1
|
Akagunduz B, Ergün Y, Dog An Akagündüz DD, Akbas N, Akagündüz D, Karaog Lu A, Soysal P. Blood-based biomarkers of frailty in older patients with cancer. Curr Opin Support Palliat Care 2025; 19:25-32. [PMID: 39888831 DOI: 10.1097/spc.0000000000000747] [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: 02/02/2025]
Abstract
PURPOSE OF REVIEW This review aims to evaluate the current evidence on blood-based biomarkers for frailty detection in older cancer patients. It explores the potential of various biomarkers, including inflammatory markers and microRNAs (miRNAs), to serve as indicators of frailty and examines the limitations of existing studies. The review also highlights the need for further research to validate these biomarkers and improve their clinical applicability. RECENT FINDINGS Recent studies have examined blood biomarkers associated with frailty in older cancer patients. Findings suggest that elevated granulocyte levels and lower macrophage-derived stem cells and regulatory T cells are linked to frailty. Inflammatory biomarkers such as interleukin-6 and specific miRNAs, as well as higher neutrophil-to-lymphocyte ratios, have also been identified as potential indicators of frailty. While these biomarkers show promise, no single marker has proven sufficient, and combining them may improve frailty detection. Further research is needed to validate their clinical usefulness in this population. SUMMARY Blood-based biomarkers show potential for detecting frailty in older patients with cancer, but further research is needed, particularly beyond an inflammatory focus and with more robust study designs.
Collapse
Affiliation(s)
- Baran Akagunduz
- Department of Medical Oncology, King Hamad University Hospıtal Bahrain Oncology Center, Muharraq, Bahrain
| | - Yakup Ergün
- Department of Medical Oncology, Diyarbakır Bower Hospital, Diyarbakır, Turkey
| | | | - Nergis Akbas
- Department of Biochemistry, Yalova University School of Medicine, Yalova, Turkey
| | - Dilara Akagündüz
- Department of Internal Medicine, Ankara Bilkent City Hospital, Ankara, Turkey
| | - Aziz Karaog Lu
- Department of Medical Oncology, Dokuz Eylül University. School of Medicine, İzmir, Turkey
| | - Pinar Soysal
- Department of Geriatrics, Bezmialem University School of Medicine, İstanbul, Turkey
| |
Collapse
|
2
|
Ceja-Galicia ZA, Cespedes-Acuña CLA, El-Hafidi M. Protection Strategies Against Palmitic Acid-Induced Lipotoxicity in Metabolic Syndrome and Related Diseases. Int J Mol Sci 2025; 26:788. [PMID: 39859502 PMCID: PMC11765695 DOI: 10.3390/ijms26020788] [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: 11/26/2024] [Revised: 12/26/2024] [Accepted: 01/03/2025] [Indexed: 01/27/2025] Open
Abstract
Diets rich in carbohydrate and saturated fat contents, when combined with a sedentary lifestyle, contribute to the development of obesity and metabolic syndrome (MetS), which subsequently increase palmitic acid (PA) levels. At high concentrations, PA induces lipotoxicity through several mechanisms involving endoplasmic reticulum (ER) stress, mitochondrial dysfunction, inflammation and cell death. Nevertheless, there are endogenous strategies to mitigate PA-induced lipotoxicity through its unsaturation and elongation and its channeling and storage in lipid droplets (LDs), which plays a crucial role in sequestering oxidized lipids, thereby reducing oxidative damage to lipid membranes. While extended exposure to PA promotes mitochondrial reactive oxygen species (ROS) generation leading to cell damage, acute exposure of ß-cells to PA increases glucose-stimulated insulin secretion (GSIS), through the activation of free fatty acid receptors (FFARs). Subsequently, the activation of FFARs by exogenous agonists has been suggested as a potential therapeutic strategy to prevent PA-induced lipotoxicity in ß cells. Moreover, some saturated fatty acids, including oleic acid, can counteract the negative impact of PA on cellular health, suggesting a complex interaction between different dietary fats and cellular outcomes. Therefore, the challenge is to prevent the lipid peroxidation of dietary unsaturated fatty acids through the utilization of natural antioxidants. This complexity indicates the necessity for further research into the function of palmitic acid in diverse pathological conditions and to find the main therapeutic target against its lipotoxicity. The aim of this review is, therefore, to examine recent data regarding the mechanism underlying PA-induced lipotoxicity in order to identify strategies that can promote protection mechanisms against lipotoxicity, dysfunction and apoptosis in MetS and obesity.
Collapse
Affiliation(s)
- Zeltzin Alejandra Ceja-Galicia
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico;
| | | | - Mohammed El-Hafidi
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico;
| |
Collapse
|
3
|
Han Z, Zhu Y, Xia Z, Deng Q, He H, Yin Q, Zhang H, Yuan M, Yang C, Tian G, Mi J, Xu F. Genetic analyses identify circulating genes related to brain structures associated with Parkinson's disease. NPJ Parkinsons Dis 2025; 11:17. [PMID: 39809793 PMCID: PMC11733288 DOI: 10.1038/s41531-024-00859-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 12/19/2024] [Indexed: 01/16/2025] Open
Abstract
Magnetic resonance imaging and circulating molecular testing are potential methods for diagnosing and treating Parkinson's disease (PD). However, their relationships remain insufficiently studied. Using genome-wide association summary statistics, we found in the general population a genetic negative correlation between white matter tract mean diffusivity and PD (-0.17 < Rg < -0.11, p < 0.05), and a positive correlation with intracellular volume fraction (0.12 < Rg < 0.2, p < 0.05). Additionally, 1345 circulating genes causally linked with white matter tract diffusivity were enriched for muscle physiological abnormalities (padj < 0.05). Notable genes, including LRRC37A4P (effect size = 15.7, p = 1.23E-55) and KANSL1-AS1 (effect size = -15.3, p = 1.13E-52), were directly associated with PD. Moreover, 23 genes were found linked with genetically correlated PD-IDP pairs (PPH4 > 0.8), including SH2B1 and TRIM10. Our study bridges the gap between molecular genetics, neuroimaging, and PD pathology, and suggests novel targets for diagnosis and treatment.
Collapse
Affiliation(s)
- Zhe Han
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, China
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Yanping Zhu
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, China
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Zhenhong Xia
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, China
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Qing Deng
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, China
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Hongjie He
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, China
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Quanting Yin
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, China
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Hui Zhang
- Baotou Cancer Hospital, Baotou, China
| | | | - Chunhua Yang
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, China
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Geng Tian
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, China
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Jia Mi
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, China.
- School of Pharmacy, Binzhou Medical University, Yantai, China.
| | - Fuyi Xu
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, China.
- School of Pharmacy, Binzhou Medical University, Yantai, China.
| |
Collapse
|
4
|
Pebriana RB, Simsek S, Heijink M, Giera M. Rapid Analysis of Sterols in Blood-Derived Samples. Methods Mol Biol 2025; 2855:291-302. [PMID: 39354314 DOI: 10.1007/978-1-0716-4116-3_17] [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] [Indexed: 10/03/2024]
Abstract
Dysregulations of cholesterol biosynthesis are known to be associated with several pathologies. Due to the rapid growth of clinical investigations in this research area, a specific, fast, and valid method for analyzing cholesterol, its precursors, and metabolites is required. Here, we describe a rapid method for sample preparation, separation, and quantification of sterols in blood-derived samples using polymeric solid phase extraction followed by gas chromatography-mass spectrometry. The validated method demonstrates a reliable quantification of cholesterol, its precursors, and metabolites.
Collapse
Affiliation(s)
- Ratna Budhi Pebriana
- Leiden University Medical Center, Center for Proteomics and Metabolomics, Leiden, The Netherlands
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Selma Simsek
- Leiden University Medical Center, Center for Proteomics and Metabolomics, Leiden, The Netherlands
| | - Marieke Heijink
- Leiden University Medical Center, Center for Proteomics and Metabolomics, Leiden, The Netherlands
| | - Martin Giera
- Leiden University Medical Center, Center for Proteomics and Metabolomics, Leiden, The Netherlands.
| |
Collapse
|
5
|
Nakamura K, Takeda S, Sakurai T, Ukawa S, Okada E, Nakagawa T, Imae A, Hui SP, Chiba H, Tamakoshi A. Relationships of the Surface Charge of Low-Density Lipoprotein (LDL) with the Serum LDL-Cholesterol and Atherosclerosis Levels in a Japanese Population: The DOSANCO Health Study. J Atheroscler Thromb 2025; 32:34-47. [PMID: 38960632 PMCID: PMC11706988 DOI: 10.5551/jat.64961] [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: 03/11/2024] [Accepted: 05/16/2024] [Indexed: 07/05/2024] Open
Abstract
AIM This study investigated the associations of the surface charge of low-density lipoprotein (LDL) with the serum LDL-cholesterol and atherosclerosis levels in a community-based Japanese population. METHODS The study had a cross-sectional design and included 409 community residents aged 35-79 years who did not take medications for dyslipidemia. The potential electric charge of LDL and the zeta potential, which indicate the surface charge of LDL, were measured by laser Doppler microelectrophoresis. The correlations of the zeta potential of LDL (-mV) with the serum LDL-cholesterol levels (mg/dL), cardio-ankle vascular index (CAVI), and serum high-sensitivity C-reactive protein (hsCRP) levels (log-transformed values, mg/L) were examined using Pearson's correlation coefficient (r). Linear regression models were constructed to examine these associations after adjusting for potential confounding factors. RESULTS A total of 201 subjects with correctly stored samples were included in the primary analysis for zeta potential measurement. An inverse correlation was observed between the LDL zeta potential and the serum LDL-cholesterol levels (r=-0.20; p=0.004). This inverse association was observed after adjusting for sex, age, dietary cholesterol intake, smoking status, alcohol intake, body mass index, and the serum levels of the major classes of free fatty acids (standardized β=-6.94; p=0.005). However, the zeta potential of LDL showed almost no association with CAVI or the serum hsCRP levels. Similar patterns were observed in the 208 subjects with compromised samples as well as all the original 409 subjects. CONCLUSION A higher electronegative surface charge of LDL was associated with lower serum LDL-cholesterol levels in the general Japanese population.
