1
|
Hoseini Z, Behpour N, Hoseini R. Aerobic training and vitamin D modulate hepatic miRNA expression to improve lipid metabolism and insulin resistance in type 2 diabetes. Sci Rep 2025; 15:16764. [PMID: 40369056 PMCID: PMC12078466 DOI: 10.1038/s41598-025-01757-x] [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] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 05/08/2025] [Indexed: 05/16/2025] Open
Abstract
The prevalence of type 2 diabetes mellitus (T2DM) has reached epidemic proportions globally, posing a significant burden on public health. Dysregulation of lipid metabolism and insulin resistance in T2DM often leads to hepatic complications, making the modulation of microRNAs (miRNAs) associated with these pathways a promising therapeutic target. This study aimed to investigate the protective effects of aerobic training (AT) and vitamin D supplementation on the liver of individuals with T2DM by examining the modulation of miRNAs related to lipid metabolism and insulin resistance. Specifically, the miRNAs examined in this study were miR-33, miR-122, miR-29, and miR-9. Thirty-two male Wistar rats with T2DM were randomly assigned to four groups: Control (C), AT, moderate dose of Vitamin D supplementation (MD; 5,000 IU), and high dose of Vitamin D supplementation (HD; 10,000 IU). The AT group underwent an eight-week program consisting of treadmill running sessions, five days per week, with a gradual increase in intensity and duration. The vitamin D supplementation groups received either 5,000 or 10,000 IU of vitamin D, administered via injection once weekly for 8 weeks. The study used the STZ + HFD rat model and collected liver tissue samples for analysis. Total RNA, including miRNA, was extracted from the liver tissue samples, and the miRNA expression levels were quantified using quantitative real-time PCR (qRT-PCR). Statistical analyses were performed using one-way ANOVA followed by Tukey's post hoc test. AT led to significantly lower fasting plasma insulin levels (p < 0.05) and a notable improvement in the homeostatic model assessment of insulin resistance (HOMA-IR) index, indicating enhanced insulin sensitivity compared with the control and other groups. It also resulted in significantly lower triglyceride levels (p < 0.01) and a favorable shift in the HDL/LDL ratio, indicative of improved lipid metabolism. Vitamin D supplementation showed a dose-dependent reduction in insulin resistance, with the 10,000 IU group demonstrating a more pronounced improvement compared with the 5,000 IU group. Rats supplemented with vitamin D exhibited a dose-dependent modulation of lipid profile, with the 10,000 IU group demonstrating a more significant decrease in triglycerides and an increase in HDL/LDL ratio. The expression of miR-33, miR-122, miR-29, and miR-9 differed significantly among the experimental groups. The AT group exhibited a significant downregulation of miR-122 and miR-9 while showing a significant upregulation of miR-33 and miR-29 compared to the C and the MD groups. The HD group showed significant downregulation of miR-122 and miR-9 compared to the C and the MD groups. Both AT and high-dose vitamin D supplementation have beneficial effects on insulin levels, insulin resistance, and lipid metabolism in rats with T2DM by modulating miRNA expression, thereby inhibiting insulin resistance and improving T2DM.
Collapse
Affiliation(s)
- Zahra Hoseini
- PhD of Exercise Physiology, Department of Exercise Physiology, Faculty of Sport Sciences, Razi University, Kermanshah, Iran
| | - Nasser Behpour
- Department of Exercise Physiology, Faculty of Sport Sciences, Razi University, P.O.Box. 6714967346, Kermanshah, Iran.
| | - Rastegar Hoseini
- Department of Exercise Physiology, Faculty of Sport Sciences, Razi University, P.O.Box. 6714967346, Kermanshah, Iran
| |
Collapse
|
2
|
Thakur MR, Tupe RS. l-Arginine: A multifaceted regulator of diabetic cardiomyopathy. Biochem Biophys Res Commun 2025; 761:151720. [PMID: 40186920 DOI: 10.1016/j.bbrc.2025.151720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/25/2025] [Accepted: 03/27/2025] [Indexed: 04/07/2025]
Abstract
In diabetes mellitus, dysregulated glucose and lipid metabolism lead to diabetic cardiomyopathy (DCM) by imparting pathological myocardial remodeling and cellular injury. Accelerated glycation, oxidative stress, and activated inflammatory pathways culminate in cardiac fibrosis and hypertrophy in DCM. The regulatory effects of l-Arginine (L-Arg) have been elucidated in the pathological changes of DCM, including myocardial fibrosis, hypertrophy, and apoptosis, by inhibiting glycation and oxidative stress-induced inflammation. Disturbed L-Arg metabolism and decreased intracellular L-Arg pool are correlated with the progression of DCM; therefore, L-Arg supplementation has been prescribed for various cardiovascular dysfunctions. This review expands the therapeutic potential of L-Arg supplementation in DCM by elucidating its molecular mechanism of action and exploring potential clinical outcomes.
Collapse
Affiliation(s)
- Muskan R Thakur
- Symbiosis School of Biological Sciences (SSBS), Symbiosis International (Deemed University) (SIU), Lavale, Pune, 412115, Maharashtra, India
| | - Rashmi S Tupe
- Symbiosis School of Biological Sciences (SSBS), Symbiosis International (Deemed University) (SIU), Lavale, Pune, 412115, Maharashtra, India.
| |
Collapse
|
3
|
Beheshti AS, Qazvini MM, Abeq M, Abedi E, Fadaei MS, Fadaei MR, Baradaran Rahimi V, Askari VR. Molecular, cellular, and metabolic insights of cinnamon (Cinnamomum zeylanicum) advantages in diabetes and related complications: condiment or medication? NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:3513-3526. [PMID: 39589531 DOI: 10.1007/s00210-024-03644-0] [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: 06/08/2024] [Accepted: 11/15/2024] [Indexed: 11/27/2024]
Abstract
Diabetes mellitus (DM) is a growing concern in public health, which affects about 10% of the population. There are several chronic complications due to DM, including kidney failure, blindness, amputations, myocardial infarction, and stroke. Cinnamon zeylanicum (C. verum, Ceylon cinnamon, or true cinnamon) has been shown to have desirable effects such as anti-obesity, anti-diabetic, anti-dyslipidemia, and anti-inflammatory effects in experimental studies. In this regard, Scopus, PubMed, and Google Scholar databases have been investigated with keywords of "Cinnamon," "Cinnamomum zeylanicum," "diabetes mellitus," "diabetes complication," "hypoglycemic," "anti-hyperglycemic," and "anti-diabetic" from incept to June 2024. This study aimed to review all pharmacological effects and molecular pathways of C. zeylanicum in DM and its complications in vitro, in vivo, and in clinical. Based on these studies, C. zeylanicum has good potential to design human studies for controlling and modifying DM and its related disorders.
Collapse
Affiliation(s)
- Anahita Sadat Beheshti
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Mahdi Qazvini
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahsa Abeq
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ermia Abedi
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Saleh Fadaei
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Reza Fadaei
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vafa Baradaran Rahimi
- Department of Cardiovascular Diseases, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vahid Reza Askari
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
4
|
Albahri G, Badran A, Baki ZA, Alame M, Hijazi A, Daou A, Mesmar JE, Baydoun E. Mandragora autumnalis Distribution, Phytochemical Characteristics, and Pharmacological Bioactivities. Pharmaceuticals (Basel) 2025; 18:328. [PMID: 40143106 PMCID: PMC11944648 DOI: 10.3390/ph18030328] [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: 01/16/2025] [Revised: 02/21/2025] [Accepted: 02/24/2025] [Indexed: 03/28/2025] Open
Abstract
In the Mediterranean and Himalayan regions, the genus Mandragora (family Solanaceae), sometimes called mandrake, is widely utilized in herbal therapy and is well-known for its mythical associations. Objective: To compile up-to-date information on M. autumnalis's therapeutic properties. Its pharmacological properties and phytochemical composition are particularly covered in managing several illnesses, including diabetes, cancer, and heart disease. Methods: Articles on the review topic were found by searching major scientific literature databases, such as PubMed, Scopus, ScienceDirect, SciFinder, Chemical Abstracts, and Medicinal and Aromatic Plants Abstracts. Additionally, general online searches were conducted using Google Scholar and Google. The time frame for the search included items released from 1986 to 2023. Results:Mandragora has been shown to contain a variety of phytochemicals, including coumarins, withanolides, and alkaloids. The pharmacological characteristics of M. autumnalis, such as increasing macrophage anti-inflammatory activity, free radicals inhibition, bacterial and fungal growth inhibition, cytotoxic anticancer activities in vivo and in vitro against cancer cell lines, and enzyme-inhibitory properties, are attributed to these phytochemicals. Furthermore, M. autumnalis also inhibits cholinesterase, tyrosinase, α-amylase, α-glucosidase, and free radicals. On the other hand, metabolic risk factors, including the inhibition of diabetes-causing enzymes and obesity, have been treated using dried ripe berries. Conclusions: Investigations into the pharmacological and phytochemical characteristics of M. autumnalis have revealed that this plant is a rich reservoir of new bioactive substances. This review aims to provide insight into the botanical and ecological characteristics of Mandragora autumnalis, including a summary of its phytochemical components and antioxidant, antimicrobial, antidiabetic, anticancer, enzyme-inhibitory properties, as well as toxicological implications, where its low cytotoxic activity against the normal VERO cell line has been shown. More research on this plant is necessary to ensure its efficacy and safety. Still, it is also necessary to understand the molecular mechanism of action behind the observed effects to clarify its therapeutic potential.
Collapse
Affiliation(s)
- Ghosoon Albahri
- Doctoral School of Science and Technology-Platform of Research and Analysis in Environmental Sciences (EDST-PRASE), Beirut P.O. Box 657314, Lebanon; (G.A.); (M.A.); (A.H.)
- Department of Biology, Faculty of Arts and Sciences, American University of Beirut, Riad El Solh, Beirut 1107, Lebanon;
| | - Adnan Badran
- Department of Nutrition, University of Petra Amman Jordan, Amman P.O. Box 961343, Jordan;
| | - Zaher Abdel Baki
- College of Engineering and Technology, American University of the Middle East, Egaila 54200, Kuwait;
| | - Mohamad Alame
- Doctoral School of Science and Technology-Platform of Research and Analysis in Environmental Sciences (EDST-PRASE), Beirut P.O. Box 657314, Lebanon; (G.A.); (M.A.); (A.H.)
| | - Akram Hijazi
- Doctoral School of Science and Technology-Platform of Research and Analysis in Environmental Sciences (EDST-PRASE), Beirut P.O. Box 657314, Lebanon; (G.A.); (M.A.); (A.H.)
- Faculty of Sciences, Kut University College, Wasit 52001, Iraq
| | - Anis Daou
- Pharmaceutical Sciences Department, College of Pharmacy, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
| | - Joelle Edward Mesmar
- Department of Biology, Faculty of Arts and Sciences, American University of Beirut, Riad El Solh, Beirut 1107, Lebanon;
| | - Elias Baydoun
- Department of Biology, Faculty of Arts and Sciences, American University of Beirut, Riad El Solh, Beirut 1107, Lebanon;
| |
Collapse
|
5
|
Szkudelski T, Szkudelska K. The relevance of the heme oxygenase system in alleviating diabetes-related hormonal and metabolic disorders. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167552. [PMID: 39490940 DOI: 10.1016/j.bbadis.2024.167552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 11/05/2024]
Abstract
Heme oxygenase (HO) is an enzyme that catalyzes heme degradation. HO dysfunction is linked to various pathological conditions, including diabetes. Results of animal studies indicate that HO expression and activity are downregulated in experimentally induced diabetes. This is associated with severe hormonal and metabolic disturbances. However, these pathological changes have been shown to be reversed by therapy with HO activators. In animals with experimentally induced diabetes, HO was upregulated by genetic manipulation or by pharmacological activators such as hemin and cobalt protoporphyrin. Induction of HO alleviated elevated blood glucose levels and improved insulin action, among other effects. This effect resulted from beneficial changes in the main insulin-sensitive tissues, i.e., the skeletal muscle, the liver, and the adipose tissue. The action of HO activators was due to positive alterations in pivotal signaling molecules and regulatory enzymes. Furthermore, diabetes-related oxidative and inflammatory stress was reduced due to HO induction. HO upregulation was effective in various animal models of type 1 and type 2 diabetes. These data suggest the possibility of testing HO activators as a potential tool for alleviating hormonal and metabolic disorders in people with diabetes.
Collapse
Affiliation(s)
- Tomasz Szkudelski
- Department of Animal Physiology, Biochemistry and Biostructure, Poznan University of Life Sciences, Wolynska 35, 60-637 Poznan, Poland.
| | - Katarzyna Szkudelska
- Department of Animal Physiology, Biochemistry and Biostructure, Poznan University of Life Sciences, Wolynska 35, 60-637 Poznan, Poland.
| |
Collapse
|
6
|
Kaur A, Singh S, Mujwar S, Singh TG. Molecular Mechanisms Underlying the Therapeutic Potential of Plant-Based α-Amylase Inhibitors for Hyperglycemic Control in Diabetes. Curr Diabetes Rev 2025; 21:e020724231486. [PMID: 38956911 DOI: 10.2174/0115733998304373240611110224] [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: 02/17/2024] [Revised: 05/14/2024] [Accepted: 05/30/2024] [Indexed: 07/04/2024]
Abstract
BACKGROUND Diabetes mellitus (DM), arising from pancreatic β-cell dysfunction and disrupted alpha-amylase secretion, manifests as hyperglycemia. Synthetic inhibitors of alphaamylase like acarbose manage glucose but pose adverse effects, prompting interest in plantderived alternatives rich in antioxidants and anti-inflammatory properties. OBJECTIVE The current review investigates plant-based alpha-amylase inhibitors, exploring their potential therapeutic roles in managing DM. Focusing on their ability to modulate postprandial hyperglycemia by regulating alpha-amylase secretion, it assesses their efficacy, health benefits, and implications for diabetes treatment. METHODS This review examines plant-derived alpha-amylase inhibitors as prospective diabetic mellitus treatments using PubMed, Google Scholar, and Scopus data. RESULTS Plant-derived inhibitors, including A. deliciosa, B. egyptiaca, and N. nucifera, exhibit anti-inflammatory and antioxidant properties, effectively reducing alpha-amylase levels in diabetic conditions. Such alpha-amylase inhibitors showed promising alternative treatment in managing diabetes with reduced adverse effects. CONCLUSION The current literature concludes that plant-derived alpha-amylase inhibitors present viable therapeutic avenues for diabetes management by modulating alpha-amylase secretion by regulating inflammatory, oxidative stress, and apoptotic mechanisms involved in the pathogenesis of diabetes. Further investigation into their formulations and clinical efficacy may reveal their more comprehensive diabetes therapeutic significance, emphasizing their potential impact on glucose regulation and overall health.
Collapse
Affiliation(s)
- Amritpal Kaur
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India
| | - Shareen Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India
| | - Somdutt Mujwar
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India
| |
Collapse
|
7
|
Zhou JX, Zheng ZY, Peng ZX, Yang YT, Ni HG. Predictive model in silicon and pathogenicity mechanism of metabolic syndrome: Impacts of heavy metal exposure. JOURNAL OF ENVIRONMENTAL MANAGEMENT 2025; 373:124001. [PMID: 39746257 DOI: 10.1016/j.jenvman.2024.124001] [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: 08/29/2024] [Revised: 12/03/2024] [Accepted: 12/29/2024] [Indexed: 01/04/2025]
Abstract
Although the association between heavy metals in human and the development of metabolic syndrome (MetS) have been extensively studied, the pathogenic mechanism of MetS affected by metals is not clear to date. In this study, a predictive model was developed with machine learning base on the large-scale dataset. These proposed models were evaluated via comparatively analysis of their accuracy and robustness. With the optimal model, two metals significantly correlated with MetS were screened and were employed to infer the pathogenicity mechanism of MetS via molecular docking. Significant associations between heavy metals and MetS were found. Molecular docking provided insights into the interactions between metal ions and key protein receptors involved in metabolic regulation, suggesting a mechanism by which heavy metals interfere with metabolic functions. Specifically, Ba and Cd affect the development of MetS thru their interactions with insulin and estrogen receptors. This study attempted to explore heavy metals' potential roles in MetS at the molecular level. These findings emphasize the importance of addressing environmental exposures in the prevention and treatment of MetS.
