Abeywardena M, O'Callaghan N, Lionetti L, Luscombe-Marsh N. Mitochondrial (Dys)function and Insulin Resistance: From Pathophysiological Molecular Mechanisms to the Impact of Diet. Front Physiol 2019; 10:532. [PMID: 31130874 PMCID: PMC6510277 DOI: 10.3389/fphys.2019.00532] [Citation(s) in RCA: 185] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 04/15/2019] [Indexed: 12/17/2022] Open
Abstract
Mitochondrial dysfunction has been implicated in the pathogenesis of insulin resistance, the hallmark of type 2 diabetes mellitus (T2DM). However, the cause-effect relationship remains to be fully elucidated. Compelling evidence suggests that boosting mitochondrial function may represent a valuable therapeutic tool to improve insulin sensitivity. Mitochondria are highly dynamic organelles, which adapt to short- and long-term metabolic perturbations by undergoing fusion and fission cycles, spatial rearrangement of the electron transport chain complexes into supercomplexes and biogenesis governed by peroxisome proliferator-activated receptor γ co-activator 1α (PGC 1α). However, these processes appear to be dysregulated in type 2 diabetic individuals. Herein, we describe the mechanistic link between mitochondrial dysfunction and insulin resistance in skeletal muscle alongside the intracellular pathways orchestrating mitochondrial bioenergetics. We then review current evidence on nutritional tools, including fatty acids, amino acids, caloric restriction and food bioactive derivatives, which may enhance insulin sensitivity by therapeutically targeting mitochondrial function and biogenesis.
Collapse
Affiliation(s)
- Domenico Sergi
- Nutrition and Health Substantiation Group, Nutrition and Health Program, Health and Biosecurity, Commonwealth Scientific and Industrial Research Organisation (CSIRO), Adelaide, SA, Australia.,Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| | - Nenad Naumovski
- Faculty of Health, University of Canberra, Canberra, ACT, Australia.,Collaborative Research in Bioactives and Biomarkers (CRIBB) Group, Canberra, ACT, Australia
| | | | - Mahinda Abeywardena
- Nutrition and Health Substantiation Group, Nutrition and Health Program, Health and Biosecurity, Commonwealth Scientific and Industrial Research Organisation (CSIRO), Adelaide, SA, Australia
| | - Nathan O'Callaghan
- Nutrition and Health Substantiation Group, Nutrition and Health Program, Health and Biosecurity, Commonwealth Scientific and Industrial Research Organisation (CSIRO), Adelaide, SA, Australia
| | - Lillà Lionetti
- Department of Chemistry and Biology "A. Zambelli", University of Salerno, Fisciano, Italy
| | - Natalie Luscombe-Marsh
- Nutrition and Health Substantiation Group, Nutrition and Health Program, Health and Biosecurity, Commonwealth Scientific and Industrial Research Organisation (CSIRO), Adelaide, SA, Australia.,Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
49
|
Kim SH, Abbasi F. Myths about Insulin Resistance: Tribute to Gerald Reaven. Endocrinol Metab (Seoul) 2019; 34:47-52. [PMID: 30912338 PMCID: PMC6435844 DOI: 10.3803/enm.2019.34.1.47] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 02/14/2019] [Accepted: 02/22/2019] [Indexed: 12/12/2022] Open
Abstract
Gerald Reaven was often called the "father of insulin resistance." On the 1-year anniversary of his death in 2018, we challenge three myths associated with insulin resistance: metformin improves insulin resistance; measurement of waist circumference predicts insulin resistance better than body mass index; and insulin resistance causes weight gain. In this review, we highlight Reaven's relevant research that helped to dispel these myths associated with insulin resistance.
Collapse
Affiliation(s)
- Sun H Kim
- Division of Endocrinology, Gerontology, and Metabolism, Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA, USA.
| | - Fahim Abbasi
- Division of Cardiovascular Medicine, Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
50
|
Mu W, Cheng XF, Liu Y, Lv QZ, Liu GL, Zhang JG, Li XY. Potential Nexus of Non-alcoholic Fatty Liver Disease and Type 2 Diabetes Mellitus: Insulin Resistance Between Hepatic and Peripheral Tissues. Front Pharmacol 2019; 9:1566. [PMID: 30692925 PMCID: PMC6339917 DOI: 10.3389/fphar.2018.01566] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 12/24/2018] [Indexed: 12/21/2022] Open
Abstract
The liver is the central metabolic organ and plays a pivotal role in regulating homeostasis of glucose and lipid metabolism. Aberrant liver metabolism promotes insulin resistance, which is reported to be a common characteristic of metabolic diseases such as non-alcoholic fatty liver disease (NAFLD) and type 2 diabetes mellitus (T2DM). There is a complex and bidirectional relationship between NAFLD and T2DM. NAFLD patients with hepatic insulin resistance generally share a high risk of impaired fasting glucose associated with early diabetes; most patients with T2DM experience non-alcoholic fatty liver (NAFL), non-alcoholic steatohepatitis (NASH), and other more severe liver complications such as cirrhosis and hepatocellular carcinoma (HCC). Additionally, hepatic insulin resistance, which is caused by diacylglycerol-mediated activation of protein kinase C epsilon (PKC𝜀), may be the critical pathological link between NAFLD and T2DM. Therefore, this review aims to illuminate current insights regarding the complex and strong association between NAFLD and T2DM and summarize novel and emerging targets for the treatment of hepatic insulin resistance based on established mechanistic knowledge.
Collapse
Affiliation(s)
- Wan Mu
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xue-Fang Cheng
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Liu
- Department of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qian-Zhou Lv
- Department of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Gao-Lin Liu
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ji-Gang Zhang
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao-Yu Li
- Department of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|