Collapse
Affiliation(s)
- Koshi Nakamura
- Department of Public Health and Epidemiology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
- Department of Public Health, Hokkaido University Faculty of Medicine, Sapporo, Japan
| | - Seiji Takeda
- Faculty of Pharmaceutical Sciences, Hokkaido University of Science, Sapporo, Japan
- Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | | | - Shigekazu Ukawa
- Department of Public Health, Hokkaido University Faculty of Medicine, Sapporo, Japan
- Research Unit of Advanced Interdisciplinary Care Science, Osaka Metropolitan University Graduate School of Human Life Science, Osaka, Japan
| | - Emiko Okada
- Department of Public Health, Hokkaido University Faculty of Medicine, Sapporo, Japan
- Public Interest Incorporated Foundation, the Health Care Science Institute, Tokyo, Japan
| | | | | | - Shu-Ping Hui
- Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Hitoshi Chiba
- Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
- Department of Nutrition, Sapporo University of Health Sciences, Sapporo, Japan
| | - Akiko Tamakoshi
- Department of Public Health, Hokkaido University Faculty of Medicine, Sapporo, Japan
| |
Collapse
|
6
|
Soares De Oliveira L, Ritter MJ. Thyroid hormone and the Liver. Hepatol Commun 2025; 9:e0596. [PMID: 39699315 DOI: 10.1097/hc9.0000000000000596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 10/16/2024] [Indexed: 12/20/2024] Open
Abstract
It is known that thyroid hormone can regulate hepatic metabolic pathways including cholesterol, de novo lipogenesis, fatty acid oxidation, lipophagy, and carbohydrate metabolism. Thyroid hormone action is mediated by the thyroid hormone receptor (THR) isoforms and their coregulators, and THRβ is the main isoform expressed in the liver. Dysregulation of thyroid hormone levels, as seen in hypothyroidism, has been associated with dyslipidemia and metabolic dysfunction-associated fatty liver disease. Given the beneficial effects of thyroid hormone in liver metabolism and the advances illuminating the use of thyroid hormone analogs such as resmetirom as therapeutic agents in the treatment of metabolic dysfunction-associated fatty liver disease, this review aims to further explore the relationship between TH, the liver, and metabolic dysfunction-associated fatty liver disease. Herein, we summarize the current clinical therapies and highlight future areas of research.
Collapse
Affiliation(s)
- Lorraine Soares De Oliveira
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston Medical Center, Boston, Massachusetts, USA
| | | |
Collapse
|
7
|
Li Y, Gao R, Yang Z, Zong H, Li Y. Liraglutide modulates lipid metabolism via ZBTB20-LPL pathway. Life Sci 2025; 360:123267. [PMID: 39608448 DOI: 10.1016/j.lfs.2024.123267] [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: 07/20/2024] [Revised: 11/15/2024] [Accepted: 11/19/2024] [Indexed: 11/30/2024]
Abstract
OBJECTIVE To investigate the mechanism of liraglutide affecting lipid metabolism by regulating lipolysis and lipogenesis in cells and ob/ob mice. METHODS 3 T3-L1 cells were treated with liraglutide in vitro, and differentially expressed genes were screened by RNA sequencing. Gene Ontology (GO) and KEGG (Kvoto Encyclopedia of Genes and Genomes) enrichment analyses identified target genes for lipid regulation of liraglutide. 3 T3-L1 preadipocytes were induced to differentiate into adipocytes using a "cocktail method". Western blot and immunofluorescence were used to detect the expression of target genes and the lipid regulatory effect of liraglutide. 3 T3-L1 preadipocytes were transfected with lentivirus overexpressing Zbtb20 to study its role in adipogenesis, and gene expression was analyzed by RT-qPCR and Western blot. In vivo, ob/ob mice were subcutaneously injected with liraglutide or saline for 4 weeks. Blood lipids, adipose tissue volume and adipocyte size were detected. Immunohistochemical analysis and RT-qPCR were used to detect the expression of target genes in adipose tissue. RESULTS Liraglutide reduced lipid droplets and TG levels and altered the expression of genes related to fatty acid metabolism, lipogenesis, fatty acid oxidation, and adipocyte browning. The results of PCR, Western blot and immunofluorescence confirmed that liraglutide could regulate the adipogenesis by downregulating the transcriptional suppressor ZBTB20, and overexpression of Zbtb20 inhibited the expression of LPL, the key enzyme for lipohydrolysis. CONCLUSIONS Liraglutide regulates lipid metabolism through ZBTB20-LPL pathway to reveal its molecular mechanism.
Collapse
Affiliation(s)
- Yue Li
- Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan- Hospital, Jinan, Shandong, China; Department of Medicine, Qilu Institute of Technology, Jinan, China.
| | - Rui Gao
- Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan- Hospital, Jinan, Shandong, China.
| | - Zhiyan Yang
- Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan- Hospital, Jinan, Shandong, China; Department of Pharmacy, Jining No.1 People's Hospital, Jining, China.
| | - Huiying Zong
- Department of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China.
| | - Yan Li
- Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan- Hospital, Jinan, Shandong, China.
| |
Collapse
|
8
|
Raudeniece J, Justamente I, Ozolina-Moll L, Sobolevs A, Zolovs M, Dela F, Reihmane D. Cardiorespiratory Fitness and Body Mass Index as Predictors of Metabolic Syndrome in Schoolchildren (PACH Study). Diabetes Metab Syndr Obes 2024; 17:4675-4687. [PMID: 39665085 PMCID: PMC11633296 DOI: 10.2147/dmso.s487309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 11/06/2024] [Indexed: 12/13/2024] Open
Abstract
Purpose Metabolic syndrome (MetS) has become a condition not rarely diagnosed in children and adolescents, leading to changes in physical and mental health. Simple and cost-effective screening methods applied in schools are needed to take preventive measures and reduce the risk of the development of MetS in children. Methods This prospective longitudinal study aims to investigate the prevalence of MetS and its risk factors in 8-10-year-old schoolchildren (46 boys and 60 girls) over 3 consecutive years. General Linear Mixed Model (GLMM) was used to assess the effect of recommended daily levels of moderate to vigorous physical activity (MVPA), body mass index (BMI), waist circumference (WC), cardiorespiratory fitness (CRF), and obesity level on a new set of orthogonal variables formed from various parameters of MetS (eg blood pressure (BP), lipid panel and glucose homeostasis) determined by Principal Component Analysis (PCA). Results The prevalence of MetS was 2% in the years 2017, 2018 and 2019, while in 2020 prevalence reached 7.7%. The most prevalent combination of criteria defining MetS syndrome in children was increased WC, BP, and blood triglycerides (TG). PCA identified non-high-density lipoprotein (non-HDL), low-density lipoprotein (LDL), and total cholesterol (TCHOL) as important predictors of metabolic syndrome (MetS). Additionally, cardiorespiratory fitness (CRF) and body mass index (BMI) were found to significantly influence the variance in MetS criteria. However, moderate to vigorous physical activity (MVPA) did not have a notable effect on the variance of these criteria. Conclusion The prevalence of MetS in children is increasing with age. Non-HDL turned out to be the most influential parameter across all principal components. The CRF, being accessible, simple to use, non-invasive and cost-effective, proved to be a superior predictor of variance of glucose homeostasis compared to BMI.
Collapse
Affiliation(s)
- Jelena Raudeniece
- Department of Human and Animal Physiology, Faculty of Biology, University of Latvia, Riga, Latvia
- Department of Human Physiology and Biochemistry, Riga Stradiņš University, Riga, Latvia
- Laboratory of Sports and Nutrition Research, Riga Stradiņš University, Riga, Latvia
| | - Ilze Justamente
- Department of Human and Animal Physiology, Faculty of Biology, University of Latvia, Riga, Latvia
- Department of Human Physiology and Biochemistry, Riga Stradiņš University, Riga, Latvia
- Laboratory of Sports and Nutrition Research, Riga Stradiņš University, Riga, Latvia
| | - Liga Ozolina-Moll
- Department of Human and Animal Physiology, Faculty of Biology, University of Latvia, Riga, Latvia
| | - Artjoms Sobolevs
- Department of Pathology, Faculty of Medicine, Riga Stradiņš University, Riga, Latvia
| | - Maksims Zolovs
- Statistics Unit, Riga Stradiņš University, Riga, Latvia
- Institute of Life Sciences and Technology, Department of Biosystematics, Daugavpils University, Daugavpils, Latvia
| | - Flemming Dela
- Department of Human Physiology and Biochemistry, Riga Stradiņš University, Riga, Latvia
- Xlab, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Dace Reihmane
- Department of Human Physiology and Biochemistry, Riga Stradiņš University, Riga, Latvia
- Laboratory of Sports and Nutrition Research, Riga Stradiņš University, Riga, Latvia
| |
Collapse
|
9
|
Rustamzadeh A, Sadigh N, Vahabi Z, Khamseh F, Mohebi N, Ghobadi Z, Moradi F. Effects silymarin and rosuvastatin on amyloid-carriers level in dyslipidemic Alzheimer's patients: A double-blind placebo-controlled randomized clinical trial. IBRO Neurosci Rep 2024; 17:108-121. [PMID: 39139290 PMCID: PMC11321388 DOI: 10.1016/j.ibneur.2024.07.002] [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: 03/06/2024] [Revised: 06/19/2024] [Accepted: 07/18/2024] [Indexed: 08/15/2024] Open
Abstract
Purpose The production/excretion rate of Amyloid-β (Aβ) is the basis of the plaque burden in alzheimer's disease (AD), which depends on both central and peripheral clearance. In this study, the effect of silymarin and rosuvastatin on serum markers and clinical outcomes in dyslipidemic AD patients was investigated. Methods Participants (n=36) were randomized to silymarin (140 mg), placebo, and rosuvastatin 10 mg orally three times a day for 6 months. Serum collection and clinical outcome tests were performed at baseline and after completion of treatment. Lipid profile markers, oxidative stress markers, Aβ1-42/Aβ1-40 ratio, and Soluble Low-density lipoprotein receptor-Related Protein-1 (sLRP1)/Soluble Receptor for Advanced Glycation End Products (sRAGE) ratio were measured. Results There was a statistically significant increase in Δ-high density lipoprotein (ΔHDL) between silymarin and placebo (P<0.000) and also between rosuvastatin and placebo (p=0.044). The level of Δ-triglycerides (ΔTG) in the silymarin group has a significant decrease compared to both the placebo and the rosuvastatin group (p<0.000 and p=0.036, respectively). The Δ-superoxide dismutase (ΔSOD) level in the silymarin group compared to placebo and rosuvastatin had a significant increase (p<0.000 and p=0.008, respectively). The ΔAβ1-42/Aβ1-40 in the silymarin group compared to both the placebo and rosuvastatin groups had a significant increase (p<0.05). There was an inverse relationship between ΔTG and ΔAβ1-42/Aβ1-40 (p=-0.493 and p=0.004). ΔAβ1-42/Aβ1-40 has a direct statistical relationship with ΔSOD marker (p=0.388 and p=0.031). Also, there was a direct correlation between the level of ΔAβ1-42/Aβ1-40 and ΔsLRP1/sRAGE (p=0.491 and p=0.005). Conclusion Our study showed the relationship between plasma lipids, especially ΔTG and ΔHDL, with ΔAβ1-42/Aβ1-40 in dyslipidemic AD patients, and modulation of these lipid factors can be used to monitor the response to treatments.