Collapse
Affiliation(s)
- Jing-Xuan Zhou
- School of Urban Planning and Design, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Zi-Yi Zheng
- School of Urban Planning and Design, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Zhao-Xing Peng
- School of Urban Planning and Design, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Yu-Ting Yang
- School of Urban Planning and Design, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Hong-Gang Ni
- School of Urban Planning and Design, Peking University Shenzhen Graduate School, Shenzhen, 518055, China.
| |
Collapse
|
8
|
Islam MS, Wei P, Suzauddula M, Nime I, Feroz F, Acharjee M, Pan F. The interplay of factors in metabolic syndrome: understanding its roots and complexity. Mol Med 2024; 30:279. [PMID: 39731011 DOI: 10.1186/s10020-024-01019-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 11/29/2024] [Indexed: 12/29/2024] Open
Abstract
Metabolic syndrome (MetS) is an indicator and diverse endocrine syndrome that combines different metabolic defects with clinical, physiological, biochemical, and metabolic factors. Obesity, visceral adiposity and abdominal obesity, dyslipidemia, insulin resistance (IR), elevated blood pressure, endothelial dysfunction, and acute or chronic inflammation are the risk factors associated with MetS. Abdominal obesity, a hallmark of MetS, highlights dysfunctional fat tissue and increased risk for cardiovascular disease and diabetes. Insulin, a vital peptide hormone, regulates glucose metabolism throughout the body. When cells become resistant to insulin's effects, it disrupts various molecular pathways, leading to IR. This condition is linked to a range of disorders, including obesity, diabetes, fatty liver disease, cardiovascular disease, and polycystic ovary syndrome. Atherogenic dyslipidemia is characterized by three key factors: high levels of small, low-dense lipoprotein (LDL) particles and triglycerides, alongside low levels of high-density lipoprotein (HDL), the "good" cholesterol. Such a combination is a major player in MetS, where IR is a driving force. Atherogenic dyslipidemia contributes significantly to the development of atherosclerosis, which can lead to cardiovascular disease. On top of that, genetic alteration and lifestyle factors such as diet and exercise influence the complexity and progression of MetS. To enhance our understanding and consciousness, it is essential to understand the fundamental pathogenesis of MetS. This review highlights current advancements in MetS research including the involvement of gut microbiome, epigenetic regulation, and metabolomic profiling for early detection of Mets. In addition, this review emphasized the epidemiology and fundamental pathogenesis of MetS, various risk factors, and their preventive measures. The goal of this effort is to deepen understanding of MetS and encourage further research to develop effective strategies for preventing and managing complex metabolic diseases.
Collapse
Affiliation(s)
- Md Sharifull Islam
- Center for Cancer Immunology, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Department of Microbiology, Stamford University Bangladesh, 51, Siddeswari Road, Dhaka, 1217, Bangladesh
| | - Ping Wei
- Center for Cancer Immunology, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Department of Pediatric Otolaryngology Head and Neck Surgery, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Md Suzauddula
- Department of Food Nutrition Dietetics and Health, Kansas State University, Manhattan, KS, 66506, USA
| | - Ishatur Nime
- Key Laboratory of Environment Correlative Dietology, College of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Farahnaaz Feroz
- Department of Microbiology, Stamford University Bangladesh, 51, Siddeswari Road, Dhaka, 1217, Bangladesh
| | - Mrityunjoy Acharjee
- Department of Microbiology, Stamford University Bangladesh, 51, Siddeswari Road, Dhaka, 1217, Bangladesh
| | - Fan Pan
- Center for Cancer Immunology, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
9
|
Bredefeld CL, Choi P, Cullen T, Nicolich-Henkin SJ, Waters L. Statin Use and Hyperglycemia: Do Statins Cause Diabetes? Curr Atheroscler Rep 2024; 27:18. [PMID: 39699704 DOI: 10.1007/s11883-024-01266-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2024] [Indexed: 12/20/2024]
Abstract
PURPOSE OF REVIEW Atherosclerotic cardiovascular disease (ASCVD) and diabetes are leading causes of morbidity and mortality in the United States and globally. Statin medications, a cornerstone of ASCVD prevention and treatment strategies, have been demonstrated to cause hyperglycemia and new onset diabetes mellitus (NODM). The purpose of this review is to summarize existing and emerging knowledge around the intersection of statins and these two important clinical problems. RECENT FINDINGS Since initial reporting of statin-induced hyperglycemia and NODM, the totality of available data corroborates an association between incident diabetes and statin use. A consensus that high-intensity statin and individuals with obesity or glycemic parameters approximating diabetes thresholds constitute the majority of risk exists. Alterations in insulin signaling, glucose transport and gastrointestinal microbiota are leading hypotheses underlying the mechanisms of statin-induced hyperglycemia. The probability of NODM based on an individual's risk factors and statin specific properties can be anticipated. This risk needs to be contextualized with the risk of ASCVD. In order to effectively adjudicate the risk of NODM, improvement in formulating and ultimately conveying a comprehensive ASCVD risk assessment to patients is necessary.
Collapse
Affiliation(s)
- Cindy L Bredefeld
- Department of Medicine, New York University Grossman Long Island School of Medicine, NYU Langone Hospital-Long Island, Garden City, NY, 11530, USA.
- Department of Foundations of Medicine, New York University Grossman Long Island School of Medicine, NYU Langone Hospital-Long Island, Mineola, NY, 11501, USA.
| | - Paula Choi
- Department of Medicine, New York University Grossman Long Island School of Medicine, NYU Langone Hospital-Long Island, Garden City, NY, 11530, USA
| | - Tiffany Cullen
- Department of Medicine, New York University Grossman Long Island School of Medicine, NYU Langone Hospital-Long Island, Garden City, NY, 11530, USA
| | - Sophie J Nicolich-Henkin
- Department of Medicine, New York University Grossman Long Island School of Medicine, NYU Langone Hospital-Long Island, Garden City, NY, 11530, USA
| | - Lauren Waters
- Department of Medicine, New York University Grossman Long Island School of Medicine, NYU Langone Hospital-Long Island, Garden City, NY, 11530, USA
| |
Collapse
|
10
|
Ashraf MS, Tuli K, Moiz S, Sharma SK, Sharma D, Adnan M. AMP kinase: A promising therapeutic drug target for post-COVID-19 complications. Life Sci 2024; 359:123202. [PMID: 39489398 DOI: 10.1016/j.lfs.2024.123202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 10/27/2024] [Accepted: 10/29/2024] [Indexed: 11/05/2024]
Abstract
The COVID-19 pandemic, caused by SARS-CoV-2, has resulted in severe respiratory issues and persistent complications, particularly affecting glucose metabolism. Patients with or without pre-existing diabetes often experience worsened symptoms, highlighting the need for innovative therapeutic approaches. AMPK, a crucial regulator of cellular energy balance, plays a pivotal role in glucose metabolism, insulin sensitivity, and inflammatory responses. AMPK activation, through allosteric or kinase-dependent mechanisms, impacts cellular processes like glucose uptake, fatty acid oxidation, and autophagy. The tissue-specific distribution of AMPK emphasizes its role in maintaining metabolic homeostasis throughout the body. Intriguingly, SARS-CoV-2 infection inhibits AMPK, contributing to metabolic dysregulation and post-COVID-19 complications. AMPK activators like capsaicinoids, curcumin, phytoestrogens, cilostazol, and momordicosides have demonstrated the potential to regulate AMPK activity. Compounds from various sources improve fatty acid oxidation and insulin sensitivity, with metformin showing opposing effects on AMPK activation compared to the virus, suggesting potential therapeutic options. The diverse effects of AMPK activation extend to its role in countering viral infections, further highlighting its significance in COVID-19. This review explores AMPK activation mechanisms, its role in metabolic disorders, and the potential use of natural compounds to target AMPK for post-COVID-19 complications. Also, it aims to review the possible methods of activating AMPK to prevent post-COVID-19 diabetes and cardiovascular complications. It also explores the use of natural compounds for their therapeutic effects in targeting the AMPK pathways. Targeting AMPK activation emerges as a promising avenue to mitigate the long-term effects of COVID-19, offering hope for improved patient outcomes and a better quality of life.
Collapse
Affiliation(s)
- Mohammad Saquib Ashraf
- Department of Medical Laboratory Science College of Pharmacy, Nursing and Medical Science Riyadh ELM University, Riyadh, P.O. Box 12734, Saudi Arabia.
| | - Kanika Tuli
- Guru Nanak Institute of Pharmacy, Dalewal, Hoshiarpur 144208, Punjab, India
| | - Shadman Moiz
- Department of Biotechnology, Lalit Narayan Mithila University, Darbhanga 846004, Bihar, India
| | - Satish Kumar Sharma
- Department of Pharmacology, Glocal School of Pharmacy, The Glocal University, Saharanpur, India
| | - Deepa Sharma
- UMM Matrix Innovations Private Limited, Delhi 110044, India
| | - Mohd Adnan
- Department of Biology, College of Science, University of Ha'il, Ha'il, P.O. Box 2440, Saudi Arabia; Center for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 602105, India
| |
Collapse
|
11
|
Laddha AP, Wahane A, Bahal R, Manautou JE. Investigating the crosstalk between ABCC4 and ABCC5 in 3T3-L1 adipocyte differentiation. Front Mol Biosci 2024; 11:1498946. [PMID: 39717760 PMCID: PMC11663720 DOI: 10.3389/fmolb.2024.1498946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 11/18/2024] [Indexed: 12/25/2024] Open
Abstract
Introduction The plasma membrane-bound protein, multi-drug resistance-associated protein 4 (MRP4/ABCC4), has gained attention for its pivotal role in facilitating the efflux of a wide range of endogenous and xenobiotic molecules. Its significance in adipogenesis and fatty acid metabolism has been brought to light by recent studies. Notably, research on ABCC4 knockout (ABCC4 -/- ) mice has established a link between the absence of ABCC4 and the development of obesity and diabetes. Nevertheless, the specific contribution of ABCC4 within adipose tissue remains largely unexplored. Methods To address this gap, we conducted a study to elucidate the role of the ABCC4 transporter in mature adipocytes, using siRNA constructs to silence its gene function. Results The successful knockdown of ABCC4 significantly altered lipid status and adipogenic gene expression in mature 3T3-L1 adipocytes. Intriguingly, this knockdown also altered the gene expression patterns of other ABCC transporter family members in 3T3-L1 cells. The downregulation of ABCC5 expression was particularly noteworthy, suggesting potential crosstalk between ABCC transporters in mature adipocytes. Additionally, knocking down ABCC5 resulted in significantly higher adipogenic and lipogenic gene expression levels. Oil Red O staining confirmed increased lipid accumulation following the knockdown of ABCC4 and ABCC5. Surprisingly, the simultaneous knockdown of both transporters did not show a cumulative effect on adipogenesis, rather it led to higher levels of intracellular cAMP and extracellular prostaglandin metabolite, both of which are essential signaling molecules in adipogenesis. Conclusion These results highlight the complex interplay between ABCC4 and ABCC5 transporters in adipocyte function and suggest their individual contributions toward obesity and related disorders.
Collapse
Affiliation(s)
| | | | | | - José E. Manautou
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, United States
| |
Collapse
|
12
|
Wang K, Zhao H, Zhao X, Zhang X, Zhang W, Cheng Y, Ge J. Photobiomodulation for diabetes and its complications: a review of general presentation, mechanisms and efficacy. Ann Med 2024; 56:2433684. [PMID: 39607829 PMCID: PMC11610354 DOI: 10.1080/07853890.2024.2433684] [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: 04/08/2024] [Revised: 06/12/2024] [Accepted: 08/01/2024] [Indexed: 11/30/2024] Open
Abstract
Diabetes mellitus is a metabolic disease that is marked by persistent hyperglycemia due to inadequate insulin secretion or insulin resistance. Its prevalence is increasing yearly. Diabetes mellitus can lead to serious health complications that are the primary cause of mortality and disability among diabetic patients, including diabetic retinopathy, diabetic foot ulcers, diabetic peripheral neuropathy, and diabetic periodontitis, and so on. Traditional treatments for diabetes and its complications still suffer from limited clinical efficacy and high therapeutic side effects. Photobiomodulation (PBM), which utilizes low levels of red or near-infrared laser to irradiate cells and tissues, has been shown to be efficacious for a wide range of organ damage. In this study, we focus on the application of PBM in diabetes and its complications and mechanisms, as well as the advantages, disadvantages with the aim of developing new ideas for the application of PBM.
Collapse
Affiliation(s)
- Keyan Wang
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, China
- Department of Physiology, College of Basic Medical Sciences, Jilin University, Changchun, China
- China Japan Union Hospital of Jilin University, Changchun, China
| | - Hongwei Zhao
- Department of Physiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Xiaoqing Zhao
- China Japan Union Hospital of Jilin University, Changchun, China
| | - Xiaoyu Zhang
- China Japan Union Hospital of Jilin University, Changchun, China
| | - Wei Zhang
- Department of Physiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Yan Cheng
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, China
| | - Jingyan Ge
- Department of Physiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| |
Collapse
|
13
|
Kato Y, Ariyoshi K, Nohara Y, Matsunaga N, Shimauchi T, Shindo N, Nishimura A, Mi X, Kim SG, Ide T, Kawanishi E, Ojida A, Nakashima N, Mori Y, Nishida M. Inhibition of dynamin-related protein 1-filamin interaction improves systemic glucose metabolism. Br J Pharmacol 2024; 181:4328-4347. [PMID: 38986570 DOI: 10.1111/bph.16487] [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: 09/22/2023] [Revised: 05/05/2024] [Accepted: 05/18/2024] [Indexed: 07/12/2024] Open
Abstract
BACKGROUND AND PURPOSE Maintaining mitochondrial quality is attracting attention as a new strategy to treat diabetes and diabetic complications. We previously reported that mitochondrial hyperfission by forming a protein complex between dynamin-related protein (Drp) 1 and filamin, mediates chronic heart failure and cilnidipine, initially developed as an L/N-type Ca2+ channel blocker, improves heart failure by inhibiting Drp1-filamin protein complex. We investigated whether cilnidipine improves hyperglycaemia of various diabetic mice models. EXPERIMENTAL APPROACH Retrospective analysis focusing on haemoglobin A1c (HbA1c) was performed in hypertensive and hyperglycaemic patients taking cilnidipine and amlodipine. After developing diabetic mice by streptozotocin (STZ) treatment, an osmotic pump including drug was implanted intraperitoneally, followed by weekly measurements of blood glucose levels. Mitochondrial morphology was analysed by electron microscopy. A Ca2+ channel-insensitive cilnidipine derivative (1,4-dihydropyridine [DHP]) was synthesized and its pharmacological effect was evaluated using obese (ob/ob) mice fed with high-fat diet (HFD). KEY RESULTS In patients, cilnidipine was superior to amlodipine in HbA1c lowering effect. Cilnidipine treatment improved systemic hyperglycaemia and mitochondrial morphological abnormalities in STZ-exposed mice, without lowering blood pressure. Cilnidipine failed to improve hyperglycaemia of ob/ob mice, with suppressing insulin secretion. 1,4-DHP improved hyperglycaemia and mitochondria abnormality in ob/ob mice fed HFD. 1,4-DHP and cilnidipine improved basal oxygen consumption rate of HepG2 cells cultured under 25 mM glucose. CONCLUSION AND IMPLICATIONS Inhibition of Drp1-filamin protein complex formation becomes a new strategy for type 2 diabetes treatment.
Collapse
Affiliation(s)
- Yuri Kato
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Kohei Ariyoshi
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Yasunobu Nohara
- Faculty of Advanced Science and Technology, Kumamoto University, Kumamoto, Japan
| | - Naoya Matsunaga
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Tsukasa Shimauchi
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
- Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences, Okazaki, Aichi, Japan
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Aichi, Japan
| | - Naoya Shindo
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Akiyuki Nishimura
- National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences, Okazaki, Aichi, Japan
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Aichi, Japan
- SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi, Japan
| | - Xinya Mi
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Sang Geon Kim
- College of Pharmacy, Dongguk University-Seoul, Goyang-si, South Korea
| | - Tomomi Ide
- Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Eiji Kawanishi
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Akio Ojida
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Naoki Nakashima
- Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yasuo Mori
- Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Motohiro Nishida
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
- National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences, Okazaki, Aichi, Japan
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Aichi, Japan
- SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi, Japan
| |
Collapse
|
14
|
Ansari P, Khan JT, Chowdhury S, Reberio AD, Kumar S, Seidel V, Abdel-Wahab YHA, Flatt PR. Plant-Based Diets and Phytochemicals in the Management of Diabetes Mellitus and Prevention of Its Complications: A Review. Nutrients 2024; 16:3709. [PMID: 39519546 PMCID: PMC11547802 DOI: 10.3390/nu16213709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/27/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Diabetes mellitus (DM) is currently regarded as a global public health crisis for which lifelong treatment with conventional drugs presents limitations in terms of side effects, accessibility, and cost. Type 2 diabetes (T2DM), usually associated with obesity, is characterized by elevated blood glucose levels, hyperlipidemia, chronic inflammation, impaired β-cell function, and insulin resistance. If left untreated or when poorly controlled, DM increases the risk of vascular complications such as hypertension, nephropathy, neuropathy, and retinopathy, which can be severely debilitating or life-threatening. Plant-based foods represent a promising natural approach for the management of T2DM due to the vast array of phytochemicals they contain. Numerous epidemiological studies have highlighted the importance of a diet rich in plant-based foods (vegetables, fruits, spices, and condiments) in the prevention and management of DM. Unlike conventional medications, such natural products are widely accessible, affordable, and generally free from adverse effects. Integrating plant-derived foods into the daily diet not only helps control the hyperglycemia observed in DM but also supports weight management in obese individuals and has broad health benefits. In this review, we provide an overview of the pathogenesis and current therapeutic management of DM, with a particular focus on the promising potential of plant-based foods.
Collapse
Affiliation(s)
- Prawej Ansari
- Comprehensive Diabetes Center, Heersink School of Medicine, University of Alabama, Birmingham (UAB), Birmingham, AL 35233, USA
- School of Pharmacy and Public Health, Department of Pharmacy, Independent University, Bangladesh (IUB), Dhaka 1229, Bangladesh
- Centre for Diabetes Research, School of Biomedical Sciences, Ulster University, Coleraine BT52 1SA, UK; (Y.H.A.A.-W.); (P.R.F.)
| | - Joyeeta T. Khan
- School of Pharmacy and Public Health, Department of Pharmacy, Independent University, Bangladesh (IUB), Dhaka 1229, Bangladesh
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences (UAMS), Little Rock, AR 72205, USA
| | - Suraiya Chowdhury
- School of Pharmacy and Public Health, Department of Pharmacy, Independent University, Bangladesh (IUB), Dhaka 1229, Bangladesh
| | - Alexa D. Reberio
- School of Pharmacy and Public Health, Department of Pharmacy, Independent University, Bangladesh (IUB), Dhaka 1229, Bangladesh
| | - Sandeep Kumar
- Comprehensive Diabetes Center, Heersink School of Medicine, University of Alabama, Birmingham (UAB), Birmingham, AL 35233, USA
| | - Veronique Seidel
- Natural Products Research Laboratory, Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK;
| | - Yasser H. A. Abdel-Wahab
- Centre for Diabetes Research, School of Biomedical Sciences, Ulster University, Coleraine BT52 1SA, UK; (Y.H.A.A.-W.); (P.R.F.)
| | - Peter R. Flatt
- Centre for Diabetes Research, School of Biomedical Sciences, Ulster University, Coleraine BT52 1SA, UK; (Y.H.A.A.-W.); (P.R.F.)
| |
Collapse
|
15
|
Abeysekera MV, Ni D, Gilbert L, Hibbert E, Nanan R. Linking the reversal of gestational insulin resistance to postpartum depression. BMC Med 2024; 22:433. [PMID: 39379948 PMCID: PMC11462660 DOI: 10.1186/s12916-024-03659-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 09/26/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND Postpartum depression (PPD) constitutes a significant mental health disorder affecting almost one fifth of pregnancies globally. Despite extensive research, the precise etiological mechanisms underlying PPD remain elusive. However, several risk factors like genetic predisposition, hormonal fluctuations, and stress-related environmental and psychosocial triggers have been found to be implicated in its development. MAIN: Recently, an increased risk of PPD has been reported to be associated with gestational diabetes mellitus (GDM), which is characterized by the disruption of glucose metabolism, primarily attributed to the emergence of insulin resistance (IR). While IR during pregnancy seems to be an evolutionary adaptative mechanism to handle the profound metabolic alterations during pregnancy, its subsequent resolution following delivery necessitates a reconfiguration of the metabolic landscape in both peripheral tissues and the central nervous system (CNS). Considering the pivotal roles of energy metabolism, particularly glucose metabolism, in CNS functions, we propose a novel model that such pronounced changes in IR and the associated glucose metabolism seen postpartum might account for PPD development. This concept is based on the profound influences from insulin and glucose metabolism on brain functions, potentially via modulating neurotransmitter actions of dopamine and serotonin. Their sudden postpartum disruption is likely to be linked to mood changes, as observed in PPD. CONCLUSIONS The detailed pathogenesis of PPD might be multifactorial and still remains to be fully elucidated. Nevertheless, our hypothesis might account in part for an additional etiological factor to PPD development. If our concept is validated, it can provide guidance for future PPD prevention, diagnosis, and intervention.
Collapse
Affiliation(s)
| | - Duan Ni
- Nepean Hospital, Nepean Blue Mountains Local Health District, Sydney, NSW, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Leah Gilbert
- Nepean Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Emily Hibbert
- Nepean Hospital, Nepean Blue Mountains Local Health District, Sydney, NSW, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- Nepean Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Ralph Nanan
- Nepean Hospital, Nepean Blue Mountains Local Health District, Sydney, NSW, Australia.
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia.
- Nepean Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
16
|
Raza A, Mushtaq MN, Hassan S, Sharif A, Akhtar B, Akhtar MF. Mitigation of Diabetes Mellitus Using Euphorbia helioscopia Leaf Ethanolic Extract by Modulating GCK, GLUT4, IGF, and G6P Expressions in Streptozotocin-Induced Diabetic Rats. J Diabetes Res 2024; 2024:5497320. [PMID: 39329045 PMCID: PMC11424858 DOI: 10.1155/2024/5497320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 06/27/2024] [Accepted: 08/12/2024] [Indexed: 09/28/2024] Open
Abstract
Diabetes mellitus is a metabolic disorder. Synthetic antidiabetics are the commonly used treatment options associated with complications. The objective of this study was to explore the antioxidative and antidiabetic potential of Euphorbia helioscopia whole plant ethanolic extract using in vitro and in vivo models. For that purpose, the antioxidative potential was explored by using 2,2-diphenyl-1-picrylhydrazyl analysis. In vitro antidiabetic potential of the extract was evaluated using amylase inhibitory analysis. In vivo antidiabetic activity of the extract was assessed in diabetic rats using streptozotocin/nicotinamide (60 mg/kg/120 mg/kg) as an inducing agent. Metformin was used as standard. The results indicated the presence of significant quantities of phenolic 82.18 ± 1.28 mgg-1 gallic acid equivalent (GAE) and flavonoid 66.55±1.22 mgg-1 quercetin equivalent (QE) contents in the extract. Quantitation of phytoconstituents exhibited the presence of sinapic acid, myricetin, and quercetin using HPLC analysis. The extract inhibited α-amylase by 84.71%, and an antiglycemic potential of 50.34% was assessed in the OGTT assay. Biochemical analysis demonstrated a reduction in urea, creatinine, cholesterol, low-density lipoprotein, and alkaline phosphatase (p < 0.001) as compared to diabetic control rats at the dose of 500 mg/kg. An upregulation in the expressions of glucokinase, glucose transporter 4, peroxisome proliferator-activated receptor γ, and insulin-like growth factor was observed in treated rats in contrast to G6P expression, which was downregulated upon treatment. In conclusion, this study provided evidence of the antioxidative and antidiabetic potential of E. helioscopia whole plant ethanolic extract through in vitro and in vivo analysis and emphasized its promising role as a natural alternative.
Collapse
Affiliation(s)
- Ahmed Raza
- Faculty of PharmacyThe University of Lahore, Lahore 54000, Pakistan
| | | | - Sadia Hassan
- Department of Biomedical Engineering and SciencesSchool of Mechanical and Manufacturing EngineeringNational University of Science and Technology, Islamabad 24090, Pakistan
| | - Ali Sharif
- Department of PharmacologyFaculty of Pharmaceutical and Allied Health SciencesLahore College for Women University, Lahore 54000, Pakistan
| | - Bushra Akhtar
- Department of PharmacyUniversity of Agriculture, Faisalabad 38000, Pakistan
| | - Muhammad Furqan Akhtar
- Riphah Institute of Pharmaceutical SciencesRiphah International UniversityLahore Campus, Lahore 54000, Pakistan
| |
Collapse
|
17
|
Cabral RP, Ribeiro APD, Monte MG, Fujimori ASS, Tonon CR, Ferreira NF, Zanatti SG, Minicucci MF, Zornoff LAM, Paiva SARD, Polegato BF. Pera orange juice ( Citrus sinensis L. Osbeck) alters lipid metabolism and attenuates oxidative stress in the heart and liver of rats treated with doxorubicin. Heliyon 2024; 10:e36834. [PMID: 39263053 PMCID: PMC11388782 DOI: 10.1016/j.heliyon.2024.e36834] [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: 03/27/2024] [Revised: 08/14/2024] [Accepted: 08/22/2024] [Indexed: 09/13/2024] Open
Abstract
Background Doxorubicin (DOX) is a highly effective chemotherapy drug widely used to treat cancer, but its use is limited due to multisystemic toxicity. Lipid metabolism is also affected by doxorubicin. Orange juice can reduce dyslipidemia in other clinical situations and has already been shown to attenuate cardiotoxicity. Our aim is to evaluate the effects of Pera orange juice (Citrus sinensis L. Osbeck) on mitigating lipid metabolism imbalance, metabolic pathways, and DOX induced cytotoxic effects in the heart and liver. Methods Twenty-four male Wistar rats were allocated into 3 groups: Control (C); DOX (D); and DOX plus Pera orange juice (DOJ). DOJ received orange juice for 4 weeks, while C and D received water. At the end of each week, D and DOJ groups received 4 mg/kg/week DOX, intraperitoneal. At the end of 4 weeks animals were submitted to echocardiography and euthanasia. Results Animals treated with DOX decreased water intake and lost weight over time. At echocardiography, DOX treated rats presented morphologic alterations in the heart. DOX increased aspartate aminotransferase (AST), alanine aminotransferase (ALT), total cholesterol, high density lipoprotein (HDL), low-density lipoprotein (LDL), and triglycerides. It also reduced superoxide dismutase (SOD) activity, increased protein carbonylation in the heart and dihydroethidium (DHE) expression in the liver, decreased glucose transporter type 4 (GLUT4) and the nuclear receptor peroxisome proliferator-activated receptor γ (PPARγ1) in the heart, and reduced carnitine palmitoyltransferase I (CPT1) in the liver. Conclusion DOX caused dyslipidemia, liver and cardiac toxicity by increasing oxidative stress, and altered energy metabolic parameters in both organs. Despite not improving changes in left ventricular morphology, orange juice did attenuate oxidative stress and mitigate the metabolic effects of DOX.
Collapse
|
18
|
Shekho D, Mishra R, Kamal R, Bhatia R, Awasthi A. Breaking Barriers in Alzheimer's Disease: the Role of Advanced Drug Delivery Systems. AAPS PharmSciTech 2024; 25:207. [PMID: 39237748 DOI: 10.1208/s12249-024-02923-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 08/18/2024] [Indexed: 09/07/2024] Open
Abstract
Alzheimer's disease (AD), characterized by cognitive impairment, brain plaques, and tangles, is a global health concern affecting millions. It involves the build-up of amyloid-β (Aβ) and tau proteins, the formation of neuritic plaques and neurofibrillary tangles, cholinergic system dysfunction, genetic variations, and mitochondrial dysfunction. Various signaling pathways and metabolic processes are implicated in AD, along with numerous biomarkers used for diagnosis, risk assessment, and research. Despite these, there is no cure or effective treatment for AD. It is critically important to address this immediately to develop novel drug delivery systems (NDDS) capable of targeting the brain and delivering therapeutic agents to modulate the pathological processes of AD. This review summarizes AD, its pathogenesis, related signaling pathways, biomarkers, conventional treatments, the need for NDDS, and their application in AD treatment. It also covers preclinical, clinical, and ongoing trials, patents, and marketed AD formulations.
Collapse
Affiliation(s)
- Devank Shekho
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Ritika Mishra
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Raj Kamal
- Department of Quality Assurance, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Rohit Bhatia
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Ankit Awasthi
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, 142001, Punjab, India.
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India.
| |
Collapse
|
19
|
U N, R C T, R KR, Mahalingam G. Glucose transporters and their energy homeostasis function in various organs. VITAMINS AND HORMONES 2024; 128:1-47. [PMID: 40097247 DOI: 10.1016/bs.vh.2024.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Glucose transporters (GLUTs) belong to a membrane-protein family that essentially participates in easing the transportation and absorption of glucose molecules throughout the cellular membranes. From the brain to the eyes, each section delves into the intricate mechanisms of glucose uptake and utilization, shedding light on the unique adaptations and regulatory pathways in different anatomical structures. Beginning with the brain, known for its high energy demands, the chapter explicates the specialized GLUT expression patterns crucial for neuronal function and synaptic transmission. Moving to metabolic powerhouses like the liver, muscles, and adipose tissue, it elucidates the dynamic interplay of GLUT isoforms in energy storage, mobilization, and insulin responsiveness. Furthermore, the chapter navigates through the kidneys, lungs, skin, and reproductive organs, unveiling the diverse roles of GLUTs in renal glucose reabsorption, pulmonary-epithelial transportation, skin barrier associated functions, and gonadal development. It also explores the significance of placental GLUTs in fatal nutrient supply and the implications of cardiac GLUTs in myocardial energy metabolism. Additionally, it examines the intricate regulation of GLUTs at key barriers like the BBB (Blood-Brain Barrier) and placenta, as well as in endocrine glands such as the pancreas, adrenal medulla and thyroid. Moreover, it further elucidates the less-explored territories of GLUT expression in the bones, gastrointestinal tract, and ocular tissues like the retina, unraveling their implications in immune function, bone metabolism, intestinal glucose-sensing, and retinal physiology.
Collapse
Affiliation(s)
- Nithya U
- Department of Bio-Medical Sciences, School of Bio, sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Theijeswini R C
- Department of Bio-Medical Sciences, School of Bio, sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Karthick Raja R
- Department of Bio-Medical Sciences, School of Bio, sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Gayathri Mahalingam
- Department of Bio-Medical Sciences, School of Bio, sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India.
| |
Collapse
|
20
|
Li Y, Ye Z, Zhao Y, Xu B, Xue W, Wang Z, An R, Wang F, Wu R. Ling-gui-zhu-gan granules reduces obesity and ameliorates metabolic disorders by inducing white adipose tissue browning in obese mice. Front Physiol 2024; 15:1427722. [PMID: 39156823 PMCID: PMC11329929 DOI: 10.3389/fphys.2024.1427722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 07/17/2024] [Indexed: 08/20/2024] Open
Abstract
Background Ling-gui-zhu-gan (LGZG) formula has been demonstrated to effectively ameliorate the clinical symptoms of patients with obesity or metabolic syndrome. This study aimed to explore both the effect and the underlying mechanisms of LGZG against obesity. Methods Male C57BL/6N mice were randomized into four groups (n = 8): normal control (NC), obese (OB), metformin (Met), and LGZG. After 8 weeks of gavage administration, the pharmacological effects of LGZG on obesity and metabolism were investigated using biochemical parameters, histomorphological examination, and lipidomics techniques. Pivotal factors associated with white adipose tissue browning were evaluated using quantitative real-time polymerase chain reaction and western blotting. Results The results revealed that LGZG reduced the levels of obesity markers, including body weights, body fat mass and food intake in obese mice. Further evaluations highlighted that LGZG restored glucose homeostasis and significantly improved insulin sensitivity in obese mice. Importantly, LGZG could adjust serum lipid profiles and regulate the lipidomic spectrum of intestinal contents, with noticeable shifts in the levels of certain lipids, particularly diacylglycerols and monoacylglycerols. Histopathological examinations of LGZG-treated mice also revealed more favorable adipose tissue structures than their obese counterparts. Furthermore, we found that LGZG upregulated the expression of several key thermogenesis-related factors, such as UCP1, PRDM16, PGC-1α, PPARα, PPARγ, CTBP1, and CTBP2 in white adipose tissues. Conclusion Our findings position LGZG as a novel strategy for preventing obesity and improving metabolic health.
Collapse
Affiliation(s)
- Yuxiu Li
- Department of Endocrinology, Guang’anmen Hospital South Campus, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zimengwei Ye
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yi Zhao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Bingrui Xu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Wanying Xue
- College of Integrative Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Zhufeng Wang
- Department of Endocrinology, Guang’anmen Hospital South Campus, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ran An
- Department of Endocrinology, Guang’anmen Hospital South Campus, China Academy of Chinese Medical Sciences, Beijing, China
| | - Fan Wang
- Department of Endocrinology, Guang’anmen Hospital South Campus, China Academy of Chinese Medical Sciences, Beijing, China
| | - Rui Wu
- Department of Endocrinology, Guang’anmen Hospital South Campus, China Academy of Chinese Medical Sciences, Beijing, China
- Department of Endocrinology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
21
|
Wang M, Yang N, Guo W, Yang Y, Bao B, Zhang X, Zhang D. RNAi-mediated glucose transporter 4 (Glut4) silencing inhibits ovarian development and enhances deltamethrin-treated energy depletion in Locusta migratoria. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2024; 203:106014. [PMID: 39084805 DOI: 10.1016/j.pestbp.2024.106014] [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: 04/29/2024] [Revised: 06/22/2024] [Accepted: 07/02/2024] [Indexed: 08/02/2024]
Abstract
Energy metabolism is essential for insect development, reproduction and detoxification. Insects often reallocate energy and resources to manage external stress, balancing the demands of detoxification and reproduction. Glucose transport 4 (Glut4), a glucose transporter, is involved in glucose and lipid metabolism. However, the specific molecular mechanism of Glut4 in insect reproduction, and its role in the response to insecticide-induced oxidative stress remain unclear. In this study, LmGlut4 was identified and analyzed in Locusta migratoria. Silencing of LmGlut4 significantly reduced vitellogenin (Vg) biosynthesis in the fat body and Vg absorption by oocytes, ultimately hindering ovarian development and oocyte maturation. Knockdown of LmGlut4 also inhibited the biosynthesis of key insect hormones, such as juvenile hormone (JH), 20-hydroxyecdysone (20E) and insulin. Furthermore, LmGlut4 knockdown led to reduced triglyceride (TG) and glycogen content in the fat body and ovary, as well as decreased capacity for trehalose biosynthesis in adipocytes. Additionally, dsLmGlut4-treated locusts showed heightened sensitivity to deltamethrin, leading to increased triglyceride depletion during detoxification. This study sheds light on the biological function of LmGlut4 in the ovary and provides potential target genes for exploring biological pest management strategies.