Collapse
Affiliation(s)
- Auob Rustamzadeh
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Department of Anatomical Sciences, Faculty of Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Nader Sadigh
- Department of Emergency Medicine, School of Medicine, Trauma and Injury Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Vahabi
- Department of Geriatric Medicine, Ziaeian Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Khamseh
- Department of Neurology, Faculty of Medicine, Islamic Azad University, Tehran, Iran
| | - Nafiseh Mohebi
- Department of Neurology, Rasool Akram Hospital, School of Medicine, Iran University of Medial Sciences, Tehran, Iran
| | - Zahra Ghobadi
- Neuroimaging and Clinical Biomarkers Research Group, Pars Darman Medical Imaging Center, Karaj, Iran
| | - Fatemeh Moradi
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Pan BY, Chen CS, Chen FY, Shen MY. Multifaceted Role of Apolipoprotein C3 in Cardiovascular Disease Risk and Metabolic Disorder in Diabetes. Int J Mol Sci 2024; 25:12759. [PMID: 39684468 PMCID: PMC11641554 DOI: 10.3390/ijms252312759] [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: 11/14/2024] [Revised: 11/26/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Apolipoprotein C3 (APOC3) plays a critical role in regulating triglyceride levels and serves as a key predictor of cardiovascular disease (CVD) risk, particularly in patients with diabetes. While APOC3 is known to inhibit lipoprotein lipase, recent findings reveal its broader influence across lipoprotein metabolism, where it modulates the structure and function of various lipoproteins. Therefore, this review examines the complex metabolic cycle of APOC3, emphasizing the impact of APOC3-containing lipoproteins on human metabolism, particularly in patients with diabetes. Notably, APOC3 affects triglyceride-rich lipoproteins and causes structural changes in high-, very low-, intermediate-, and low-density lipoproteins, thereby increasing CVD risk. Evidence suggests that elevated APOC3 levels-above the proposed safe range of 10-15 mg/dL-correlate with clinically significant CVD outcomes. Recognizing APOC3 as a promising biomarker for CVD, this review underscores the urgent need for high-throughput, clinically feasible methods to further investigate its role in lipoprotein physiology in both animal models and human studies. Additionally, we analyze the relationship between APOC3-related genes and lipoproteins, reinforcing the value of large-population studies to understand the impact of APOC3 on metabolic diseases. Ultimately, this review supports the development of therapeutic strategies targeting APOC3 reduction as a preventive approach for diabetes-related CVD.
Collapse
Affiliation(s)
- Bo-Yi Pan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan; (B.-Y.P.); (F.-Y.C.)
| | - Chen-Sheng Chen
- The Ph.D. Program for Cancer Biology and Drug Discovery, China Medical University and Academia Sinica, Taichung 40402, Taiwan;
| | - Fang-Yu Chen
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan; (B.-Y.P.); (F.-Y.C.)
| | - Ming-Yi Shen
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan; (B.-Y.P.); (F.-Y.C.)
- Department of Medical Research, China Medical University Hospital, Taichung 40402, Taiwan
- Department of Nursing, Asia University, Taichung 413305, Taiwan
| |
Collapse
|
11
|
Lee MS, Lee HY, Oh SH, Kim CB, Kim JH, Yoo SH, Yoo YJ, Lee SY, Lee BC. Salvia miltiorrhiza and Its Compounds as Complementary Therapy for Dyslipidemia: A Meta-Analysis of Clinical Efficacy and In Silico Mechanistic Insights. Pharmaceuticals (Basel) 2024; 17:1426. [PMID: 39598338 PMCID: PMC11597782 DOI: 10.3390/ph17111426] [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: 09/19/2024] [Revised: 10/10/2024] [Accepted: 10/21/2024] [Indexed: 11/29/2024] Open
Abstract
Background/Objectives: Dyslipidemia is a significant risk factor for atherosclerotic cardiovascular disease (ASCVD), a leading cause of death worldwide. Salvia miltiorrhiza Burge is widely used in East Asia for cardiovascular health, showing potential benefits in lowering cholesterol and reducing inflammation. Methods: This study systematically reviewed and conducted a meta-analysis of randomized controlled trials (RCTs) to assess the clinical effectiveness of Salvia miltiorrhiza in treating dyslipidemia. Moreover, network pharmacology and molecular docking analyses were performed to explore the mechanisms underlying the effects of Salvia miltiorrhiza. Results: The meta-analysis revealed that when Salvia miltiorrhiza is combined with statin therapy, it significantly enhances lipid profiles, including reductions in total cholesterol, low-density lipoprotein cholesterol (LDL-C), and triglycerides and improvements in high-density lipoprotein cholesterol (HDL-C), compared to statin therapy alone. The in silico analyses indicated that Salvia miltiorrhiza may influence key biological pathways, such as the PI3K/Akt, JAK/STAT, and HMGCR pathways, which are involved in inflammation, lipid metabolism, and the development of atherosclerosis. Conclusions:Salvia miltiorrhiza shows potential as a complementary therapy for dyslipidemia, offering additional lipid-lowering and anti-inflammatory benefits.
Collapse
Affiliation(s)
- Min-Seong Lee
- Department of Clinical Korean Medicine, Graduate School, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea; (M.-S.L.); (H.-Y.L.); (S.-H.O.); (C.-B.K.); (J.-H.K.); (S.-H.Y.); (Y.-J.Y.)
| | - Han-Young Lee
- Department of Clinical Korean Medicine, Graduate School, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea; (M.-S.L.); (H.-Y.L.); (S.-H.O.); (C.-B.K.); (J.-H.K.); (S.-H.Y.); (Y.-J.Y.)
| | - Seung-Hyun Oh
- Department of Clinical Korean Medicine, Graduate School, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea; (M.-S.L.); (H.-Y.L.); (S.-H.O.); (C.-B.K.); (J.-H.K.); (S.-H.Y.); (Y.-J.Y.)
| | - Chang-Bum Kim
- Department of Clinical Korean Medicine, Graduate School, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea; (M.-S.L.); (H.-Y.L.); (S.-H.O.); (C.-B.K.); (J.-H.K.); (S.-H.Y.); (Y.-J.Y.)
| | - Ji-Han Kim
- Department of Clinical Korean Medicine, Graduate School, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea; (M.-S.L.); (H.-Y.L.); (S.-H.O.); (C.-B.K.); (J.-H.K.); (S.-H.Y.); (Y.-J.Y.)
| | - Seung-Hoon Yoo
- Department of Clinical Korean Medicine, Graduate School, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea; (M.-S.L.); (H.-Y.L.); (S.-H.O.); (C.-B.K.); (J.-H.K.); (S.-H.Y.); (Y.-J.Y.)
| | - Yeon-Joo Yoo
- Department of Clinical Korean Medicine, Graduate School, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea; (M.-S.L.); (H.-Y.L.); (S.-H.O.); (C.-B.K.); (J.-H.K.); (S.-H.Y.); (Y.-J.Y.)
| | - Su-Yeon Lee
- College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Byung-Cheol Lee
- Department of Clinical Korean Medicine, Graduate School, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea; (M.-S.L.); (H.-Y.L.); (S.-H.O.); (C.-B.K.); (J.-H.K.); (S.-H.Y.); (Y.-J.Y.)
| |
Collapse
|
12
|
Antelo-Cea DA, Martínez-Rojas L, Cabrerizo-Ibáñez I, Roudi Rashtabady A, Hernández-Alvarez MI. Regulation of Mitochondrial and Peroxisomal Metabolism in Female Obesity and Type 2 Diabetes. Int J Mol Sci 2024; 25:11237. [PMID: 39457018 PMCID: PMC11508381 DOI: 10.3390/ijms252011237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
Obesity and type 2 diabetes (T2D) are widespread metabolic disorders that significantly impact global health today, affecting approximately 17% of adults worldwide with obesity and 9.3% with T2D. Both conditions are closely linked to disruptions in lipid metabolism, where peroxisomes play a pivotal role. Mitochondria and peroxisomes are vital organelles responsible for lipid and energy regulation, including the β-oxidation and oxidation of very long-chain fatty acids (VLCFAs), cholesterol biosynthesis, and bile acid metabolism. These processes are significantly influenced by estrogens, highlighting the interplay between these organelles' function and hormonal regulation in the development and progression of metabolic diseases, such as obesity, metabolic dysfunction-associated fatty liver disease (MAFLD), and T2D. Estrogens modulate lipid metabolism through interactions with nuclear receptors, like peroxisome proliferator-activated receptors (PPARs), which are crucial for maintaining metabolic balance. Estrogen deficiency, such as in postmenopausal women, impairs PPAR regulation, leading to lipid accumulation and increased risk of metabolic disorders. The disruption of peroxisomal-mitochondrial function and estrogen regulation exacerbates lipid imbalances, contributing to insulin resistance and ROS accumulation. This review emphasizes the critical role of these organelles and estrogens in lipid metabolism and their implications for metabolic health, suggesting that therapeutic strategies, including hormone replacement therapy, may offer potential benefits in treating and preventing metabolic diseases.
Collapse
Affiliation(s)
- Damián A. Antelo-Cea
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain; (D.A.A.-C.); (L.M.-R.); (I.C.-I.); (A.R.R.)
- IBUB Universitat de Barcelona—Institut de Biomedicina de la Universitat de Barcelona, 08028 Barcelona, Spain
| | - Laura Martínez-Rojas
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain; (D.A.A.-C.); (L.M.-R.); (I.C.-I.); (A.R.R.)
| | - Izan Cabrerizo-Ibáñez
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain; (D.A.A.-C.); (L.M.-R.); (I.C.-I.); (A.R.R.)
| | - Ayda Roudi Rashtabady
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain; (D.A.A.-C.); (L.M.-R.); (I.C.-I.); (A.R.R.)
- IBUB Universitat de Barcelona—Institut de Biomedicina de la Universitat de Barcelona, 08028 Barcelona, Spain
| | - María Isabel Hernández-Alvarez
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain; (D.A.A.-C.); (L.M.-R.); (I.C.-I.); (A.R.R.)
- IBUB Universitat de Barcelona—Institut de Biomedicina de la Universitat de Barcelona, 08028 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
13
|
Xu X, Gao J, Sun J, Liu R, Chen W. The role of metabolic factors in the association between obesity and cholelithiasis: A two-step, two-sample multivariable mendelian randomization study. Clinics (Sao Paulo) 2024; 79:100520. [PMID: 39427492 PMCID: PMC11533479 DOI: 10.1016/j.clinsp.2024.100520] [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: 08/30/2023] [Revised: 07/16/2024] [Accepted: 09/16/2024] [Indexed: 10/22/2024] Open
Abstract
BACKGROUND AND PURPOSE The extent to which the effects of BMI on cholelithiasis are mediated by metabolic factors (including blood pressure, blood lipids, body mass, and fasting blood glucose) is unclear. Therefore, in this study, the authors used genetic evidence to test the effects of these characteristics. METHODS Summary-level data for exposures and main outcomes were extracted from GWAS consortia. The authors used a two-step, two-sample Multivariable Mendelian Randomization (MVMR) analysis to illustrate the effect of BMI on cholelithiasis and a stepwise test method to quantify the possible mediating effects of cardiometabolic factors on cholelithiasis. RESULTS For each one-unit logarithmic increase in body mass index, the risk of cholelithiasis increased by 98 % (Odds Ratio [OR = 1.98], 95 % CI: 1.73 %‒2.28 %). After mediation analysis, the authors found that high-density lipoprotein and triglycerides were the main mediating factors, while the mediating effects of other metabolic factors were not significant. The total effect ratios of HDL and TG on cholelithiasis were 7.3 % (95 % CI: 8.51 %‒12.85 %) and 3.5 % (95 % CI: 3.59 %‒6.50 %), respectively. HDL and TG played a significant role in regulating cholelithiasis, but there was no evidence to show the regulatory effect of LDL on cholelithiasis. The total effects of BMI and triglycerides on cholelithiasis were 10.7 % and 5.0 %, respectively. CONCLUSION The authors found that among the metabolic factors evaluated, the decrease of HDL and the increase of TG mediated a high proportion of the effect of BMI on cholelithiasis. Therefore, intervention with these factors may reduce the increased risk of cholelithiasis in patients with high BMI.
Collapse
Affiliation(s)
- Xiangrong Xu
- Department of General Surgery, Kunshan Hospital Affiliated to Jiangsu University, Kunshan, PR China
| | - Jiawei Gao
- Department of General Surgery, Second Affiliated Hospital of Soochow University, Suzhou, PR China
| | - Jun Sun
- Department of General Surgery, Kunshan Hospital Affiliated to Jiangsu University, Kunshan, PR China
| | - Ruiwen Liu
- Department of General Surgery, Kunshan Hospital Affiliated to Jiangsu University, Kunshan, PR China.
| | - Wei Chen
- Department of General Surgery, Second Affiliated Hospital of Soochow University, Suzhou, PR China
| |
Collapse
|
14
|
Burks KH, Stitziel NO, Davidson NO. Molecular Regulation and Therapeutic Targeting of VLDL Production in Cardiometabolic Disease. Cell Mol Gastroenterol Hepatol 2024; 19:101409. [PMID: 39406347 PMCID: PMC11609389 DOI: 10.1016/j.jcmgh.2024.101409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 09/19/2024] [Accepted: 09/19/2024] [Indexed: 11/16/2024]
Abstract
There exists a complex relationship between steatotic liver disease (SLD) and atherosclerotic cardiovascular disease (CVD). CVD is a leading cause of morbidity and mortality among individuals with SLD, particularly those with metabolic dysfunction-associated SLD (MASLD), a significant proportion of whom also exhibit features of insulin resistance. Recent evidence supports an expanded role of very low-density lipoprotein (VLDL) in the pathogenesis of CVD in patients, both with and without associated metabolic dysfunction. VLDL represents the major vehicle for exporting neutral lipid from hepatocytes, with each particle containing one molecule of apolipoproteinB100 (APOB100). VLDL production becomes dysregulated under conditions characteristic of MASLD including steatosis and insulin resistance. Insulin resistance not only affects VLDL production but also mediates the pathogenesis of atherosclerotic CVD. VLDL assembly and secretion therefore represents an important pathway in the setting of cardiometabolic disease and offers several candidates for therapeutic targeting, particularly in metabolically complex patients with MASLD at increased risk of atherosclerotic CVD. Here we review the clinical significance as well as the translational and therapeutic potential of key regulatory steps impacting VLDL initiation, maturation, secretion, catabolism, and clearance.
Collapse
Affiliation(s)
- Kendall H Burks
- Division of Cardiology, Department of Medicine, Center for Cardiovascular Research, Washington University School of Medicine, Saint Louis, Missouri
| | - Nathan O Stitziel
- Division of Cardiology, Department of Medicine, Center for Cardiovascular Research, Washington University School of Medicine, Saint Louis, Missouri
| | - Nicholas O Davidson
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri.
| |
Collapse
|
15
|
Tauil RB, Golono PT, de Lima EP, de Alvares Goulart R, Guiguer EL, Bechara MD, Nicolau CCT, Yanaguizawa Junior JL, Fiorini AMR, Méndez-Sánchez N, Abenavoli L, Direito R, Valente VE, Laurindo LF, Barbalho SM. Metabolic-Associated Fatty Liver Disease: The Influence of Oxidative Stress, Inflammation, Mitochondrial Dysfunctions, and the Role of Polyphenols. Pharmaceuticals (Basel) 2024; 17:1354. [PMID: 39458995 PMCID: PMC11510109 DOI: 10.3390/ph17101354] [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: 09/26/2024] [Revised: 10/05/2024] [Accepted: 10/07/2024] [Indexed: 10/28/2024] Open
Abstract
Metabolic-Associated Fatty Liver Disease (MAFLD) is a clinical-pathological scenario that occurs due to the accumulation of triglycerides in hepatocytes which is considered a significant cause of liver conditions and contributes to an increased risk of death worldwide. Even though the possible causes of MAFLD can involve the interaction of genetics, hormones, and nutrition, lifestyle (diet and sedentary lifestyle) is the most influential factor in developing this condition. Polyphenols comprise many natural chemical compounds that can be helpful in managing metabolic diseases. Therefore, the aim of this review was to investigate the impact of oxidative stress, inflammation, mitochondrial dysfunction, and the role of polyphenols in managing MAFLD. Some polyphenols can reverse part of the liver damage related to inflammation, oxidative stress, or mitochondrial dysfunction, and among them are anthocyanin, baicalin, catechin, curcumin, chlorogenic acid, didymin, epigallocatechin-3-gallate, luteolin, mangiferin, puerarin, punicalagin, resveratrol, and silymarin. These compounds have actions in reducing plasma liver enzymes, body mass index, waist circumference, adipose visceral indices, lipids, glycated hemoglobin, insulin resistance, and the HOMA index. They also reduce nuclear factor-KB (NF-KB), interleukin (IL)-1β, IL-6, tumor necrosis factor-α (TNF-α), blood pressure, liver fat content, steatosis index, and fibrosis. On the other hand, they can improve HDL-c, adiponectin levels, and fibrogenesis markers. These results show that polyphenols are promising in the prevention and treatment of MAFLD.