Collapse
Affiliation(s)
- Mingjun Wang
- Key Laboratory of Zoological Systematics and Application of Hebei Province, Institute of Life Sciences and Green Development, College of Life Sciences, Hebei University, Baoding 071002, China
| | - Ningxin Yang
- Key Laboratory of Zoological Systematics and Application of Hebei Province, Institute of Life Sciences and Green Development, College of Life Sciences, Hebei University, Baoding 071002, China
| | - Wenhui Guo
- Key Laboratory of Zoological Systematics and Application of Hebei Province, Institute of Life Sciences and Green Development, College of Life Sciences, Hebei University, Baoding 071002, China
| | - Yong Yang
- Key Laboratory of Zoological Systematics and Application of Hebei Province, Institute of Life Sciences and Green Development, College of Life Sciences, Hebei University, Baoding 071002, China
| | - Bowen Bao
- Key Laboratory of Zoological Systematics and Application of Hebei Province, Institute of Life Sciences and Green Development, College of Life Sciences, Hebei University, Baoding 071002, China
| | - Xiaohong Zhang
- Key Laboratory of Zoological Systematics and Application of Hebei Province, Institute of Life Sciences and Green Development, College of Life Sciences, Hebei University, Baoding 071002, China.
| | - Daochuan Zhang
- Key Laboratory of Zoological Systematics and Application of Hebei Province, Institute of Life Sciences and Green Development, College of Life Sciences, Hebei University, Baoding 071002, China; Hebei Basic Science Center for Biotic Interaction, Hebei University, Baoding 071002, China.
| |
Collapse
|
22
|
Ivanova N, Atanasova M, Terzieva D, Georgieva K, Tchekalarova J. The Role of Piromelatine on Peripheral and Hippocampal Insulin Resistance in Rat Offspring Exposed to Chronic Maternal Stress. Int J Mol Sci 2024; 25:7022. [PMID: 39000130 PMCID: PMC11241293 DOI: 10.3390/ijms25137022] [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: 05/31/2024] [Revised: 06/23/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024] Open
Abstract
Prenatal stress (PNS), which alters the hypothalamic-pituitary-adrenal axis function in the offspring, predisposes to insulin resistance (IR) in later life and is associated with numerous disorders, including cognitive and memory impairments. At present, our main goal is to assess the effects of chronic piromelatine (Pir) administration, a melatonin analogue, on PNS-provoked IR in the periphery and the hippocampus in male and female offspring. Pregnant Sprague-Dawley rats were exposed to chronic stress (one short-term stressor on a daily basis and one long-term stressor on a nightly basis) from the first gestation week until birth. Vehicle or Pir 20 mg/kg were administered intraperitoneally for 21 days. Plasma glucose, serum insulin levels, and the homeostasis model assessment of insulin resistance (HOMA-IR) were determined as markers of peripheral IR. For the hippocampal IR assessment, insulin receptors (IRs) and glucose transporter 4 (GLUT4) were examined. Prenatally stressed offspring of both sexes indicated enhanced plasma glucose and serum insulin concentrations, increased HOMA-IR, and decreased hippocampal GLUT4 only in male rats. The PNS-induced changes were corrected by chronic treatment with Pir. The present results suggest that the melatoninergic compound Pir exerts beneficial effects on altered glucose/insulin homeostasis in PNS-exposed offspring.
Collapse
Affiliation(s)
- Natasha Ivanova
- Institute of Neurobiology, Bulgarian Academy of Sciences (BAS), 1113 Sofia, Bulgaria
| | - Milena Atanasova
- Department of Biology, Medical University of Pleven, 5800 Pleven, Bulgaria;
| | - Dora Terzieva
- Department of Clinical Laboratory, Medical Faculty, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria;
| | - Katerina Georgieva
- Department of Physiology, Medical University of Plovdiv, 5800 Pleven, Bulgaria;
| | - Jana Tchekalarova
- Institute of Neurobiology, Bulgarian Academy of Sciences (BAS), 1113 Sofia, Bulgaria
| |
Collapse
|
23
|
Yesuf HA, Molla MD, Malik T, Seyoum Wendimagegn Z, Yimer Y. MicroRNA-29-mediated cross-talk between metabolic organs in the pathogenesis of diabetes mellitus and its complications: A narrative review. Cell Biochem Funct 2024; 42:e4053. [PMID: 38773932 DOI: 10.1002/cbf.4053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 04/27/2024] [Accepted: 05/07/2024] [Indexed: 05/24/2024]
Abstract
Diabetes mellitus (DM) is a heterogeneous group of disorders characterized by hyperglycemia. Microribonucleic acids (microRNAs) are noncoding RNA molecules synthesized in the nucleus, modified, and exported to the extracellular environment to bind to their complementary target sequences. It regulates protein synthesis in the targeted cells by inhibiting translation or triggering the degradation of the target messenger. MicroRNA-29 is one of noncoding RNA that can be secreted by adipose tissue, hepatocytes, islet cells, and brain cells. The expression level of the microRNA-29 family in several metabolic organs is regulated by body weight, blood concentrations of inflammatory mediators, serum glucose levels, and smoking habits. Several experimental studies have demonstrated the effect of microRNA-29 on the expression of target genes involved in glucose metabolism, insulin synthesis and secretion, islet cell survival, and proliferation. These findings shed new light on the role of microRNA-29 in the pathogenesis of diabetes and its complications, which plays a vital role in developing appropriate therapies. Different molecular pathways have been proposed to explain how microRNA-29 promotes the development of diabetes and its complications. However, to the best of our knowledge, no published review article has summarized the molecular mechanism of microRNA-29-mediated initiation of DM and its complications. Therefore, this narrative review aims to summarize the role of microRNA-29-mediated cross-talk between metabolic organs in the pathogenesis of diabetes and its complications.
Collapse
Affiliation(s)
- Hassen Ahmed Yesuf
- Department of Biomedical Science, School of Medicine, College of Health Sciences, Woldia University, Woldia, Ethiopia
| | - Meseret Derbew Molla
- Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
- Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Tabarak Malik
- Department of Biomedical Sciences, Institute of Health, Jimma University, Jimma, Ethiopia
- Division of Research and Development, Lovely Professional University, Phagwara, India
| | - Zeru Seyoum Wendimagegn
- Department of Biomedical Science, School of Medicine, College of Health Sciences, Woldia University, Woldia, Ethiopia
| | - Yadelew Yimer
- Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
24
|
Zhou Y, Zhang S, Jia Y, Wang X, Liu Y, Zhang H, Yuan Z, Han Y, Weng Q. Regulation and Role of Adiponectin Secretion in Rat Ovarian Granulosa Cells. Int J Mol Sci 2024; 25:5155. [PMID: 38791193 PMCID: PMC11120769 DOI: 10.3390/ijms25105155] [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: 03/28/2024] [Revised: 04/25/2024] [Accepted: 05/03/2024] [Indexed: 05/26/2024] Open
Abstract
Adiponectin is an important adipokine involved in glucose and lipid metabolism, but its secretion and potential role in regulating glucose utilization during ovarian development remains unclear. This study aims to investigate the mechanism and effects of follicle-stimulating hormones (FSHs) on adiponectin secretion and its following impact on glucose transport in the granulosa cells of rat ovaries. A range of experimental techniques were utilized to test our research, including immunoblotting, immunohistochemistry, immunofluorescence, ELISA, histological staining, real-time quantitative PCR, and transcriptome analysis. The immunohistochemistry results indicated that adiponectin was primarily located in the granulosa cells of rat ovaries. In primary granulosa cells cultured in vitro, both Western blot and immunofluorescence assays demonstrated that FSH significantly induced adiponectin secretion within 2 h of incubation, primarily via the PKA signaling pathway rather than the PI3K/AKT pathway. Concurrently, the addition of the AdipoR1/AdipoR2 dual agonist AdipoRon to the culture medium significantly stimulated the protein expression of GLUT1 in rat granulosa cells, resulting in enhanced glucose absorption. Consistent with these in vitro findings, rats injected with eCG (which shares structural and functional similarities with FSH) exhibited significantly increased adiponectin levels in both the ovaries and blood. Moreover, there was a notable elevation in mRNA and protein levels of AdipoRs and GLUTs following eCG administration. Transcriptomic analysis further revealed a positive correlation between the expression of the intraovarian adiponectin system and glucose transporter. The present study represents a novel investigation, demonstrating that FSH stimulates adiponectin secretion in ovarian granulosa cells through the PKA signaling pathway. This mechanism potentially influences glucose transport (GLUT1) and utilization within the ovaries.
Collapse
Affiliation(s)
- Yue Zhou
- College of Biological Science and Technology, Beijing Forestry University, Beijing 100083, China; (Y.Z.); (Y.J.); (X.W.); (Y.L.); (H.Z.); (Z.Y.)
| | - Shuhao Zhang
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China;
| | - Yurong Jia
- College of Biological Science and Technology, Beijing Forestry University, Beijing 100083, China; (Y.Z.); (Y.J.); (X.W.); (Y.L.); (H.Z.); (Z.Y.)
| | - Xi Wang
- College of Biological Science and Technology, Beijing Forestry University, Beijing 100083, China; (Y.Z.); (Y.J.); (X.W.); (Y.L.); (H.Z.); (Z.Y.)
| | - Yuning Liu
- College of Biological Science and Technology, Beijing Forestry University, Beijing 100083, China; (Y.Z.); (Y.J.); (X.W.); (Y.L.); (H.Z.); (Z.Y.)
| | - Haolin Zhang
- College of Biological Science and Technology, Beijing Forestry University, Beijing 100083, China; (Y.Z.); (Y.J.); (X.W.); (Y.L.); (H.Z.); (Z.Y.)
| | - Zhengrong Yuan
- College of Biological Science and Technology, Beijing Forestry University, Beijing 100083, China; (Y.Z.); (Y.J.); (X.W.); (Y.L.); (H.Z.); (Z.Y.)
| | - Yingying Han
- College of Biological Science and Technology, Beijing Forestry University, Beijing 100083, China; (Y.Z.); (Y.J.); (X.W.); (Y.L.); (H.Z.); (Z.Y.)
| | - Qiang Weng
- College of Biological Science and Technology, Beijing Forestry University, Beijing 100083, China; (Y.Z.); (Y.J.); (X.W.); (Y.L.); (H.Z.); (Z.Y.)
| |
Collapse
|
25
|
Li T, Hu X, Fan L, Yang Y, He K. Myricanol improves metabolic profiles in dexamethasone induced lipid and protein metabolism disorders in mice. Biomed Pharmacother 2024; 174:116557. [PMID: 38583337 DOI: 10.1016/j.biopha.2024.116557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 03/27/2024] [Accepted: 04/04/2024] [Indexed: 04/09/2024] Open
Abstract
Myricanol (MY) is one of the main active components from bark of Myrica Rubra. It is demonstrated that MY rescues dexamethasone (DEX)-induced muscle dysfunction via activating silent information regulator 1 (SIRT1) and increasing adenosine 5'-monophosphate-activated protein kinase (AMPK) phosphorylation. Since SIRT1 and AMPK are widely involved in the metabolism of nutrients, we speculated that MY may exert beneficial effects on DEX-induced metabolic disorders. This study for the first time applied widely targeted metabolomics to investigate the beneficial effects of MY on glucose, lipids, and protein metabolism in DEX-induced metabolic abnormality in mice. The results showed that MY significantly reversed DEX-induced soleus and gastrocnemius muscle weight loss, muscle fiber damage, and muscle strength loss. MY alleviated DEX-induced metabolic disorders by increasing SIRT1 and glucose transporter type 4 (GLUT4) expressions. Additionally, myricanol prevented muscle cell apoptosis and atrophy by inhibiting caspase 3 cleavages and muscle ring-finger protein-1 (MuRF1) expression. Metabolomics showed that MY treatment reversed the serum content of carnitine ph-C1, palmitoleic acid, PS (16:0_17:0), PC (14:0_20:5), PE (P-18:1_16:1), Cer (t18:2/38:1(2OH)), four amino acids and their metabolites, and 16 glycerolipids in DEX mice. Kyoto encyclopedia of genes and genomes (KEGG) and metabolic set enrichment analysis (MSEA) analysis revealed that MY mainly affected metabolic pathways, glycerolipid metabolism, lipolysis, fat digestion and absorption, lipid and atherosclerosis, and cholesterol metabolism pathways through regulation of metabolites involved in glutathione, butanoate, vitamin B6, glycine, serine and threonine, arachidonic acid, and riboflavin metabolism. Collectively, MY can be used as an attractive therapeutic agent for DEX-induced metabolic abnormalities.
Collapse
Affiliation(s)
- Tiandan Li
- Hunan Provincial Key Laboratory of Dong Medicine, Hunan Provincial Key Laboratory for Synthetic Biology of Traditional Chinese Medicine, School of Pharmaceutical Science, Hunan University of Medicine, Huaihua, Hunan 418000, China
| | - Xiaochao Hu
- Hunan Provincial Key Laboratory of Dong Medicine, Hunan Provincial Key Laboratory for Synthetic Biology of Traditional Chinese Medicine, School of Pharmaceutical Science, Hunan University of Medicine, Huaihua, Hunan 418000, China
| | - Lingyang Fan
- Hunan Provincial Key Laboratory of Dong Medicine, Hunan Provincial Key Laboratory for Synthetic Biology of Traditional Chinese Medicine, School of Pharmaceutical Science, Hunan University of Medicine, Huaihua, Hunan 418000, China
| | - Yong Yang
- chool of Pharmacy, Hunan University of Traditional Chinese Medicine, Changsha, Hunan 410208, China.
| | - Kai He
- Hunan Provincial Key Laboratory of Dong Medicine, Hunan Provincial Key Laboratory for Synthetic Biology of Traditional Chinese Medicine, School of Pharmaceutical Science, Hunan University of Medicine, Huaihua, Hunan 418000, China.
| |
Collapse
|
26
|
Mallardo M, Daniele A, Musumeci G, Nigro E. A Narrative Review on Adipose Tissue and Overtraining: Shedding Light on the Interplay among Adipokines, Exercise and Overtraining. Int J Mol Sci 2024; 25:4089. [PMID: 38612899 PMCID: PMC11012884 DOI: 10.3390/ijms25074089] [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: 03/18/2024] [Revised: 03/30/2024] [Accepted: 04/01/2024] [Indexed: 04/14/2024] Open
Abstract
Lifestyle factors, particularly physical inactivity, are closely linked to the onset of numerous metabolic diseases. Adipose tissue (AT) has been extensively studied for various metabolic diseases such as obesity, type 2 diabetes, and immune system dysregulation due to its role in energy metabolism and regulation of inflammation. Physical activity is increasingly recognized as a powerful non-pharmacological tool for the treatment of various disorders, as it helps to improve metabolic, immune, and inflammatory functions. However, chronic excessive training has been associated with increased inflammatory markers and oxidative stress, so much so that excessive training overload, combined with inadequate recovery, can lead to the development of overtraining syndrome (OTS). OTS negatively impacts an athlete's performance capabilities and significantly affects both physical health and mental well-being. However, diagnosing OTS remains challenging as the contributing factors, signs/symptoms, and underlying maladaptive mechanisms are individualized, sport-specific, and unclear. Therefore, identifying potential biomarkers that could assist in preventing and/or diagnosing OTS is an important objective. In this review, we focus on the possibility that the endocrine functions of AT may have significant implications in the etiopathogenesis of OTS. During physical exercise, AT responds dynamically, undergoing remodeling of endocrine functions that influence the production of adipokines involved in regulating major energy and inflammatory processes. In this scenario, we will discuss exercise about its effects on AT activity and metabolism and its relevance to the prevention and/or development of OTS. Furthermore, we will highlight adipokines as potential markers for diagnosing OTS.