Collapse
Affiliation(s)
- Raissa Bulaty Tauil
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
| | - Paula Takano Golono
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
| | - Enzo Pereira de Lima
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
| | - Ricardo de Alvares Goulart
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
| | - Elen Landgraf Guiguer
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
- Department of Biochemistry and Nutrition, School of Food and Technology of Marília (FATEC), Marília 17500-000, São Paulo, Brazil
| | - Marcelo Dib Bechara
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
| | - Claudia C. T. Nicolau
- Department of Biochemistry and Nutrition, School of Food and Technology of Marília (FATEC), Marília 17500-000, São Paulo, Brazil
| | - José Luiz Yanaguizawa Junior
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
| | - Adriana M. R. Fiorini
- Department of Biochemistry and Nutrition, School of Food and Technology of Marília (FATEC), Marília 17500-000, São Paulo, Brazil
| | - Nahum Méndez-Sánchez
- Liver Research Unit, Medica Sur Clinic & Foundation, Mexico City 14050, Mexico;
- Faculty of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico
| | - Ludovico Abenavoli
- Department of Health Sciences, University “Magna Graecia”, Viale Europa, 88100 Catanzaro, Italy;
| | - Rosa Direito
- Laboratory of Systems Integration Pharmacology, Clinical and Regulatory Science, Research Institute for Medicines, Universidade de Lisboa (iMed.ULisboa), Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal;
| | - Vitor Engrácia Valente
- Autonomic Nervous System Center, School of Philosophy and Sciences, São Paulo State University, Marília 17525-902, São Paulo, Brazil
| | - Lucas Fornari Laurindo
- Department of Biochemistry and Pharmacology, School of Medicine, Faculdade de Medicina de Marília (FAMEMA), Marília 17519-030, São Paulo, Brazil;
| | - Sandra Maria Barbalho
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
- Department of Biochemistry and Nutrition, School of Food and Technology of Marília (FATEC), Marília 17500-000, São Paulo, Brazil
- Research Coordination, UNIMAR Charity Hospital, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
| |
Collapse
|
16
|
Alami F, Mousavi Shalmani SH, Mahmoudi Z, Nooriani N, Mousavi Z, Amjadi A, Masoumvand M, Mohajerani M, Abbasi Mobarakeh K, Harsini AR, Shafaei H, Omidi S, Khoshdooz S, Doaei S, Khosravi M. Comparison of the Effect of Omega-3 vs. MCT Supplementation on Iron-Related Indices in Patients Undergoing Dialysis. Biol Trace Elem Res 2024:10.1007/s12011-024-04349-4. [PMID: 39377959 DOI: 10.1007/s12011-024-04349-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 08/15/2024] [Indexed: 10/09/2024]
Abstract
Chronic kidney disease (CKD), characterized by progressive kidney failure, significantly increases mortality and comorbidity risks such as anemia. This study contrasts the impacts of omega-3 and medium-chain triglycerides (MCT) oil on levels of iron, ferritin, total iron-binding capacity (TIBC), hemoglobin (Hb), and transferrin saturation in patients with CKD undergoing dialysis. This interventional trial was conducted on 120 patients with CKD undergoing dialysis in Rasht, Iran. For 8 weeks, the omega-3 group was orally administered three 1000-mg capsules of omega-3 fatty acid supplement, and the MCT group was administered three 1000-mg capsules containing MCT oil daily. Serum concentrations of ferritin, iron, TIBC, Hb, and transferrin saturation were assessed pre-intervention and after the intervention. There was a significant increase in serum iron levels in the MCT group compared to the omega-3 group (103.72 ± 57.8 vs. 77.48±40.13; P = 0.031). No effect was found regarding other iron-related factors such as TIBC, Hb, transferrin saturation, and ferritin levels. The results of our study indicated that taking MCT oil increased serum iron levels compared to omega-3 supplementation in patients with CKD undergoing dialysis. Further research is needed to better understand the potential benefits of MCT oils in patients with CKD.
Collapse
Affiliation(s)
- Farkhondeh Alami
- Student Research Committee, Department of Nutrition, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | | | - Zahra Mahmoudi
- Department of Nutrition, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Narjes Nooriani
- Department of Community Nutrition, School of Nutrition and Food Sciences, Nutrition and Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Zahra Mousavi
- Nursing and Midwifery school, Shahed University, Tehran, Iran
| | - Arezoo Amjadi
- Department of Nutrition, School of Nutritional Sciences and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohammad Masoumvand
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Malikeh Mohajerani
- Department of Nutrition, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Khadijeh Abbasi Mobarakeh
- Department of Community Nutrition, School of Nutrition and Food Sciences, Nutrition and Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Asma Rajabi Harsini
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Hanieh Shafaei
- Master's Student in Pediatric Nursing, Shahid Beheshti College of Midwifery, Guilan University of Medical Sciences, Rasht, Iran
| | - Saeed Omidi
- School of Health, Guilan University of Medical Sciences, Rasht, Iran
| | - Sara Khoshdooz
- Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Saeid Doaei
- Department of Community Nutrition, Faculty of Nutrition Sciences and Food Technology, School of Nutrition and Food Sciences, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Masoud Khosravi
- Urology Research Center, Razi Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran.
| |
Collapse
|
17
|
Zhao Y, Mao W, Liu B, Wang YF, Zhang SY, Guo LL, Qian YH, Gong ZG, Zhao JM, Yang XL, Qu GG, Hasi SR, Bai YT, Cao JS. Preparation of ceftiofur-encapsulated hen-egg low-density lipoproteins and their antibacterial effects on intracellular Staphylococcus aureus. Int J Biol Macromol 2024; 278:134840. [PMID: 39217040 DOI: 10.1016/j.ijbiomac.2024.134840] [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: 02/28/2024] [Revised: 08/03/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024]
Abstract
Hen egg low-density lipoprotein (heLDL), as alternative of serum-derived LDL, was used as drug delivery system of ceftiofur (CEF). The CEF-loaded hen egg low-density lipoprotein (CEF-heLDL) with complete apolipoprotein structure and high drug loading rate was synthesized, possesses suitable particle size. CEF-heLDL undergoes cellular uptake and colocalizes with lysosomes in vitro. An intracellular infection model of the bovine endometrial epithelial cells and a coeliac-induced inflammation model of mice by Staphylococcus aureus (S. aureus) were established, and significantly lower intracellular S. aureus levels of CEF-heLDL group than CEF-free group (P < 0.001) was observed. The antibacterial efficacy was sustained for 24 h. Up to 400 mg/kg of CEF-heLDL, 20 times the clinical practice, were intraperitoneally administrated, and no significant toxicity signs on mice were observed. HeLDLs is an effective, safe, and cheap drug carrier, and could also be used for transmembrane delivering other antibiotics.
Collapse
Affiliation(s)
- Yi Zhao
- Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot City, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot City, China
| | - Wei Mao
- Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot City, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot City, China
| | - Bo Liu
- Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot City, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot City, China
| | - Yong-Fei Wang
- Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot City, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot City, China; Inner Mongolia Medical University, Hohhot 010030, China
| | - Shuang-Yi Zhang
- Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot City, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot City, China
| | - Li-Li Guo
- Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot City, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot City, China
| | - Ying-Hong Qian
- Inner Mongolia Academy of Agricultural & Animal Husbandry Science, Hohhot 010010, China
| | - Zhi-Guo Gong
- Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot City, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot City, China
| | - Jia-Min Zhao
- Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot City, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot City, China
| | - Xiao-Lin Yang
- Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot City, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot City, China
| | - Gang-Gang Qu
- Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot City, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot City, China
| | - Su-Rong Hasi
- Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot City, China.
| | - Yu-Ting Bai
- Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot City, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot City, China.
| | - Jin-Shan Cao
- Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot City, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot City, China.
| |
Collapse
|
18
|
Liu Y, Li T, Xu J, Li S, Li B, Elgozair M. Apolipoprotein H deficiency exacerbates alcohol-induced liver injury via gut Dysbiosis and altered bile acid metabolism. Biochim Biophys Acta Mol Cell Biol Lipids 2024; 1869:159535. [PMID: 39033850 DOI: 10.1016/j.bbalip.2024.159535] [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: 03/15/2024] [Revised: 07/06/2024] [Accepted: 07/14/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND APOH plays an essential role in lipid metabolism and the transport of lipids in the circulation. Previous studies have shown that APOH deficiency causes fatty liver and gut microbiota dysbiosis in mouse models. However, the role and potential mechanisms of APOH deficiency in the pathogenesis of alcoholic liver disease remain unclear. METHODS C57BL/6 WT and ApoH-/- mice were used to construct the binge-on-chronic alcohol feeding model. Mouse liver transcriptome, targeted bile acid metabolome, and 16S gut bacterial taxa were assayed and analyzed. Open-source human liver transcriptome dataset was analyzed. RESULTS ApoH-/- mice fed with alcohol showed severe hepatic steatosis. Liver RNAseq and RT-qPCR data indicated that APOH deficiency predominantly impacts hepatic lipid metabolism by disrupting de novo lipogenesis, cholesterol processing, and bile acid metabolism. A targeted bile acid metabolomics assay indicated significant changes in bile acid composition, including increased percentages of TCA in the liver and DCA in the gut of alcohol-fed ApoH-/- mice. The concentrations of CA, NorCA, and HCA in the liver were higher in ApoH-/- mice on an ethanol diet compared to the control mice (p < 0.05). Additionally, APOH deficiency altered the composition of gut flora, which correlated with changes in the liver bile acid composition in the ethanol-feeding mouse model. Finally, open-source transcript-level data from human ALD livers highlighted a remarkable link between APOH downregulation and steatohepatitis, as well as bile acid metabolism. CONCLUSION APOH deficiency aggravates alcohol induced hepatic steatosis through the disruption of gut microbiota homeostasis and bile acid metabolism in mice.
Collapse
Affiliation(s)
- Yaming Liu
- Department of Gastroenterology and Hepatology, The Second Hospital of Dalian Medical University, Dalian, Liaoning Province, China 116011; Department of Digestive Diseases, School of Medicine, Xiamen University, Xiamen, Fujian Province, China 361001.
| | - Tingting Li
- Department of Digestive Diseases, School of Medicine, Xiamen University, Xiamen, Fujian Province, China 361001
| | - Jun Xu
- Clinical Center of Immune-Mediated Digestive Diseases, Peking University People's Hospital, Beijing, China 100044
| | - Shanshan Li
- The Fourth Liver Disease Center, Beijing YouAn Hospital, Capital Medical University, Beijing, China 100069
| | - Binbin Li
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA 55902
| | - Mohamad Elgozair
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA 55902
| |
Collapse
|
19
|
Zhang L, Wang X, Chen XW. The biogenesis and transport of triglyceride-rich lipoproteins. Trends Endocrinol Metab 2024:S1043-2760(24)00196-6. [PMID: 39164120 DOI: 10.1016/j.tem.2024.07.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/16/2024] [Accepted: 07/19/2024] [Indexed: 08/22/2024]
Abstract
Triglyceride-rich lipoproteins (TRLs) play essential roles in human health and disease by transporting bulk lipids into the circulation. This review summarizes the fundamental mechanisms and diverse factors governing lipoprotein production, secretion, and regulation. Emphasizing the broader implications for human health, we outline the intricate landscape of lipoprotein research and highlight the potential coordination between the biogenesis and transport of TRLs in physiology, particularly the unexpected coupling of metabolic enzymes and transport machineries. Challenges and opportunities in lipoprotein biology with respect to inherited diseases and viral infections are also discussed. Further characterization of the biogenesis and transport of TRLs will advance both basic research in lipid biology and translational medicine for metabolic diseases.