Collapse
Affiliation(s)
- Marta Mallardo
- Department of Molecular and Biotechnological Medicine, University of Naples “Federico II”, 80131 Naples, Italy;
- CEINGE-Biotechnologies Advances S.c.a r.l., Via G. Salvatore 486, 80145 Naples, Italy;
| | - Aurora Daniele
- Department of Molecular and Biotechnological Medicine, University of Naples “Federico II”, 80131 Naples, Italy;
- CEINGE-Biotechnologies Advances S.c.a r.l., Via G. Salvatore 486, 80145 Naples, Italy;
| | - Giuseppe Musumeci
- Department of Biomedical and Biotechnological Sciences, Anatomy, Histology and Movement Sciences Section, School of Medicine, University of Catania, Via S. Sofia 87, 95123 Catania, Italy
- Research Center on Motor Activities (CRAM), University of Catania, 95123 Catania, Italy
| | - Ersilia Nigro
- CEINGE-Biotechnologies Advances S.c.a r.l., Via G. Salvatore 486, 80145 Naples, Italy;
- Department of Pharmaceutical, Biological, Environmental Sciences and Technologies, University of Campania “Luigi Vanvitelli”, Via G. Vivaldi 42, 81100 Caserta, Italy
| |
Collapse
|
27
|
Malacarne C, Giagnorio E, Chirizzi C, Cattaneo M, Saraceno F, Cavalcante P, Bonanno S, Mantegazza R, Moreno-Manzano V, Lauria G, Metrangolo P, Bombelli FB, Marcuzzo S. FM19G11-loaded nanoparticles modulate energetic status and production of reactive oxygen species in myoblasts from ALS mice. Biomed Pharmacother 2024; 173:116380. [PMID: 38447450 DOI: 10.1016/j.biopha.2024.116380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 02/22/2024] [Accepted: 02/29/2024] [Indexed: 03/08/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease affecting motor neurons. Considerable evidence indicates that early skeletal muscle atrophy plays a crucial role in the disease pathogenesis, leading to an altered muscle-motor neuron crosstalk that, in turn, may contribute to motor neuron degeneration. Currently, there is no effective treatment for ALS, highlighting the need to dig deeper into the pathological mechanisms for developing innovative therapeutic strategies. FM19G11 is a novel drug able to modulate the global cellular metabolism, but its effects on ALS skeletal muscle atrophy and mitochondrial metabolism have never been evaluated, yet. This study investigated whether FM19G11-loaded nanoparticles (NPs) may affect the bioenergetic status in myoblasts isolated from G93A-SOD1 mice at different disease stages. We found that FM19G1-loaded NP treatment was able to increase transcriptional levels of Akt1, Akt3, Mef2a, Mef2c and Ucp2, which are key genes associated with cell proliferation (Akt1, Akt3), muscle differentiation (Mef2c), and mitochondrial activity (Ucp2), in G93A-SOD1 myoblasts. These cells also showed a significant reduction of mitochondrial area and networks, in addition to decreased ROS production after treatment with FM19G11-loaded NPs, suggesting a ROS clearance upon the amelioration of mitochondrial dynamics. Our overall findings demonstrate a significant impact of FM19G11-loaded NPs on muscle cell function and bioenergetic status in G93A-SOD1 myoblasts, thus promising to open new avenues towards possible adoption of FM19G11-based nanotherapies to slow muscle degeneration in the frame of ALS and muscle disorders.
Collapse
Affiliation(s)
- Claudia Malacarne
- Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, Milan 20133, Italy
| | - Eleonora Giagnorio
- Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, Milan 20133, Italy.
| | - Cristina Chirizzi
- Neuroradiology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, Milan 20133, Italy; Laboratory of Supramolecular and Bio-Nanomaterials (SupraBioNano Lab), Department of Chemistry, Materials and Chemical Engineering, "Giulio Natta", Politecnico di Milano, Milan 20131, Italy
| | - Marco Cattaneo
- Neuroalgology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, Milan 20133, Italy; PhD Program in Pharmacological Biomolecular Sciences, Experimental and Clinical, University of Milano, Via G.Balzaretti 9, Milan 20133, Italy
| | - Fulvia Saraceno
- Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, Milan 20133, Italy; Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11/a, Parma 43124, Italy
| | - Paola Cavalcante
- Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, Milan 20133, Italy
| | - Silvia Bonanno
- Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, Milan 20133, Italy
| | - Renato Mantegazza
- Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, Milan 20133, Italy
| | - Victoria Moreno-Manzano
- Neuronal and Tissue Regeneration Laboratory, Prince Felipe Research Center, Carrer d´Eduardo Primo Yúfera 3, Valencia 46012, Spain
| | - Giuseppe Lauria
- Neuroalgology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, Milan 20133, Italy; Department of Medical Biotechnology and Translational Medicine, University of Milano, Milan 20133, Italy
| | - Pierangelo Metrangolo
- Laboratory of Supramolecular and Bio-Nanomaterials (SupraBioNano Lab), Department of Chemistry, Materials and Chemical Engineering, "Giulio Natta", Politecnico di Milano, Milan 20131, Italy; Brains Lab, Joint Research Platform, Fondazione IRCCS Istituto Neurologico Carlo Besta-Politecnico di Milano, Via Celoria 11, 20133 Milan, Italy
| | - Francesca Baldelli Bombelli
- Laboratory of Supramolecular and Bio-Nanomaterials (SupraBioNano Lab), Department of Chemistry, Materials and Chemical Engineering, "Giulio Natta", Politecnico di Milano, Milan 20131, Italy
| | - Stefania Marcuzzo
- Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, Milan 20133, Italy; Brains Lab, Joint Research Platform, Fondazione IRCCS Istituto Neurologico Carlo Besta-Politecnico di Milano, Via Celoria 11, 20133 Milan, Italy.
| |
Collapse
|
28
|
Odongo K, Abe A, Kawasaki R, Kawabata K, Ashida H. Two Prenylated Chalcones, 4-Hydroxyderricin, and Xanthoangelol Prevent Postprandial Hyperglycemia by Promoting GLUT4 Translocation via the LKB1/AMPK Signaling Pathway in Skeletal Muscle Cells. Mol Nutr Food Res 2024; 68:e2300538. [PMID: 38267744 DOI: 10.1002/mnfr.202300538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/30/2023] [Indexed: 01/26/2024]
Abstract
SCOPE Stimulation of glucose uptake in the skeletal muscle is crucial for the prevention of postprandial hyperglycemia. Insulin and certain polyphenols enhance glucose uptake through the translocation of glucose transporter 4 (GLUT4) in the skeletal muscle. The previous study reports that prenylated chalcones, 4-hydroxyderricin (4-HD), and xanthoangelol (XAG) promote glucose uptake and GLUT4 translocation in L6 myotubes, but their underlying molecular mechanism remains unclear. This study investigates the mechanism in L6 myotubes and confirms antihyperglycemia by 4-HD and XAG. METHODS AND RESULTS In L6 myotubes, 4-HD and XAG promote glucose uptake and GLUT4 translocation through the activation of adenosine monophosphate-activated protein kinase (AMPK) and liver kinase B1 (LKB1) signaling pathway without activating phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) and Janus kinases (JAKs)/signal transducers and activators of transcriptions (STATs) pathways. Moreover, Compound C, an AMPK-specific inhibitor, as well as siRNA targeting AMPK and LKB1 completely canceled 4-HD and XAG-increased glucose uptake. Consistently, oral administration of 4-HD and XAG to male ICR mice suppresses acute hyperglycemia in an oral glucose tolerance test. CONCLUSION In conclusion, LKB1/AMPK pathway and subsequent GLUT4 translocation in skeletal muscle cells are involved in Ashitaba chalcone-suppressed acute hyperglycemia.
Collapse
Affiliation(s)
- Kevin Odongo
- Department of Agrobioscience, Graduate School of Agricultural Science, Kobe University, Kobe, 657-8501, Japan
| | - Ayane Abe
- Department of Agrobioscience, Graduate School of Agricultural Science, Kobe University, Kobe, 657-8501, Japan
| | - Rina Kawasaki
- Department of Agrobioscience, Graduate School of Agricultural Science, Kobe University, Kobe, 657-8501, Japan
| | - Kyuichi Kawabata
- Faculty of Clinical Nutrition and Dietetics, Konan Women's University, Kobe, 658-0001, Japan
| | - Hitoshi Ashida
- Department of Agrobioscience, Graduate School of Agricultural Science, Kobe University, Kobe, 657-8501, Japan
| |
Collapse
|
29
|
Swain SK, Dash UC, Kanhar S, Sahoo AK. Network pharmacology-based elucidation of bioactive compounds and experimental exploration of antidiabetic mechanisms of Hydrolea zeylanica. Cell Signal 2024; 114:110999. [PMID: 38052370 DOI: 10.1016/j.cellsig.2023.110999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 11/21/2023] [Accepted: 12/01/2023] [Indexed: 12/07/2023]
Abstract
This investigation systematically explored the underlying antidiabetic mechanism of Hydrolea zeylanica (HZH) by the approach of network pharmacology and experimental validation in restoring glucose homeostasis, and inflammation in high fat diet fed-streptozotocin (HFD/STZ)-induced type II diabetes (T2DM) rats. Network pharmacology analysis was conducted on 32 bioactive components of HZH. In silico ADME prediction, and physicochemical analysis of 32 compounds were used to assess their drug-likeness. Common targets between selected compounds, and T2DM were subjected for GO enrichment. Compound-target-pathway network was predicted with selected compounds and targets. HZH (300 and 400 mg/kg) were considered for GLUTs expression, and inflammation cytokines in T2DM rats. Network pharmacology showed the core relationship between 13 selected compounds, and 194 key target genes in insulin resistance, type II diabetes mellitus, insulin signaling pathways in T2DM. AKT1, AKT2, GSK3B, IL6, INSR, MAPK8, PPARA, GLUT1, GLUT2, GLUT4 were observed as the key targets in PPI network. HZH-400 significantly restored glucose homeostasis, and inflammatory markers in T2DM rats. It altered GLUT2, GLUT4 expression in liver, and skeletal muscle to normal. Bioactive compounds of HZH were found to control blood sugar level by modulating insulin resistance, type II diabetes mellitus, insulin signaling pathways, and glucose homeostasis, which in turn improved glucose uptake, insulin production in diabetes as shown in network pharmacology and glucose transporter expression studies.
Collapse
Affiliation(s)
- Sandeep Kumar Swain
- Regional Plant Resource Centre, Medicinal & Aromatic Plant Division, Forest, Environment & Climate Change Department, Govt. of Odisha, Nayapalli, Bhubaneswar 751015, India
| | - Umesh Chandra Dash
- Regional Plant Resource Centre, Medicinal & Aromatic Plant Division, Forest, Environment & Climate Change Department, Govt. of Odisha, Nayapalli, Bhubaneswar 751015, India
| | - Satish Kanhar
- Regional Plant Resource Centre, Medicinal & Aromatic Plant Division, Forest, Environment & Climate Change Department, Govt. of Odisha, Nayapalli, Bhubaneswar 751015, India
| | - Atish Kumar Sahoo
- Regional Plant Resource Centre, Medicinal & Aromatic Plant Division, Forest, Environment & Climate Change Department, Govt. of Odisha, Nayapalli, Bhubaneswar 751015, India.
| |
Collapse
|
30
|
Wen J, Chen C. From Energy Metabolic Change to Precision Therapy: a Holistic View of Energy Metabolism in Heart Failure. J Cardiovasc Transl Res 2024; 17:56-70. [PMID: 37450209 DOI: 10.1007/s12265-023-10412-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 07/04/2023] [Indexed: 07/18/2023]
Abstract
Heart failure (HF) is a complex and multifactorial disease that affects millions of people worldwide. It is characterized by metabolic disturbances of substrates such as glucose, fatty acids (FAs), ketone bodies, and amino acids, which lead to changes in cardiac energy metabolism pathways. These metabolic alterations can directly or indirectly promote myocardial remodeling, thereby accelerating the progression of HF, resulting in a vicious cycle of worsening symptoms, and contributing to the increased hospitalization and mortality among patients with HF. In this review, we summarized the latest researches on energy metabolic profiling in HF and provided the related translational therapeutic strategies for this devastating disease. By taking a holistic approach to understanding energy metabolism changes in HF, we hope to provide comprehensive insights into the pathophysiology of this challenging condition and identify novel precise targets for the development of more effective treatments.
Collapse
Affiliation(s)
- Jianpei Wen
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Avenue, Wuhan, 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China
| | - Chen Chen
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Avenue, Wuhan, 430030, China.
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China.
| |
Collapse
|
31
|
Yang J, Shay C, Saba NF, Teng Y. Cancer metabolism and carcinogenesis. Exp Hematol Oncol 2024; 13:10. [PMID: 38287402 PMCID: PMC10826200 DOI: 10.1186/s40164-024-00482-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 01/22/2024] [Indexed: 01/31/2024] Open
Abstract
Metabolic reprogramming is an emerging hallmark of cancer cells, enabling them to meet increased nutrient and energy demands while withstanding the challenging microenvironment. Cancer cells can switch their metabolic pathways, allowing them to adapt to different microenvironments and therapeutic interventions. This refers to metabolic heterogeneity, in which different cell populations use different metabolic pathways to sustain their survival and proliferation and impact their response to conventional cancer therapies. Thus, targeting cancer metabolic heterogeneity represents an innovative therapeutic avenue with the potential to overcome treatment resistance and improve therapeutic outcomes. This review discusses the metabolic patterns of different cancer cell populations and developmental stages, summarizes the molecular mechanisms involved in the intricate interactions within cancer metabolism, and highlights the clinical potential of targeting metabolic vulnerabilities as a promising therapeutic regimen. We aim to unravel the complex of metabolic characteristics and develop personalized treatment approaches to address distinct metabolic traits, ultimately enhancing patient outcomes.
Collapse
Affiliation(s)
- Jianqiang Yang
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, 201 Dowman Dr, Atlanta, GA, 30322, USA
| | - Chloe Shay
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30322, USA
| | - Nabil F Saba
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, 201 Dowman Dr, Atlanta, GA, 30322, USA
| | - Yong Teng
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, 201 Dowman Dr, Atlanta, GA, 30322, USA.
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30322, USA.
| |
Collapse
|
32
|
Montserrat-de la Paz S, D Miguel-Albarreal A, Gonzalez-de la Rosa T, Millan-Linares MC, Rivero-Pino F. Protein-based nutritional strategies to manage the development of diabetes: evidence and challenges in human studies. Food Funct 2023; 14:9962-9973. [PMID: 37873616 DOI: 10.1039/d3fo02466k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Type 2 diabetes mellitus (T2DM) is one of the most prevalent diseases in modern society, governed by both genetic and environmental factors, such as nutritional habits. This metabolic disorder is characterized by insulin resistance, which is related to high blood glucose levels, implying negative health effects in humans, hindering the healthy ageing of people. The relationship between food and health is clear, and the ingestion of specific nutrients modulates some physiological processes, potentially implying biologically relevant changes, which can translate into a health benefit. This review aims to summarize human studies published in which the purpose was to investigate the effect of protein ingestion (in native state or as hydrolysates) on human metabolism. Overall, several studies showed how protein ingestion might induce a decrease of glucose concentration in the postprandial state (area under the curve), although it is highly dependent on the source and the dose. Other studies showed no biological effects upon protein consumption, mostly with fish-derived products. In addition, the major challenges and perspectives in this research field are highlighted, suggesting the future directions, towards which scientists should focus on. The dietary intake of proteins has been proven to likely exert a beneficial effect on diabetes-related parameters, which can have a biological relevance in the prevention and pre-treatment of diabetes. However, the number of well-designed human studies carried out to date to demonstrate the effects of specific proteins or protein hydrolysates in vivo is still scarce.
Collapse
Affiliation(s)
- Sergio Montserrat-de la Paz
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, University of Seville, Av. Sanchez Pizjuan s/n, 41009 Seville, Spain.
| | - Antonio D Miguel-Albarreal
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, University of Seville, Av. Sanchez Pizjuan s/n, 41009 Seville, Spain.
| | - Teresa Gonzalez-de la Rosa
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, University of Seville, Av. Sanchez Pizjuan s/n, 41009 Seville, Spain.
| | - Maria C Millan-Linares
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, University of Seville, Av. Sanchez Pizjuan s/n, 41009 Seville, Spain.
| | - Fernando Rivero-Pino
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, University of Seville, Av. Sanchez Pizjuan s/n, 41009 Seville, Spain.
| |
Collapse
|
33
|
Ding YY, Lan J, Fang Y, Pan Y, Gu Z, Xue J, Yang Y, Jiang M, Ge Y, Shen Q. Dityrosine Aggravates Hepatic Insulin Resistance in Obese Mice by Altering Gut Microbiota and the LPS/TLR4/NF-κB Inflammatory Pathway. Mol Nutr Food Res 2023; 67:e2300373. [PMID: 37726250 DOI: 10.1002/mnfr.202300373] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 07/28/2023] [Indexed: 09/21/2023]
Abstract
SCOPE Dityrosine is the main product of protein oxidation, which has been proved to be a threat to human health. This study aims to investigate whether dityrosine exacerbates insulin resistance by inducing gut flora disturbance and associated inflammatory responses. METHODS AND RESULTS Mice fed with normal diet or high-fat diet (HFD) received daily gavage of dityrosine (320 µg kg-1 BW) or saline for consecutive 13 weeks. The effects of dityrosine on gut microbiota are verified by in vitro fermentation using fecal microbiota from db/m mice and db/db mice. As a result, dityrosine causes the insulin resistance in mice fed normal diet, and aggravates the effects of HFD on insulin sensitivity. Dityrosine increases the levels of lipopolysaccharide (LPS), lipopolysaccharide-binding protein (LBP), toll-like receptor 4 (TLR4), nuclear factor kappa-B (NF-κB), tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and interleukin-8 (IL-8) but decreases levels of interleukin-10 (IL-10) in the plasma of CON and HFD-fed mice. The changes of gut flora composition caused by dityrosine are significantly correlated with the changes of inflammatory biomarkers. CONCLUSION The effects of dityrosine on insulin resistance may be attributed to the reshaping of the gut microbiota composition and promoting the activity of the LPS/TLR4/NF-κB inflammatory pathway in HFD-induced obese individuals.
Collapse
Affiliation(s)
- Yin-Yi Ding
- Food Safety Key Laboratory of Zhejiang Province, Food Nutrition Science Centre, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, China
- Department of Clinical Laboratory, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, China
| | - Jinchi Lan
- Food Safety Key Laboratory of Zhejiang Province, Food Nutrition Science Centre, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, China
| | - Yumeng Fang
- Food Safety Key Laboratory of Zhejiang Province, Food Nutrition Science Centre, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, China
| | - Yuxiang Pan
- Food Safety Key Laboratory of Zhejiang Province, Food Nutrition Science Centre, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, China
| | - Zhenyu Gu
- Food Safety Key Laboratory of Zhejiang Province, Food Nutrition Science Centre, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, China
| | - Jing Xue
- Food Safety Key Laboratory of Zhejiang Province, Food Nutrition Science Centre, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, China
- Collaborative Innovation Center of Seafood Deep Processing, Zhejiang Province Joint Key Laboratory of Aquatic Products Processing, Institute of Seafood, Zhejiang Gongshang University, Hangzhou, 310018, China
| | - Ying Yang
- Institute of Food Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China
| | - Mengqi Jiang
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Yujun Ge
- Central blood station of Jiaxing, Jiaxing, 314000, China
| | - Qing Shen
- Department of Clinical Laboratory, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, China
- Collaborative Innovation Center of Seafood Deep Processing, Zhejiang Province Joint Key Laboratory of Aquatic Products Processing, Institute of Seafood, Zhejiang Gongshang University, Hangzhou, 310018, China
| |
Collapse
|
34
|
Samy JVRA, Kumar N, Singaravel S, Krishnamoorthy R, Alshuniaber MA, Gatasheh MK, Venkatesan A, Natesan V, Kim SJ. Effect of Prunetin on Streptozotocin-Induced Diabetic Nephropathy in Rats - a Biochemical and Molecular Approach. Biomol Ther (Seoul) 2023; 31:619-628. [PMID: 37818618 PMCID: PMC10616515 DOI: 10.4062/biomolther.2023.068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 07/25/2023] [Accepted: 07/30/2023] [Indexed: 10/12/2023] Open
Abstract
In the modern era, chronic kidney failure due to diabetes has spread across the globe. Prunetin (PRU), a component of herbal medicines, has a broad variety of pharmacological activities; these may help to slow the onset of diabetic kidney disease. The anti-nephropathic effects of PRU have not yet been reported. The present study explored the potential nephroprotective actions of PRU in diabetic rats. For 28 days, nephropathic rats were given oral doses of PRU (20, 40, and 80 mg/kg). Body weight, blood urea, creatinine, total protein, lipid profile, liver marker enzymes, carbohydrate metabolic enzymes, C-reactive protein, antioxidants, lipid peroxidative indicators, and the expression of insulin receptor substrate 1 (IRS-1) and glucose transporter 2 (GLUT-2) mRNA genes were all examined. Histological examinations of the kidneys, liver, and pancreas were also performed. The oral treatment of PRU drastically lowered the blood glucose, HbA1c, blood urea, creatinine, serum glutamic-oxaloacetic transaminase, serum glutamic pyruvic transaminase, alkaline phosphatase, lipid profile, and hexokinase. Meanwhile, the levels of fructose 1,6-bisphosphatase, glucose-6-phosphatase, and phosphoenol pyruvate carboxykinase were all elevated, but glucose-6-phosphate dehydrogenase dropped significantly. Inflammatory marker antioxidants and lipid peroxidative markers were also less persistent due to this administration. PRU upregulated the IRS-1 and GLUT-2 gene expression in the nephropathic group. The possible renoprotective properties of PRU were validated by histopathology of the liver, kidney, and pancreatic tissues. It is therefore proposed that PRU (80 mg/kg) has considerable renoprotective benefits in diabetic nephropathy in rats.
Collapse
Affiliation(s)
- Jose Vinoth Raja Antony Samy
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Annamalainagar 608002, Tamil Nadu, India
| | - Nirubama Kumar
- Department of Biochemistry, Kongunadu Arts and Science College, Coimbatore 641029, Tamil Nadu, India
| | | | - Rajapandiyan Krishnamoorthy
- Department of Food Science and Nutrition, College of Food and Agriculture Sciences, King Saud University, Riyadh 11451, Kingdom of Saudi Arabia
| | - Mohammad A Alshuniaber
- Department of Food Science and Nutrition, College of Food and Agriculture Sciences, King Saud University, Riyadh 11451, Kingdom of Saudi Arabia
| | - Mansour K. Gatasheh
- Department of Biochemistry, College of Science, King Saud University, Riyadh 11451, Kingdom of Saudi Arabia
| | - Amalan Venkatesan
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Annamalainagar 608002, Tamil Nadu, India
| | - Vijayakumar Natesan
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Annamalainagar 608002, Tamil Nadu, India
| | - Sung-Jin Kim
- Department of Pharmacology and Toxicology, Metabolic Diseases Research Laboratory, School of Dentistry, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
35
|
Konadu B, Cox CK, Garrett MR, Gibert Y. Excess glucose or fat differentially affects metabolism and appetite-related gene expression during zebrafish embryogenesis. iScience 2023; 26:107063. [PMID: 37534154 PMCID: PMC10391732 DOI: 10.1016/j.isci.2023.107063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 03/28/2023] [Accepted: 06/02/2023] [Indexed: 08/04/2023] Open
Abstract
Zebrafish embryos use their yolk sac reserve as the sole nutrient source during embryogenesis. The two main forms of energy fuel can be found in the form of glucose or fat. Zebrafish embryos were exposed to glucose or injected with free fatty acid/Triacylglycerol (FFA/TAG) into the yolk sac at 24 hpf. At 72 hpf, glucose exposed or FFA/TAG injected had differential effects on gene expression in embryos, with fat activating lipolysis and β-oxidation and glucose activating the insulin pathway. Bulk RNA-seq revealed that more gene expression was affected by glucose exposure compared to FFA/TAGs injection. Appetite-controlling genes were also differently affected by glucose exposure or FFA/TAG injections. Because the embryo did not yet feed itself at the time of our analysis, gene expression changes occurred in absence of actual hunger and revealed how the embryo manages its nutrient intake before active feeding.
Collapse
Affiliation(s)
- Bridget Konadu
- Department of Cell and Molecular Biology, Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Carol K. Cox
- Department of Cell and Molecular Biology, Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Michael R. Garrett
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Yann Gibert
- Department of Cell and Molecular Biology, Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, MS 39216, USA
| |
Collapse
|
36
|
Baraskar K, Thakur P, Shrivastava R, Shrivastava VK. Ameliorative effects of gallic acid on GLUT-4 expression and insulin resistance in high fat diet-induced obesity animal model mice, Mus musculus. J Diabetes Metab Disord 2023; 22:721-733. [PMID: 37255787 PMCID: PMC10225423 DOI: 10.1007/s40200-023-01194-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 02/01/2023] [Indexed: 06/01/2023]
Abstract
Reduced activity of glucose transporter type 4 isoform (GLUT-4), an insulin-sensitive glucose transporter distributed on the adipocytes, is associated with impaired insulin signaling. Insulin resistance resulting from alteration in glucose transport is responsible for exacerbating the emergence of metabolic abnormalities. The present study aimed to investigate the effects of the antidote gallic acid (GA) on expression-related changes in GLUT-4 and insulin receptor substrate-1 (IRS-1) in the visceral adipose tissue and on the subsequent development of insulin resistance in a high-fat diet (HFD)-induced obesity animal model. Methods: Twenty-four female Swiss albino mice were used and separated into the following four groups (six animals in each group): control group (standard pellet diet), HFD group, (60% HFD), HFD + GA group (60% HFD and GA 50 mg/kg body weight for 60 days), and GA group (GA 50 mg/kg body weight for 60 days). The effect of HFD on serum glucose, total cholesterol, triglycerides, high-density lipoprotein cholesterol (HDL), low-density lipoprotein (LDL) cholesterol, and insulin was evaluated. Additionally, homeostasis model assessment for insulin resistance (HOMA-IR) and glucose tolerance test (GTT) was performed. The serum antioxidative profile, which comprises oxidative parameters (superoxide dismutase [SOD], catalase [CAT], and glutathione peroxidase [GPx]) was measured. The effectiveness of GA against HFD-induced alteration in GLUT-4 and IRS-1 expression was also evaluated. Results: The experimental group that fed on GA + HFD had improved levels of serum triglycerides (p˂0.001), cholesterol (p˂0.05), and LDL cholesterol. GA administration also significantly improved hyperinsulinemia and HOMA-IR index (p˂0.001) in HFD mice. GA improved GTT results (p˂0.05); activity of SOD, CAT, and GPx (p˂0.05); and upregulated mRNA expression of GLUT-4 and IRS-1(p˂0.05) in the visceral adipose tissue in the HFD + GA experimental group. Conclusion: A link exists between insulin resistance, GLUT-4, and IRS-1 expression in the adipose tissue, and the initiation of metabolic syndrome, a condition characterized by obesity. GA may promote insulin signaling, glucose uptake, and lipid metabolism in the adipose tissues by mitigating oxidative stress. GA can also be used to manage obesity-related comorbidities including type 2 diabetes and dyslipidemia. Supplementary Information The online version contains supplementary material available at 10.1007/s40200-023-01194-5.
Collapse
Affiliation(s)
- Kirti Baraskar
- Endocrinology Unit, Biosciences Department, Barkatullah University, 462026 Bhopal, Madhya Pradesh India
| | - Pratibha Thakur
- Department of Medicine, Indira Gandhi Medical College, 171001 Shimla, Himachal Pradesh India
| | - Renu Shrivastava
- Zoology Department, Sri Sathya Sai, College for Women, 262024 Bhopal, Madhya Pradesh India
| | - Vinoy Kumar Shrivastava
- Endocrinology Unit, Biosciences Department, Barkatullah University, 462026 Bhopal, Madhya Pradesh India
| |
Collapse
|
37
|
Chen L, Li H, Ru Y, Song Y, Shen Y, Zhao L, Huang G, Chen Y, Qi Z, Li R, Dong C, Fang J, Lam TKY, Yang Z, Cai Z. Xanthine-derived reactive oxygen species exacerbates adipose tissue disorders in male db/db mice induced by real-ambient PM2.5 exposure. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 882:163592. [PMID: 37087002 DOI: 10.1016/j.scitotenv.2023.163592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 04/11/2023] [Accepted: 04/15/2023] [Indexed: 05/03/2023]
Abstract
Epidemiological and experimental data have associated exposure to fine particulate matter (PM2.5) with various metabolic dysfunctions and diseases, including overweight and type 2 diabetes. Adipose tissue is an energy pool for storing lipids, a necessary regulator of glucose homeostasis, and an active endocrine organ, playing an essential role in developing various related diseases such as diabetes and obesity. However, the molecular mechanisms underlying PM2.5-impaired functions in adipose tissue have rarely been explored. In this work, metabolomics based on liquid chromatography-mass spectrometry was performed to study the adverse impacts of PM2.5 exposure on brown adipose tissue (BAT) and white adipose tissue (WAT) in the diabetic mouse model. We found the effects of PM2.5 exposure by comparing the different metabolites in both adipose tissues of male db/db mice using real-ambient PM2.5 exposure. The results showed that PM2.5 exposure changed the purine metabolism in mice, especially the dramatic increase of xanthine content in both WAT and BAT. These changes led to significant oxidative stress. Then the results from real-time quantitative polymerase chain reaction showed that PM2.5 exposure could cause the production of inflammatory factors in both adipose tissues. Moreover, the increased reactive oxygen species (ROS) promoted triglyceride accumulation in WAT and inhibited its decomposition, causing increased WAT content in db/db mice. In addition, PM2.5 exposure significantly suppressed thermogenesis and affected energy metabolism in the BAT of male db/db mice, which may deteriorate insulin sensitivity and blood glucose regulation. This research demonstrated the impact of PM2.5 on the adipose tissue of male db/db mice, which may be necessary for public health.
Collapse
Affiliation(s)
- Leijian Chen
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong
| | - Huankai Li
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong
| | - Yi Ru
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong
| | - Yuanyuan Song
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong
| | - Yuting Shen
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong
| | - Lifang Zhao
- Institute of Environmental Science, Shanxi University, Taiyuan 030006, China
| | - Gefei Huang
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong
| | - Yi Chen
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong
| | - Zenghua Qi
- School of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou 510006, China
| | - Ruijin Li
- Institute of Environmental Science, Shanxi University, Taiyuan 030006, China
| | - Chuan Dong
- Institute of Environmental Science, Shanxi University, Taiyuan 030006, China
| | - Jiacheng Fang
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong
| | - Thomas Ka-Yam Lam
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong
| | - Zhu Yang
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong.
| | - Zongwei Cai
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong
| |
Collapse
|
38
|
Chang YC, Chan MH, Yang YF, Li CH, Hsiao M. Glucose transporter 4: Insulin response mastermind, glycolysis catalyst and treatment direction for cancer progression. Cancer Lett 2023; 563:216179. [PMID: 37061122 DOI: 10.1016/j.canlet.2023.216179] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/27/2023] [Accepted: 04/07/2023] [Indexed: 04/17/2023]
Abstract
The glucose transporter family (GLUT) consists of fourteen members. It is responsible for glucose homeostasis and glucose transport from the extracellular space to the cell cytoplasm to further cascade catalysis. GLUT proteins are encoded by the solute carrier family 2 (SLC2) genes and are members of the major facilitator superfamily of membrane transporters. Moreover, different GLUTs also have their transporter kinetics and distribution, so each GLUT member has its uniqueness and importance to play essential roles in human physiology. Evidence from many studies in the field of diabetes showed that GLUT4 travels between the plasma membrane and intracellular vesicles (GLUT4-storage vesicles, GSVs) and that the PI3K/Akt pathway regulates this activity in an insulin-dependent manner or by the AMPK pathway in response to muscle contraction. Moreover, some published results also pointed out that GLUT4 mediates insulin-dependent glucose uptake. Thus, dysfunction of GLUT4 can induce insulin resistance, metabolic reprogramming in diverse chronic diseases, inflammation, and cancer. In addition to the relationship between GLUT4 and insulin response, recent studies also referred to the potential upstream transcription factors that can bind to the promoter region of GLUT4 to regulating downstream signals. Combined all of the evidence, we conclude that GLUT4 has shown valuable unknown functions and is of clinical significance in cancers, which deserves our in-depth discussion and design compounds by structure basis to achieve therapeutic effects. Thus, we intend to write up a most updated review manuscript to include the most recent and critical research findings elucidating how and why GLUT4 plays an essential role in carcinogenesis, which may have broad interests and impacts on this field.