Collapse
Affiliation(s)
- Linqi Zhang
- State Key Laboratory of Membrane Biology, Peking University, Beijing 100871, PR China; Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, PR China
| | - Xiao Wang
- State Key Laboratory of Membrane Biology, Peking University, Beijing 100871, PR China; Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, PR China.
| | - Xiao-Wei Chen
- State Key Laboratory of Membrane Biology, Peking University, Beijing 100871, PR China; Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, PR China; Peking University (PKU)-Tsinghua University (THU) Joint Center for Life Sciences, Peking University, Beijing 100871, PR China.
| |
Collapse
|
20
|
Huang CH, Hsu HS, Chiang MT. Influence of Varied Dietary Cholesterol Levels on Lipid Metabolism in Hamsters. Nutrients 2024; 16:2472. [PMID: 39125351 PMCID: PMC11314022 DOI: 10.3390/nu16152472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 07/25/2024] [Accepted: 07/27/2024] [Indexed: 08/12/2024] Open
Abstract
Syrian hamsters are valuable models for studying lipid metabolism due to their sensitivity to dietary cholesterol, yet the precise impact of varying cholesterol levels has not been comprehensively assessed. This study examined the impact of varying dietary cholesterol levels on lipid metabolism in Syrian hamsters. Diets ranging from 0% to 1% cholesterol were administered to assess lipid profiles and oxidative stress markers. Key findings indicate specific cholesterol thresholds for inducing distinct lipid profiles: below 0.13% for normal lipids, 0.97% for elevated LDL-C, 0.43% for increased VLDL-C, and above 0.85% for heightened hepatic lipid accumulation. A cholesterol supplementation of 0.43% induced hypercholesterolemia without adverse liver effects or abnormal lipoprotein expression. Furthermore, cholesterol supplementation significantly increased liver weight, plasma total cholesterol, LDL-C, and VLDL-C levels while reducing the HDL-C/LDL-C ratio. Fecal cholesterol excretion increased, with stable bile acid levels. High cholesterol diets correlated with elevated plasma ALT activities, reduced hepatic lipid peroxidation, and altered leptin and CETP levels. These findings underscore Syrian hamsters as robust models for hyperlipidemia research, offering insights into experimental methodologies. The identified cholesterol thresholds facilitate precise lipid profile manipulation, enhancing the hamster's utility in lipid metabolism studies and potentially informing clinical approaches to managing lipid disorders.
Collapse
Affiliation(s)
| | | | - Meng-Tsan Chiang
- Department of Food Science, College of Life Sciences, National Taiwan Ocean University, Keelung 20224, Taiwan; (C.-H.H.); (H.-S.H.)
| |
Collapse
|
21
|
Díaz-Grijuela E, Hernández A, Caballero C, Fernandez R, Urtasun R, Gulak M, Astigarraga E, Barajas M, Barreda-Gómez G. From Lipid Signatures to Cellular Responses: Unraveling the Complexity of Melanoma and Furthering Its Diagnosis and Treatment. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1204. [PMID: 39202486 PMCID: PMC11356604 DOI: 10.3390/medicina60081204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 07/19/2024] [Accepted: 07/22/2024] [Indexed: 09/03/2024]
Abstract
Recent advancements in mass spectrometry have significantly enhanced our understanding of complex lipid profiles, opening new avenues for oncological diagnostics. This review highlights the importance of lipidomics in the comprehension of certain metabolic pathways and its potential for the detection and characterization of various cancers, in particular melanoma. Through detailed case studies, we demonstrate how lipidomic analysis has led to significant breakthroughs in the identification and understanding of cancer types and its potential for detecting unique biomarkers that are instrumental in its diagnosis. Additionally, this review addresses the technical challenges and future perspectives of these methodologies, including their potential expansion and refinement for clinical applications. The discussion underscores the critical role of lipidomic profiling in advancing cancer diagnostics, proposing a new paradigm in how we approach this devastating disease, with particular emphasis on its application in comparative oncology.
Collapse
Affiliation(s)
| | | | | | - Roberto Fernandez
- IMG Pharma Biotech, Research and Development Division, 48170 Zamudio, Spain;
| | - Raquel Urtasun
- Biochemistry Area, Department of Health Science, Universidad Pública de Navarra, 31006 Pamplona, Spain; (R.U.); (M.B.)
| | | | - Egoitz Astigarraga
- Betternostics SL, 31110 Noáin, Spain; (E.D.-G.); (A.H.); (C.C.)
- IMG Pharma Biotech, Research and Development Division, 48170 Zamudio, Spain;
| | - Miguel Barajas
- Biochemistry Area, Department of Health Science, Universidad Pública de Navarra, 31006 Pamplona, Spain; (R.U.); (M.B.)
| | - Gabriel Barreda-Gómez
- Betternostics SL, 31110 Noáin, Spain; (E.D.-G.); (A.H.); (C.C.)
- IMG Pharma Biotech, Research and Development Division, 48170 Zamudio, Spain;
| |
Collapse
|
22
|
Zhao F, Wang J, Zhang Y, Hu J, Li C, Liu S, Li R, Du R. In vivo Fate of Targeted Drug Delivery Carriers. Int J Nanomedicine 2024; 19:6895-6929. [PMID: 39005963 PMCID: PMC11246094 DOI: 10.2147/ijn.s465959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 06/22/2024] [Indexed: 07/16/2024] Open
Abstract
This review aimed to systematically investigate the intracellular and subcellular fate of various types of targeting carriers. Upon entering the body via intravenous injection or other routes, a targeting carrier that can deliver therapeutic agents initiates their journey. If administered intravenously, the carrier initially faces challenges presented by the blood circulation before reaching specific tissues and interacting with cells within the tissue. At the subcellular level, the car2rier undergoes processes, such as drug release, degradation, and metabolism, through specific pathways. While studies on the fate of 13 types of carriers have been relatively conclusive, these studies are incomplete and lack a comprehensive analysis. Furthermore, there are still carriers whose fate remains unclear, underscoring the need for continuous research. This study highlights the importance of comprehending the in vivo and intracellular fate of targeting carriers and provides valuable insights into the operational mechanisms of different carriers within the body. By doing so, researchers can effectively select appropriate carriers and enhance the successful clinical translation of new formulations.
Collapse
Affiliation(s)
- Fan Zhao
- Engineering Research Center of Modern Preparation Technology of TCM, Ministry of Education, Shanghai, 201203, People’s Republic of China
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| | - Jitong Wang
- Engineering Research Center of Modern Preparation Technology of TCM, Ministry of Education, Shanghai, 201203, People’s Republic of China
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| | - Yu Zhang
- Engineering Research Center of Modern Preparation Technology of TCM, Ministry of Education, Shanghai, 201203, People’s Republic of China
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| | - Jinru Hu
- Engineering Research Center of Modern Preparation Technology of TCM, Ministry of Education, Shanghai, 201203, People’s Republic of China
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| | - Chenyang Li
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, 518055, People’s Republic of China
| | - Shuainan Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Key Laboratory of Polymorphic Drugs of Beijing, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, People’s Republic of China
- Diabetes Research Center of Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Ruixiang Li
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| | - Ruofei Du
- Engineering Research Center of Modern Preparation Technology of TCM, Ministry of Education, Shanghai, 201203, People’s Republic of China
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| |
Collapse
|
23
|
Albitar O, D'Souza CM, Adeghate EA. Effects of Lipoproteins on Metabolic Health. Nutrients 2024; 16:2156. [PMID: 38999903 PMCID: PMC11243180 DOI: 10.3390/nu16132156] [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: 05/21/2024] [Revised: 06/28/2024] [Accepted: 07/03/2024] [Indexed: 07/14/2024] Open
Abstract
Lipids are primarily transported in the bloodstream by lipoproteins, which are macromolecules of lipids and conjugated proteins also known as apolipoproteins. The processes of lipoprotein assembly, secretion, transportation, modification, and clearance are crucial components of maintaining a healthy lipid metabolism. Disruption in any of these steps results in pathophysiological abnormalities such as dyslipidemia, obesity, insulin resistance, inflammation, atherosclerosis, peripheral artery disease, and cardiovascular diseases. By studying these genetic mutations, researchers can gain valuable insights into the underlying mechanisms that govern the relationship between protein structure and its physiological role. These lipoproteins, including HDL, LDL, lipoprotein(a), and VLDL, mainly serve the purpose of transporting lipids between tissues and organs. However, studies have provided evidence that apo(a) also possesses protective properties against pathogens. In the future, the field of study will be significantly influenced by the integration of recombinant DNA technology and human site-specific mutagenesis for treating hereditary disorders. Several medications are available for the treatment of dyslipoproteinemia. These include statins, fibrates, ezetimibe, niacin, PCSK9 inhibitors, evinacumab, DPP 4 inhibitors, glucagon-like peptide-1 receptor agonists GLP1RAs, GLP-1, and GIP dual receptor agonists, in addition to SGLT2 inhibitors. This current review article exhibits, for the first time, a comprehensive reflection of the available body of publications concerning the impact of lipoproteins on metabolic well-being across various pathological states.
Collapse
Affiliation(s)
- Obaida Albitar
- Department of Pharmacology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates
| | - Crystal M D'Souza
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates
| | - Ernest A Adeghate
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates
| |
Collapse
|
24
|
Li A, Hu H, Huang Y, Yang F, Mi Q, Jin L, Liu H, Zhang Q, Pan H. Effects of dietary metabolizable energy level on hepatic lipid metabolism and cecal microbiota in aged laying hens. Poult Sci 2024; 103:103855. [PMID: 38796988 PMCID: PMC11153248 DOI: 10.1016/j.psj.2024.103855] [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: 12/08/2023] [Revised: 04/27/2024] [Accepted: 05/10/2024] [Indexed: 05/29/2024] Open
Abstract
Lipid metabolic capacity, feed utilization, and the diversity of gut microbiota are reduced in the late laying stage for laying hens. This experiment aimed to investigate the effects of different levels of dietary metabolizable energy (ME) on hepatic lipid metabolism and cecal microbiota in late laying hens. The 216 Peking Pink laying hens (57-wk-old) were randomly assigned to experimental diets of 11.56 (HM = high ME), 11.14 (MM = medium ME), or 10.72 (LM = low ME) MJ of ME/kg, with each dietary treatment containing 6 replicates per group and 12 chickens per replicate. The HM group showed higher triglyceride (TG), total cholesterol (T-CHO), and low-density lipoprotein cholesterol (LDL-C) concentrations in the liver compared with the LM group; second, the HM group showed higher TG concentration and the LM group showed lower T-CHO concentration compared with MM group; finally, the HM group showed a lower hepatic lipase (HL) activity compared with the MM and LM groups (P < 0.05). There was a significant difference in the microbial community structure of the cecum between the HM and MM groups (P < 0.05). The decrease of dietary ME level resulted in a gradual decrease relative abundance of Proteobacteria. At the genus level, beneficial bacteria were significantly enriched in the LM group compared to the MM group, including Faecalibacterium, Lactobacillus, and Bifidobacterium, (linear discriminant analysis [LDA] >2, P <0.05). In addition, at the species level, Lactobacillus crispatus, Parabacteroides gordonii, Blautia caecimuris, and Lactobacillus johnsonii were significantly enriched in the LM group (LDA>2, P < 0.05). The HM group had a higher abundance of Sutterella spp. compared to the LM group (LDA>2, P <0.05). In conclusion, this research suggests that the reduction in dietary energy level did not adversely affect glycolipid metabolism or low dietary ME (10.72 MJ/kg). The findings can be helpful for maintaining intestinal homeostasis and increasing benefit for gut microbiota in late laying hens.
Collapse
Affiliation(s)
- Anjian Li
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
| | - Hong Hu
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
| | - Ying Huang
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
| | - Fuyan Yang
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
| | - Qianhui Mi
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
| | - Liqiang Jin
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
| | - Hongli Liu
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
| | - Qiang Zhang
- WOD Poultry Research Institute, Beijing, 100193, China
| | - Hongbin Pan
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, China; WOD Poultry Research Institute, Beijing, 100193, China.
| |
Collapse
|
25
|
Zaid AB, Awad SM, El-Abd MG, Saied SA, Almahdy SK, Saied AA, Elmalawany AM, AboShabaan HS, Saleh HS. Unraveling the controversy between fasting and nonfasting lipid testing in a normal population: a systematic review and meta-analysis of 244,665 participants. Lipids Health Dis 2024; 23:199. [PMID: 38937752 PMCID: PMC11210154 DOI: 10.1186/s12944-024-02169-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: 02/22/2024] [Accepted: 05/29/2024] [Indexed: 06/29/2024] Open
Abstract
BACKGROUND The final decision to fast or not fast for routine lipid profile examination in a standard, healthy population is unclear. Whereas the United States and European protocols state that fasting for regular lipid analysis is unnecessary, the North American and Chinese guidelines still recommend fasting before routine lipid testing. AIM This study aimed to unravel the contradiction between the different protocols of lipid profile testing worldwide and clarify the effect of diet on lipid profile testing only in a regular, healthy population. METHODS A literature search was conducted through May 2024. The analyses included studies performed from the date 2000 until now because the contradiction of guidelines for lipid profile testing appeared for the first time in this period. A planned internal validity evaluation was performed using the National Institute of Health (NIH) quality measurement tools for observational cohort, case‒control, controlled interventional, and cross-sectional studies. The data were synthesized according to RevMan 5.3. RESULTS Eight studies with a total of 244,665 participants were included. The standardized mean difference in cholesterol in six studies showed significant differences in overall effect among fasting and nonfasting states (P < 0.00001), as did high-density lipoprotein cholesterol (P < 0.00001). At the same time, with respect to triglycerides and low-density lipoprotein cholesterol, there were notable variations in the overall effect between the fasted and nonfasted states (P < 0.00001 and P ≤ 0.001, respectively). CONCLUSIONS This meta-analysis concluded that fasting for lipid profile testing is preferred as a conservative model to reduce variability and increase consistency in patients' metabolic status when sampling for lipid testing.
Collapse
Affiliation(s)
- Ahmed B Zaid
- Department of Clinical Pathology, National Liver Institute Hospital, Menoufia Univerisity, Shebin Elkoom, Egypt.
| | - Samah M Awad
- Department of Clinical Microbiology, National Liver Institute, Menoufia University, Shibin Elkom, 32511, Egypt
| | - Mona G El-Abd
- Department of Clinical Pathology, National Liver Institute, Menoufia University, Shibin Elkom, 32511, Egypt
| | - Sara A Saied
- Department of Clinical Pathology, National Liver Institute, Menoufia University, Shibin Elkom, 32511, Egypt
| | - Shimaa K Almahdy
- Department of Hepatology and Gastroenterology, National Liver Institute, Menoufia University, Shibin Elkom, 32511, Egypt
| | | | - Alshimaa M Elmalawany
- Department of Clinical Pathology, National Liver Institute Hospital, Menoufia Univerisity, Shebin Elkoom, Egypt
| | - Hind S AboShabaan
- Department of Clinical Pathology, National Liver Institute Hospital, Menoufia Univerisity, Shebin Elkoom, Egypt
| | - Helmy S Saleh
- Department of Microbiology, Animal Health Research Institute, Shibin Elkom, 32511, Egypt
| |
Collapse
|
26
|
Zhang Y, Wang X, Liu T, Zhang ZY, Song WG, Guo SD. Exserolide J ameliorates lipid accumulation in vitro by regulating liver X receptor alpha and peroxisome proliferator-activated receptor alpha proteins. Heliyon 2024; 10:e31861. [PMID: 38947487 PMCID: PMC11214467 DOI: 10.1016/j.heliyon.2024.e31861] [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: 01/08/2024] [Revised: 05/22/2024] [Accepted: 05/22/2024] [Indexed: 07/02/2024] Open
Abstract
Exserolides are isocoumarin derivatives containing lactone moiety. Recently, some isocoumarins have been demonstrated to ameliorate hyperlipidemia, a major factor for inducing cardiovascular diseases. However, the effects and mechanisms of action of exserolides on hyperlipidemia are not known. The aim of this study is to investigate whether the marine fungus Setosphaeria sp.-derived exserolides (compounds I, J, E, and F) exert lipid-lowering effects via improving reverse cholesterol transport (RCT) in vitro. RAW264.7 macrophages and HepG2 cells were used to establish lipid-laden models, and the levels of intracellular lipids and RCT-related proteins were determined by assay kits and Western blotting, respectively. We observed that exserolides (at a 5 μM concentration) significantly decreased intracellular cholesterol and triglyceride levels in oxidized low-density lipoprotein-laden RAW264.7 cells and markedly improved [3H]-cholesterol efflux. Among the four tested compounds, exserolide J increased the protein levels of ATP-binding cassette transporter A1, peroxisome proliferator-activated receptor α (PPARα), and liver X receptor α (LXRα). Furthermore, treatment with exserolides significantly decreased oleic acid-laden lipid accumulation in HepG2 hepatocytes. Mechanistically, exserolides enhance PPARα protein levels; furthermore, compound J increases cholesterol 7 alpha-hydroxylase A1 and LXRα protein levels. Molecular docking revealed that exserolides, particularly compound J, can interact with PPARα and LXRα proteins. These data suggest that the terminal carboxyl group of compound J plays a key role in lowering lipid levels by stimulating LXRα and PPARα proteins. In conclusion, compound J exhibits powerful lipid-lowering effects in vitro. However, its hypolipidemic effects in vivo should be investigated in the future.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Endocrinology and Metabolism, Guiqian International General Hospital, Guiyang, 550018, China
| | - Xue Wang
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Shandong Second Medical University, Weifang, 261053, China
| | - Tian Liu
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Shandong Second Medical University, Weifang, 261053, China
| | - Zi-Yi Zhang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Wen-Gang Song
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, China
| | - Shou-Dong Guo
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Shandong Second Medical University, Weifang, 261053, China
| |
Collapse
|
27
|
Huang X, Xue Z, Zhang D, Lee HJ. Pinpointing Fat Molecules: Advances in Coherent Raman Scattering Microscopy for Lipid Metabolism. Anal Chem 2024; 96:7945-7958. [PMID: 38700460 DOI: 10.1021/acs.analchem.4c01398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2024]
Affiliation(s)
- Xiangjie Huang
- College of Biomedical Engineering & Instrument Science, and Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou 310027, China
| | - Zexin Xue
- College of Biomedical Engineering & Instrument Science, and Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou 310027, China
| | - Delong Zhang
- MOE Frontier Science Center for Brain Science & Brain-Machine Integration, Zhejiang University, Hangzhou 310027, China
- Zhejiang Key Laboratory of Micro-nano Quantum Chips and Quantum Control, and School of Physics, Zhejiang University, Hangzhou 310027, China
| | - Hyeon Jeong Lee
- College of Biomedical Engineering & Instrument Science, and Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou 310027, China
- MOE Frontier Science Center for Brain Science & Brain-Machine Integration, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
28
|
Gao N, Liu Y, Liu G, Liu B, Cheng Y. Sanghuangporus vaninii extract ameliorates hyperlipidemia in rats by mechanisms identified with transcriptome analysis. Food Sci Nutr 2024; 12:3360-3376. [PMID: 38726415 PMCID: PMC11077191 DOI: 10.1002/fsn3.4002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 01/18/2024] [Accepted: 01/21/2024] [Indexed: 05/12/2024] Open
Abstract
The increasing incidence of hyperlipidemia is a serious threat to public health. The development of effective and safe lipid-lowering drugs with few side effects is necessary. The purpose of this study was to assess the lipid-lowering activity of Sanghuangporus vaninii extract (SVE) in rat experiments and reveal the molecular mechanism by transcriptome analysis. Hyperlipidemia was induced in the animals using a high-fat diet for 4 weeks. At the end of the 4th week, hyperlipidemic rats were assigned into two control groups (model and positive simvastatin control) and three treatment groups that received SVE at 200, 400, or 800 mg kg-1 day-1 for another 4 weeks. A last control group comprised normal chow-fed rats. At the end of the 8th week, rats were sacrificed and lipid serum levels, histopathology, and liver transcriptome profiles were determined. SVE was demonstrated to relieve the lipid disorder and improve histopathological liver changes in a dose-dependent manner. The transcriptomic analysis identified changes in hepatocyte gene activity for major pathways including steroid biosynthesis, bile secretion, cholesterol metabolism, AMPK signaling, thyroid hormone signaling, and glucagon signaling. The changed expression of crucial genes in the different groups was confirmed by qPCR. Collectively, this study revealed that SVE could relieve hyperlipidemia in rats, the molecular mechanism might be to promote the metabolism of lipids and the excretion of cholesterol, inhibit the biosynthesis of cholesterol by activating the AMPK signaling pathway, the thyroid hormone signaling pathway, and the glucagon signaling pathway.