Collapse
Affiliation(s)
- Yu-Chan Chang
- Department of Biomedical Imaging and Radiological Science, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ming-Hsien Chan
- Department of Biomedical Imaging and Radiological Science, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yi-Fang Yang
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Chien-Hsiu Li
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, Taipei, Taiwan; Department of Biochemistry, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
39
|
De Loof M, Renguet E, Ginion A, Bouzin C, Horman S, Beauloye C, Bertrand L, Bultot L. Enhanced protein acetylation initiates fatty acid-mediated inhibition of cardiac glucose transport. Am J Physiol Heart Circ Physiol 2023; 324:H305-H317. [PMID: 36607800 DOI: 10.1152/ajpheart.00449.2022] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Fatty acids (FAs) rapidly and efficiently reduce cardiac glucose uptake in the Randle cycle or glucose-FA cycle. This fine-tuned physiological regulation is critical to allow optimal substrate allocation during fasted and fed states. However, the mechanisms involved in the direct FA-mediated control of glucose transport have not been totally elucidated yet. We previously reported that leucine and ketone bodies, other cardiac substrates, impair glucose uptake by increasing global protein acetylation from acetyl-CoA. As FAs generate acetyl-CoA as well, we postulated that protein acetylation is enhanced by FAs and participates in their inhibitory action on cardiac glucose uptake. Here, we demonstrated that both palmitate and oleate promoted a rapid increase in protein acetylation in primary cultured adult rat cardiomyocytes, which correlated with an inhibition of insulin-stimulated glucose uptake. This glucose absorption deficit was caused by an impairment in the translocation of vesicles containing the glucose transporter GLUT4 to the plasma membrane, although insulin signaling remained unaffected. Interestingly, pharmacological inhibition of lysine acetyltransferases (KATs) prevented this increase in protein acetylation and glucose uptake inhibition induced by FAs. Similarly, FA-mediated inhibition of insulin-stimulated glucose uptake could be prevented by KAT inhibitors in perfused hearts. To summarize, enhanced protein acetylation can be considered as an early event in the FA-induced inhibition of glucose transport in the heart, explaining part of the Randle cycle.NEW & NOTEWORTHY Our results show that cardiac metabolic overload by oleate or palmitate leads to increased protein acetylation inhibiting GLUT4 translocation to the plasma membrane and glucose uptake. This observation suggests an additional regulation mechanism in the physiological glucose-FA cycle originally discovered by Randle.
Collapse
Affiliation(s)
- Marine De Loof
- Pole of Cardiovascular Research, Institute for Experimental and Clinical Research, UCLouvain, Brussels, Belgium
| | - Edith Renguet
- Pole of Cardiovascular Research, Institute for Experimental and Clinical Research, UCLouvain, Brussels, Belgium
| | - Audrey Ginion
- Pole of Cardiovascular Research, Institute for Experimental and Clinical Research, UCLouvain, Brussels, Belgium
| | - Caroline Bouzin
- Institute for Experimental and Clinical Research, Imaging platform (2IP), UCLouvain, Brussels, Belgium
| | - Sandrine Horman
- Pole of Cardiovascular Research, Institute for Experimental and Clinical Research, UCLouvain, Brussels, Belgium
| | - Christophe Beauloye
- Pole of Cardiovascular Research, Institute for Experimental and Clinical Research, UCLouvain, Brussels, Belgium.,Division of Cardiology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Luc Bertrand
- Pole of Cardiovascular Research, Institute for Experimental and Clinical Research, UCLouvain, Brussels, Belgium.,WELBIO Department, WEL Research Institute, Wavre, Belgium
| | - Laurent Bultot
- Pole of Cardiovascular Research, Institute for Experimental and Clinical Research, UCLouvain, Brussels, Belgium
| |
Collapse
|
40
|
Ding YY, Fang Y, Pan Y, Lan J, Xu T, Zhang W, Mao H, Gu Z, Chen X, Shen Q. Orally administered octacosanol improves liver insulin resistance in high-fat diet-fed mice through the reconstruction of the gut microbiota structure and inhibition of the TLR4/NF-κB inflammatory pathway. Food Funct 2023; 14:769-786. [PMID: 36594412 DOI: 10.1039/d2fo02463b] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
1-Octacosanol (Octa) is reported to possess many physiological properties. However, its relative mechanism has not been illustrated yet. Herein, we aimed to investigate the effect of Octa on insulin resistance in mice fed with a high fat diet (HFD) and used an in vitro simulated gastrointestinal tract to analyze its digestive behavior. The effects of Octa on the gut microbiota were verified by in vitro fermentation using the mouse fecal microbiota. As a result, the Octa monomer was digested into shortened saturated and unsaturated fatty acids (C10-C24) in the simulated gastrointestinal tract. Octa improved the fasting blood glucose (FBG), insulin resistance (IR), plasma lipids, and inflammatory response in HFD-fed mice in a dose-dependent manner. This study also suggested that a high-dose of Octa effectively decreased the levels of toll-like receptor 4 (TLR4), nuclear factor kappa-B (NF-κB), tumor necrosis factor-α (TNF-α), and interleukin-6 (IL-6) in the plasma of HFD-fed mice. Octa improved the oxidative stress induced by a HFD and increased the expression of the Nrf2/ARE signaling pathway. Importantly, Octa reshaped gut microbiota through decreasing Firmicutes content and increasing Bacteroidota and Verrucomicrobiota contents at the phylum level, and the changes of intestinal flora structure caused by Octa were significantly correlated with the changes of inflammatory biomarkers. In conclusion, the effects of Octa on insulin resistance might be attributed to the reconstruction of the gut microbiota structure and inhibition of the TLR4/NF-κB inflammatory pathway in HFD-induced obese individuals.
Collapse
Affiliation(s)
- Yin-Yi Ding
- Food Nutrition Science Center, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, China. .,Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, China
| | - Yumeng Fang
- Food Nutrition Science Center, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, China.
| | - Yuxiang Pan
- Food Nutrition Science Center, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, China.
| | - Jinchi Lan
- Food Nutrition Science Center, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, China.
| | - Tao Xu
- Huzhou Shengtao Biotechnology LLC, Huzhou, 313000, China
| | - Wanyue Zhang
- Food Nutrition Science Center, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, China.
| | - Huijuan Mao
- Hangzhou Linping Hospital of Traditional Chinese Medicine, Linping, Zhejiang, 311106, China.
| | - Zhenyu Gu
- Food Nutrition Science Center, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, China.
| | - Xi Chen
- Center for General Practice Medicine, Department of General Practice Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, 310014, China
| | - Qing Shen
- Food Nutrition Science Center, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, China. .,Collaborative Innovation Center of Seafood Deep Processing, Zhejiang Province Joint Key Laboratory of Aquatic Products Processing, Institute of Seafood, Zhejiang Gongshang University, Hangzhou, China, 310018
| |
Collapse
|
41
|
Sheinboim D, Parikh S, Manich P, Markus I, Dahan S, Parikh R, Stubbs E, Cohen G, Zemser-Werner V, Bell RE, Ruiz SA, Percik R, Brenner R, Leibou S, Vaknine H, Arad G, Gerber Y, Keinan-Boker L, Shimony T, Bikovski L, Goldstein N, Constantini K, Labes S, Mordechai S, Doron H, Lonescu A, Ziv T, Nizri E, Choshen G, Eldar-Finkelman H, Tabach Y, Helman A, Ben-Eliyahu S, Erez N, Perlson E, Geiger T, Ben-Zvi D, Khaled M, Gepner Y, Levy C. An Exercise-Induced Metabolic Shield in Distant Organs Blocks Cancer Progression and Metastatic Dissemination. Cancer Res 2022; 82:4164-4178. [PMID: 36084256 PMCID: PMC9762351 DOI: 10.1158/0008-5472.can-22-0237] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 06/16/2022] [Accepted: 08/31/2022] [Indexed: 01/07/2023]
Abstract
Exercise prevents cancer incidence and recurrence, yet the underlying mechanism behind this relationship remains mostly unknown. Here we report that exercise induces the metabolic reprogramming of internal organs that increases nutrient demand and protects against metastatic colonization by limiting nutrient availability to the tumor, generating an exercise-induced metabolic shield. Proteomic and ex vivo metabolic capacity analyses of murine internal organs revealed that exercise induces catabolic processes, glucose uptake, mitochondrial activity, and GLUT expression. Proteomic analysis of routinely active human subject plasma demonstrated increased carbohydrate utilization following exercise. Epidemiologic data from a 20-year prospective study of a large human cohort of initially cancer-free participants revealed that exercise prior to cancer initiation had a modest impact on cancer incidence in low metastatic stages but significantly reduced the likelihood of highly metastatic cancer. In three models of melanoma in mice, exercise prior to cancer injection significantly protected against metastases in distant organs. The protective effects of exercise were dependent on mTOR activity, and inhibition of the mTOR pathway with rapamycin treatment ex vivo reversed the exercise-induced metabolic shield. Under limited glucose conditions, active stroma consumed significantly more glucose at the expense of the tumor. Collectively, these data suggest a clash between the metabolic plasticity of cancer and exercise-induced metabolic reprogramming of the stroma, raising an opportunity to block metastasis by challenging the metabolic needs of the tumor. SIGNIFICANCE Exercise protects against cancer progression and metastasis by inducing a high nutrient demand in internal organs, indicating that reducing nutrient availability to tumor cells represents a potential strategy to prevent metastasis. See related commentary by Zerhouni and Piskounova, p. 4124.
Collapse
Affiliation(s)
- Danna Sheinboim
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shivang Parikh
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Paulee Manich
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Irit Markus
- Department of Epidemiology and Preventive Medicine, School of Public Health, Sackler Faculty of Medicine, and Sylvan Adams Sports Institute, Tel Aviv University, Tel Aviv, Israel
| | - Sapir Dahan
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Roma Parikh
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Elisa Stubbs
- Department of Epidemiology and Preventive Medicine, School of Public Health, Sackler Faculty of Medicine, and Sylvan Adams Sports Institute, Tel Aviv University, Tel Aviv, Israel
| | - Gali Cohen
- Department of Epidemiology and Preventive Medicine, School of Public Health, Sackler Faculty of Medicine, and Sylvan Adams Sports Institute, Tel Aviv University, Tel Aviv, Israel.,Stanley Steyer Institute for Cancer Epidemiology and Research, Tel Aviv University, Tel Aviv, Israel
| | | | - Rachel E. Bell
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sara Arciniegas Ruiz
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ruth Percik
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.,Institute of Endocrinology, Chaim Sheba Medical Center, Tel Hashomer, Israel
| | - Ronen Brenner
- Institute of Oncology, E. Wolfson Medical Center, Holon, Israel
| | - Stav Leibou
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Hananya Vaknine
- Institute of Pathology, E. Wolfson Medical Center, Holon, Israel
| | - Gali Arad
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yariv Gerber
- Department of Epidemiology and Preventive Medicine, School of Public Health, Sackler Faculty of Medicine, and Sylvan Adams Sports Institute, Tel Aviv University, Tel Aviv, Israel.,Stanley Steyer Institute for Cancer Epidemiology and Research, Tel Aviv University, Tel Aviv, Israel
| | - Lital Keinan-Boker
- School of Public Health, Faculty of Social Welfare and Health Sciences, University of Haifa, Haifa, Israel.,Israel Center for Disease Control, Israel Ministry of Health, Ramat Gan, Israel
| | - Tal Shimony
- Israel Center for Disease Control, Israel Ministry of Health, Ramat Gan, Israel
| | - Lior Bikovski
- The Myers Neuro-Behavioral Core Facility, Tel Aviv University, Tel Aviv, Israel.,School of Behavioral Sciences, Netanya Academic College, Netanya, Israel
| | - Nir Goldstein
- Department of Epidemiology and Preventive Medicine, School of Public Health, Sackler Faculty of Medicine, and Sylvan Adams Sports Institute, Tel Aviv University, Tel Aviv, Israel
| | - Keren Constantini
- Department of Epidemiology and Preventive Medicine, School of Public Health, Sackler Faculty of Medicine, and Sylvan Adams Sports Institute, Tel Aviv University, Tel Aviv, Israel
| | - Sapir Labes
- Department of Developmental Biology and Cancer Research, Institute of Medical Research-Israel-Canada, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Shimonov Mordechai
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Surgery, E. Wolfson Medical Center, Holon, Israel
| | - Hila Doron
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ariel Lonescu
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Tamar Ziv
- The Smoler Proteomics Center, Technion, Haifa, Israel
| | - Eran Nizri
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Dermatology, Tel Aviv Sourasky (Ichilov) Medical Center, Tel Aviv, Israel
| | - Guy Choshen
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Internal Medicine, Tel Aviv Sourasky (Ichilov) Medical Center, Tel Aviv, Israel
| | - Hagit Eldar-Finkelman
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yuval Tabach
- Department of Developmental Biology and Cancer Research, Institute of Medical Research-Israel-Canada, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Aharon Helman
- Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University, Rehovot, Israel
| | - Shamgar Ben-Eliyahu
- School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel.,Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Neta Erez
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Eran Perlson
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Tamar Geiger
- The Weizmann Institute of Science, Rehovot, Israel
| | - Danny Ben-Zvi
- Department of Developmental Biology and Cancer Research, Institute of Medical Research Israel–Canada, The Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Mehdi Khaled
- INSERM 1186, Gustave Roussy, Université Paris-Saclay, Villejuif, France.,Corresponding Authors: Carmit Levy, Human Molecular Genetics and Biochemistry, Tel Aviv University, Tel Aviv, 69978, Israel. E-mail: ; Yftach Gepner, E-mail: ; and Mehdi Khaled, E-mail:
| | - Yftach Gepner
- Department of Epidemiology and Preventive Medicine, School of Public Health, Sackler Faculty of Medicine, and Sylvan Adams Sports Institute, Tel Aviv University, Tel Aviv, Israel.,Corresponding Authors: Carmit Levy, Human Molecular Genetics and Biochemistry, Tel Aviv University, Tel Aviv, 69978, Israel. E-mail: ; Yftach Gepner, E-mail: ; and Mehdi Khaled, E-mail:
| | - Carmit Levy
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Corresponding Authors: Carmit Levy, Human Molecular Genetics and Biochemistry, Tel Aviv University, Tel Aviv, 69978, Israel. E-mail: ; Yftach Gepner, E-mail: ; and Mehdi Khaled, E-mail:
| |
Collapse
|
42
|
Afolabi OA, Alabi BA, Oluranti O. Diet-induced insulin resistance altered cardiac GLUT4 and FATP/CD36 expression in rats. BENI-SUEF UNIVERSITY JOURNAL OF BASIC AND APPLIED SCIENCES 2022. [DOI: 10.1186/s43088-022-00312-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Altered substrate transport protein expression is central to the effect of insulin resistance on cardiac metabolism. The present study was thus designed to investigate the comparative effects of high fat, high sucrose and salt-induced IR on cardiac expression of fatty acid transporter (FATP) and glucose transporter (GLUT4) in rats.
Results
Rats fed with high fat, high sucrose and salt diets developed impaired glucose tolerance (p > 0.05) and hyperinsulinemia (p < 0.05) compared with control group. Myocardial glucose transporter expression was significantly increased (p < 0.001 for salt-induced IR; p < 0.01 for sucrose-induced IR; p < 0.01 for fat-induced IR) across all IR groups compared with control. Fatty acid transporter expression was also increased (p < 0.001) in high salt diet-induced IR rats, and high fat diet-induced IR rats (p < 0.05).
Conclusions
Our results demonstrate that salt and not caloric excess has a potential role in IR alteration of myocardial substrate transport protein expression in the rat.
Collapse
|
43
|
Tan D, Tseng HHL, Zhong Z, Wang S, Vong CT, Wang Y. Glycyrrhizic Acid and Its Derivatives: Promising Candidates for the Management of Type 2 Diabetes Mellitus and Its Complications. Int J Mol Sci 2022; 23:10988. [PMID: 36232291 PMCID: PMC9569462 DOI: 10.3390/ijms231910988] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/13/2022] [Accepted: 09/15/2022] [Indexed: 11/16/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a chronic metabolic disease, which is characterized by hyperglycemia, chronic insulin resistance, progressive decline in β-cell function, and defect in insulin secretion. It has become one of the leading causes of death worldwide. At present, there is no cure for T2DM, but it can be treated, and blood glucose levels can be controlled. It has been reported that diabetic patients may suffer from the adverse effects of conventional medicine. Therefore, alternative therapy, such as traditional Chinese medicine (TCM), can be used to manage and treat diabetes. In this review, glycyrrhizic acid (GL) and its derivatives are suggested to be promising candidates for the treatment of T2DM and its complications. It is the principal bioactive constituent in licorice, one type of TCM. This review comprehensively summarized the therapeutic effects and related mechanisms of GL and its derivatives in managing blood glucose levels and treating T2DM and its complications. In addition, it also discusses existing clinical trials and highlights the research gap in clinical research. In summary, this review can provide a further understanding of GL and its derivatives in T2DM as well as its complications and recent progress in the development of potential drugs targeting T2DM.
Collapse
Affiliation(s)
| | | | | | | | - Chi Teng Vong
- Macau Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
| | - Yitao Wang
- Macau Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
| |
Collapse
|
44
|
Mengstie MA, Abebe EC, Teklemariam AB, Mulu AT, Teshome AA, Zewde EA, Muche ZT, Azezew MT. Molecular and cellular mechanisms in diabetic heart failure: Potential therapeutic targets. Front Endocrinol (Lausanne) 2022; 13:947294. [PMID: 36120460 PMCID: PMC9478122 DOI: 10.3389/fendo.2022.947294] [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: 05/18/2022] [Accepted: 08/12/2022] [Indexed: 12/15/2022] Open
Abstract
Diabetes Mellitus (DM) is a worldwide health issue that can lead to a variety of complications. DM is a serious metabolic disorder that causes long-term microvascular and macro-vascular complications, as well as the failure of various organ systems. Diabetes-related cardiovascular diseases (CVD) including heart failure cause significant morbidity and mortality worldwide. Concurrent hypertensive heart disease and/or coronary artery disease have been thought to be the causes of diabetic heart failure in DM patients. However, heart failure is extremely common in DM patients even in the absence of other risk factors such as coronary artery disease and hypertension. The occurrence of diabetes-induced heart failure has recently received a lot of attention. Understanding how diabetes increases the risk of heart failure and how it mediates major cellular and molecular alteration will aid in the development of therapeutics to prevent these changes. Hence, this review aimed to summarize the current knowledge and most recent findings in cellular and molecular mechanisms of diabetes-induced heart failure.
Collapse
Affiliation(s)
- Misganaw Asmamaw Mengstie
- Department of Biochemistry, College of Medicine and Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Endeshaw Chekol Abebe
- Department of Biochemistry, College of Medicine and Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Awgichew Behaile Teklemariam
- Department of Biochemistry, College of Medicine and Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Anemut Tilahun Mulu
- Department of Biochemistry, College of Medicine and Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Assefa Agegnehu Teshome
- Department of Anatomy, College of Medicine and Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Edgeit Abebe Zewde
- Department of Physiology, College of Medicine and Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Zelalem Tilahun Muche
- Department of Physiology, College of Medicine and Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Muluken Teshome Azezew
- Department of Physiology, College of Medicine and Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| |
Collapse
|
45
|
Østergård Jensen S, Christen M, Rondahl V, Holland CT, Jagannathan V, Leeb T, Giger U. EHBP1L1 Frameshift Deletion in English Springer Spaniel Dogs with Dyserythropoietic Anemia and Myopathy Syndrome (DAMS) or Neonatal Losses. Genes (Basel) 2022; 13:genes13091533. [PMID: 36140701 PMCID: PMC9498568 DOI: 10.3390/genes13091533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 08/20/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022] Open
Abstract
Hereditary myopathies are well documented in dogs, whereas hereditary dyserythropoietic anemias are rarely seen. The aim of this study was to further characterize the clinical and clinicopathological features of and to identify the causative genetic variant for a dyserythropoietic anemia and myopathy syndrome (DAMS) in English springer spaniel dogs (ESSPs). Twenty-six ESSPs, including five dogs with DAMS and two puppies that died perinatally, were studied. Progressive weakness, muscle atrophy—particularly of the temporal and pelvic muscles—trismus, dysphagia, and regurgitation due to megaesophagus were observed at all ages. Affected dogs had a non-regenerative, microcytic hypochromic anemia with metarubricytosis, target cells, and acanthocytes. Marked erythroid hyperplasia and dyserythropoiesis with non-orderly maturation of erythrocytes and inappropriate microcytic metarubricytosis were present. Muscle biopsies showed centralized nuclei, central pallor, lipocyte infiltrates, and fibrosis, which was consistent with centronuclear myopathy. The genome sequencing of two affected dogs was compared to 782 genomes of different canine breeds. A homozygous frameshift single-base deletion in EHBP1L1 was identified; this gene was not previously associated with DAMS. Pedigree analysis confirmed that the affected ESSPs were related. Variant genotyping showed appropriate complete segregation in the family, which was consistent with an autosomal recessive mode of inheritance. This study expands the known genotype–phenotype correlation of EHBP1L1 and the list of potential causative genes in dyserythropoietic anemias and myopathies in humans. EHBP1L1 deficiency was previously reported as perinatally lethal in humans and knockout mice. Our findings enable the genetic testing of ESSP dogs for early diagnosis and disease prevention through targeted breeding strategies.
Collapse
Affiliation(s)
- Sarah Østergård Jensen
- Clinical Pathology Laboratory, The Swedish University of Agricultural Sciences, 750 07 Uppsala, Sweden
- AniCura Small Animal Referral Hospital Bagarmossen, Ljusnevägen 17, Bagarmossen, 128 48 Stockholm, Sweden
| | - Matthias Christen
- Institute of Genetics, Vetsuisse Faculty, University of Bern, Bremgartenstrasse 109a, 3001 Bern, Switzerland
| | | | - Christopher T. Holland
- Merewether Veterinary Hospital, Suite 2, 25 Llewellyn St, Merewether, NSW 2291, Australia
| | - Vidhya Jagannathan
- Institute of Genetics, Vetsuisse Faculty, University of Bern, Bremgartenstrasse 109a, 3001 Bern, Switzerland
| | - Tosso Leeb
- Institute of Genetics, Vetsuisse Faculty, University of Bern, Bremgartenstrasse 109a, 3001 Bern, Switzerland
| | - Urs Giger
- Vetsuisse Faculty, University of Zürich, Winterthurerstrasse 260, 8057 Zürich, Switzerland
- Correspondence: ; Tel.: +1-610-565-1427
| |
Collapse
|
46
|
Li Y, Wang S, Wang S, Wang S, Tang B, Liu F. Involvement of glucose transporter 4 in ovarian development and reproductive maturation of Harmonia axyridis (Coleoptera: Coccinellidae). INSECT SCIENCE 2022; 29:691-703. [PMID: 34516727 PMCID: PMC9298200 DOI: 10.1111/1744-7917.12972] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/21/2021] [Accepted: 08/30/2021] [Indexed: 06/13/2023]
Abstract
Glucose is vital to embryogenesis, as are glucose transporters. Glucose transporter 4 (Glut4) is one of the glucose transporters, which is involved in rapid uptake of glucose by various cells and promotes glucose homeostasis. Although energy metabolism in insect reproduction is well known, the molecular mechanism of Glut4 in insect reproduction is poorly understood. We suspect that Glut4 is involved in maintaining glucose concentrations in the ovaries and affecting vitellogenesis, which is critical for subsequent oocyte maturation and insect fertility. Harmonia axyridis (Pallas) is a model organism for genetic research and a natural enemy of insect pests. We studied the influence of the Glut4 gene on the reproduction and development of H. axyridis using RNA interference technology. Reverse transcription quantitative polymerase chain reaction analysis revealed that HaGlut4 was most highly expressed in adults. Knockdown of the HaGlut4 gene reduced the transcript levels of HaGlut4, and the weight and number of eggs produced significantly decreased. In addition, the transcript levels of vitellogenin receptor and vitellogenin in the fat bodies and the ovaries of H. axyridis decreased after the interference of Glut4, and decreased the triglyceride, fatty acid, total amino acid and adenosine triphosphate content of H. axyridis. This resulted in severe blockage of ovary development and reduction of yolk formation; there was no development of ovarioles in the developing oocytes. These changes indicate that a lack of HaGlut4 can impair ovarian development and oocyte maturation and result in decreased fecundity.
Collapse
Affiliation(s)
- Yan Li
- College of Horticulture and Plant ProtectionYangzhou UniversityYangzhouJiangsu225009China
- College of Life and Environmental SciencesHangzhou Normal UniversityHangzhouZhejiang310036China
| | - Sha‐Sha Wang
- College of Life and Environmental SciencesHangzhou Normal UniversityHangzhouZhejiang310036China
| | - Su Wang
- Institute of Plant and Environment ProtectionBeijing Academy of Agricultural and Forestry SciencesBeijing100097China
| | - Shi‐Gui Wang
- College of Life and Environmental SciencesHangzhou Normal UniversityHangzhouZhejiang310036China
| | - Bin Tang
- College of Life and Environmental SciencesHangzhou Normal UniversityHangzhouZhejiang310036China
| | - Fang Liu
- College of Horticulture and Plant ProtectionYangzhou UniversityYangzhouJiangsu225009China
| |
Collapse
|
47
|
Renguet E, De Loof M, Fourny N, Ginion A, Bouzin C, Poüs C, Horman S, Beauloye C, Bultot L, Bertrand L. α-Tubulin acetylation on Lysine 40 controls cardiac glucose uptake. Am J Physiol Heart Circ Physiol 2022; 322:H1032-H1043. [PMID: 35486479 DOI: 10.1152/ajpheart.00664.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Our group previously demonstrated that an excess of nutrients, as observed in diabetes, provokes an increase in cardiac protein acetylation responsible for a reduced insulin-stimulated translocation of the glucose transporter GLUT4 to the plasma membrane. The acetylated proteins involved in this event have yet not been identified. α-Tubulin is a promising candidate as a major cytoskeleton component involved, among other things, in the translocation of GLUT4-containing vesicles from their intracellular pools towards the plasma membrane. Moreover, α-tubulin is known to be acetylated, Lys40 (K40) being its best characterized acetylated residue. The present work sought to evaluate the impact of α-tubulin K40 acetylation on cardiac glucose entry, with a particular interest in GLUT4 translocation. First, we observed that a mouse model of high-fat diet-induced obesity presented an increase in cardiac α-tubulin K40 acetylation level. Next, we showed that treatment of insulin-sensitive primary cultured adult rat cardiomyocytes with tubacin, a specific tubulin acetylation inducer, reduced insulin-stimulated glucose uptake and GLUT4 translocation. Conversely, decreasing α-tubulin K40 acetylation by expressing a non-acetylable dominant form of α-tubulin (mCherry α-tubulin K40A mutant) remarkably intensified insulin-induced glucose transport. Finally, mCherry α-tubulin K40A expression similarly improved glucose transport in insulin-resistant cardiomyocytes or after AMP-activated protein kinase activation. Taken together, our study demonstrates that modulation of α-tubulin K40 acetylation level affects glucose transport in cardiomyocytes, offering new putative therapeutic insights regarding modulation of glucose metabolism in insulin-resistant and diabetic hearts.
Collapse
Affiliation(s)
- Edith Renguet
- Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Brussels, Belgium
| | - Marine De Loof
- Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Brussels, Belgium
| | - Natacha Fourny
- Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Brussels, Belgium
| | - Audrey Ginion
- Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Brussels, Belgium
| | - Caroline Bouzin
- Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique, IREC Imaging Platform (2IP), Brussels, Belgium
| | - Christian Poüs
- Université Paris-Saclay, INSERM UMR-S-1193, Châtenay-Malabry, France; AP-HP, Biochimie-Hormonologie, Hôpital Antoine Béclère, Clamart, France
| | - Sandrine Horman
- Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Brussels, Belgium
| | - Christophe Beauloye
- Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Brussels, Belgium.,Cliniques Universitaires Saint-Luc, Division of Cardiology, Brussels, Belgium
| | - Laurent Bultot
- Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Brussels, Belgium
| | - Luc Bertrand
- Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Brussels, Belgium
| |
Collapse
|
48
|
Sakhrani N, Lee AJ, Murphy LA, Kenawy HM, Visco CJ, Ateshian GA, Shah RP, Hung CT. Toward Development of a Diabetic Synovium Culture Model. Front Bioeng Biotechnol 2022; 10:825046. [PMID: 35265601 PMCID: PMC8899218 DOI: 10.3389/fbioe.2022.825046] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 01/24/2022] [Indexed: 11/13/2022] Open
Abstract
Osteoarthritis (OA) is a degenerative joint disease characterized by articular cartilage degradation and inflammation of synovium, the specialized connective tissue that envelops the diarthrodial joint. Type 2 diabetes mellitus (DM) is often found in OA patients, with nearly double the incidence of arthritis reported in patients with diabetes (52%) than those without it (27%). The correlation between OA and DM has been attributed to similar risk factors, namely increasing age and joint loading due to obesity. However, a potential causative link is not well understood due to comorbidities involved with treating diabetic patients, such as high infection rates and poor healing response caused by hyperglycemia and insulin resistance. The purpose of this study was to investigate the effect of hyperglycemic and insulin culture conditions on synovium properties. It was hypothesized that modeling hyperglycemia-induced insulin resistance in synovium would provide novel insights of OA pathogenesis in DM patients. To simulate DM in the synovial joint, healthy synovium was preconditioned in either euglycemic (EG) or hyperglycemic (HG) glucose concentrations with insulin in order to induce the biological response of the diseased phenotype. Synovium biochemical composition was evaluated to determine ECM remodeling under hyperglycemic culture conditions. Concurrent changes in AKT phosphorylation, a signaling pathway implicated in insulin resistance, were measured along with gene expression data for insulin receptors, glucose transporters, and specific glycolysis markers involved in glucose regulation. Since fluid shear stress arising during joint articulation is a relevant upstream stimulus for fibroblast-like synoviocytes (FLS), the predominant cell type in synovium, FLS mechanotransduction was evaluated via intracellular calcium ([Ca2+]i). Incidence and length of primary cilia, a critical effector of cell mechanosensing, were measured as potential mechanisms to support differences in [Ca2+]i responses. Hyperglycemic culture conditions decreased collagen and GAG content compared to EG groups, while insulin recovered ECM constituents. FLS mechanosensitivity was significantly greater in EG and insulin conditions compared to HG and non-insulin treated groups. Hyperglycemic treatment led to decreased incidence and length of primary cilia and decreased AKT phosphorylation, providing possible links to the mechanosensing response and suggesting a potential correlation between glycemic culture conditions, diabetic insulin resistance, and OA development.
Collapse
Affiliation(s)
- Neeraj Sakhrani
- Department of Biomedical Engineering, Columbia University, New York, NY, United States
| | - Andy J Lee
- Department of Biomedical Engineering, Columbia University, New York, NY, United States
| | - Lance A Murphy
- Department of Biomedical Engineering, Columbia University, New York, NY, United States
| | - Hagar M Kenawy
- Department of Biomedical Engineering, Columbia University, New York, NY, United States
| | - Christopher J Visco
- Department of Rehabilitation and Regenerative Medicine, Columbia University, New York, NY, United States
| | - Gerard A Ateshian
- Department of Biomedical Engineering, Columbia University, New York, NY, United States.,Department of Mechanical Engineering, Columbia University, New York, NY, United States
| | - Roshan P Shah
- Department of Orthopedic Surgery, Columbia University, New York, NY, United States
| | - Clark T Hung
- Department of Biomedical Engineering, Columbia University, New York, NY, United States.,Department of Orthopedic Surgery, Columbia University, New York, NY, United States
| |
Collapse
|
49
|
Wang TN, Hu XG, Chen GX. Uses of knockout, knockdown, and transgenic models in the studies of glucose transporter 4. World J Meta-Anal 2022; 10:1-11. [DOI: 10.13105/wjma.v10.i1.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 12/10/2021] [Accepted: 02/23/2022] [Indexed: 02/06/2023] Open
|
50
|
Carpéné C, Marti L, Morin N. Increased monoamine oxidase activity and imidazoline binding sites in insulin-resistant adipocytes from obese Zucker rats. World J Biol Chem 2022; 13:15-34. [PMID: 35126867 PMCID: PMC8790288 DOI: 10.4331/wjbc.v13.i1.15] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 07/09/2021] [Accepted: 01/14/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Despite overt insulin resistance, adipocytes of genetically obese Zucker rats accumulate the excess of calorie intake in the form of lipids.
AIM To investigate whether factors can replace or reinforce insulin lipogenic action by exploring glucose uptake activation by hydrogen peroxide, since it is produced by monoamine oxidase (MAO) and semicarbazide-sensitive amine oxidase (SSAO) in adipocytes.
METHODS 3H-2-deoxyglucose uptake (2-DG) was determined in adipocytes from obese and lean rats in response to insulin or MAO and SSAO substrates such as tyramine and benzylamine. 14C-tyramine oxidation and binding of imidazolinic radioligands [3H-Idazoxan, 3H-(2-benzofuranyl)-2-imidazoline] were studied in adipocytes, the liver, and muscle. The influence of in vivo administration of tyramine + vanadium on glucose handling was assessed in lean and obese rats.
RESULTS 2-DG uptake and lipogenesis stimulation by insulin were dampened in adipocytes from obese rats, when compared to their lean littermates. Tyramine and benzylamine activation of hexose uptake was vanadate-dependent and was also limited, while MAO was increased and SSAO decreased. These changes were adipocyte-specific and accompanied by a greater number of imidazoline I2 binding sites in the obese rat, when compared to the lean. In vitro, tyramine precluded the binding to I2 sites, while in vivo, its administration together with vanadium lowered fasting plasma levels of glucose and triacylglycerols in obese rats.
CONCLUSION The adipocytes from obese Zucker rats exhibit increased MAO activity and imidazoline binding site number. However, probably as a consequence of SSAO down-regulation, the glucose transport stimulation by tyramine is decreased as much as that of insulin in these insulin-resistant adipocytes. The adipocyte amine oxidases deserve more studies with respect to their putative contribution to the management of glucose and lipid handling.
Collapse
Affiliation(s)
- Christian Carpéné
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM, Toulouse 31342, France
| | - Luc Marti
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM, Toulouse 31342, France
| | - Nathalie Morin
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM, Toulouse 31342, France
- Faculté de Pharmacie de Paris, Paris University, Paris 75270, France
| |
Collapse
|