Collapse
Affiliation(s)
- Ning Gao
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of EducationHarbinChina
- School of PharmacyHeilongjiang University of Chinese MedicineHarbinChina
| | - Yuanzhen Liu
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of EducationHarbinChina
- School of PharmacyHeilongjiang University of Chinese MedicineHarbinChina
| | - Guangjie Liu
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of EducationHarbinChina
- School of PharmacyHeilongjiang University of Chinese MedicineHarbinChina
| | - Bo Liu
- School of Pharmaceutical EngineeringHeilongjiang Agricultural Reclamation Vocational CollegeHarbinChina
| | - Yupeng Cheng
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of EducationHarbinChina
- School of PharmacyHeilongjiang University of Chinese MedicineHarbinChina
| |
Collapse
|
29
|
Zeng T, Lv J, Liang J, Xie B, Liu L, Tan Y, Zhu J, Jiang J, Xie H. Zebrafish cobll1a regulates lipid homeostasis via the RA signaling pathway. Front Cell Dev Biol 2024; 12:1381362. [PMID: 38699158 PMCID: PMC11063382 DOI: 10.3389/fcell.2024.1381362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 04/04/2024] [Indexed: 05/05/2024] Open
Abstract
Background The COBLL1 gene has been implicated in human central obesity, fasting insulin levels, type 2 diabetes, and blood lipid profiles. However, its molecular mechanisms remain largely unexplored. Methods In this study, we established cobll1a mutant lines using the CRISPR/Cas9-mediated gene knockout technique. To further dissect the molecular underpinnings of cobll1a during early development, transcriptome sequencing and bioinformatics analysis was employed. Results Our study showed that compared to the control, cobll1a -/- zebrafish embryos exhibited impaired development of digestive organs, including the liver, intestine, and pancreas, at 4 days post-fertilization (dpf). Transcriptome sequencing and bioinformatics analysis results showed that in cobll1a knockout group, the expression level of genes in the Retinoic Acid (RA) signaling pathway was affected, and the expression level of lipid metabolism-related genes (fasn, scd, elovl2, elovl6, dgat1a, srebf1 and srebf2) were significantly changed (p < 0.01), leading to increased lipid synthesis and decreased lipid catabolism. The expression level of apolipoprotein genes (apoa1a, apoa1b, apoa2, apoa4a, apoa4b, and apoea) genes were downregulated. Conclusion Our study suggest that the loss of cobll1a resulted in disrupted RA metabolism, reduced lipoprotein expression, and abnormal lipid transport, therefore contributing to lipid accumulation and deleterious effects on early liver development.
Collapse
Affiliation(s)
- Ting Zeng
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Changsha, Hunan, China
| | - Jinrui Lv
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Changsha, Hunan, China
| | - Jiaxin Liang
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Changsha, Hunan, China
| | - Binling Xie
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Changsha, Hunan, China
| | - Ling Liu
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Changsha, Hunan, China
| | - Yuanyuan Tan
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Changsha, Hunan, China
| | - Junwei Zhu
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Changsha, Hunan, China
| | - Jifan Jiang
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Changsha, Hunan, China
| | - Huaping Xie
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Changsha, Hunan, China
| |
Collapse
|
30
|
Pascual-Oliver A, Casas-Deza D, Yagüe-Caballero C, Arbones-Mainar JM, Bernal-Monterde V. Lipid Profile and Cardiovascular Risk Modification after Hepatitis C Virus Eradication. Pathogens 2024; 13:278. [PMID: 38668233 PMCID: PMC11054742 DOI: 10.3390/pathogens13040278] [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: 02/05/2024] [Revised: 03/18/2024] [Accepted: 03/21/2024] [Indexed: 04/29/2024] Open
Abstract
The eradication of the hepatitis C virus (HCV) has revolutionized the hepatology paradigm, halting the progression of advanced liver disease in patients with chronic infection and reducing the risk of hepatocarcinoma. In addition, treatment with direct-acting antivirals can reverse the lipid and carbohydrate abnormalities described in HCV patients. Although HCV eradication may reduce the overall risk of vascular events, it is uncertain whether altered lipid profiles increase the risk of cerebrovascular disease in certain patients. We have conducted a review on HCV and lipid and carbohydrate metabolism, as well as new scientific advances, following the advent of direct-acting antivirals.
Collapse
Affiliation(s)
- Andrea Pascual-Oliver
- Gastroenterology Department, Miguel Servet University Hospital, 50009 Zaragoza, Spain; (A.P.-O.); (C.Y.-C.); (V.B.-M.)
| | - Diego Casas-Deza
- Gastroenterology Department, Miguel Servet University Hospital, 50009 Zaragoza, Spain; (A.P.-O.); (C.Y.-C.); (V.B.-M.)
- Adipocyte and Fat Biology Laboratory (AdipoFat), Translational Research Unit, University Hospital Miguel Servet, 50009 Zaragoza, Spain;
- Instituto Aragones de Ciencias de la Salud (IACS), 50009 Zaragoza, Spain
- Instituto de Investigación Sanitaria (IIS) Aragon, 50009 Zaragoza, Spain
| | - Carmen Yagüe-Caballero
- Gastroenterology Department, Miguel Servet University Hospital, 50009 Zaragoza, Spain; (A.P.-O.); (C.Y.-C.); (V.B.-M.)
- Adipocyte and Fat Biology Laboratory (AdipoFat), Translational Research Unit, University Hospital Miguel Servet, 50009 Zaragoza, Spain;
- Instituto de Investigación Sanitaria (IIS) Aragon, 50009 Zaragoza, Spain
| | - Jose M. Arbones-Mainar
- Adipocyte and Fat Biology Laboratory (AdipoFat), Translational Research Unit, University Hospital Miguel Servet, 50009 Zaragoza, Spain;
- Instituto Aragones de Ciencias de la Salud (IACS), 50009 Zaragoza, Spain
- Instituto de Investigación Sanitaria (IIS) Aragon, 50009 Zaragoza, Spain
- CIBER Fisiopatología Obesidad y Nutrición (CIBERObn), Instituto Salud Carlos III, 28029 Madrid, Spain
| | - Vanesa Bernal-Monterde
- Gastroenterology Department, Miguel Servet University Hospital, 50009 Zaragoza, Spain; (A.P.-O.); (C.Y.-C.); (V.B.-M.)
- Adipocyte and Fat Biology Laboratory (AdipoFat), Translational Research Unit, University Hospital Miguel Servet, 50009 Zaragoza, Spain;
- Instituto Aragones de Ciencias de la Salud (IACS), 50009 Zaragoza, Spain
- Instituto de Investigación Sanitaria (IIS) Aragon, 50009 Zaragoza, Spain
| |
Collapse
|
31
|
Song X, Ye T, Jing D, Wei K, Ge Y, Bei X, Qi Y, Wang H, Li J, Zhang Y. Association between exposure to per- and polyfluoroalkyl substances and levels of lipid profile based on human studies. REVIEWS ON ENVIRONMENTAL HEALTH 2024; 0:reveh-2023-0146. [PMID: 38408126 DOI: 10.1515/reveh-2023-0146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 02/02/2024] [Indexed: 02/28/2024]
Abstract
Epidemiological evidence suggests that exposure to per- and polyfluoroalkyl substances (PFAS) is associated with lipid profile levels, but with inconsistent conclusions from different studies. The aim of this study was to conduct a meta-analysis of the relationship between PFAS exposure and lipid profile levels based on population-based epidemiological studies. Embase, PubMed, Ovid database, The Cochrane Library and Web of Science database were used to search appropriate studies (before September 6, 2022) on the correlation between PFAS exposure and lipid profile levels. β value, odd ratio (OR) and 95 % confidence intervals (CIs) were extracted from studies. In this study, we found that higher low-density lipoprotein (LDL) levels were associated with exposure to perfluoroundecanoic acid (PFUnDA) (β value=0.13, 95 % CIs: 0.02, 0.24) and perfluorooctane sulfonic acid (PFOS) (β value=0.13, 95 % CIs: 0.04, 0.21). PFOA, PFOS and PFNA exposure were significantly related to the higher levels of total cholesterol (TC) with the pooled effect estimates of 0.08 (95 % CI: 0.02, 0.14), 0.13 (95 % CI: 0.05, 0.21) and 0.14 (95 % CI: 0.08, 0.20) respectively. In sum, our results identified that PFOA, PFOS, PFNA and PFUnDA were the most important risk factors for abnormal levels of lipid profile, indicating that we should prevent cerebrovascular disease by reducing and controlling PFAS exposure.
Collapse
Affiliation(s)
- Xinru Song
- Department of General Surgery, Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Tingtao Ye
- Department of Public Health, Kangda College of Nanjing Medical University, Lianyungang, China
| | - Dongmei Jing
- Muchunyuan Nursing Home of Jiangsu Province Official Hospital, Nanjing, China
| | - Kai Wei
- Department of Public Health, Kangda College of Nanjing Medical University, Lianyungang, China
| | - Yue Ge
- Department of Public Health, Kangda College of Nanjing Medical University, Lianyungang, China
| | - Xinyue Bei
- Department of Public Health, Kangda College of Nanjing Medical University, Lianyungang, China
| | - Yuqian Qi
- Department of Public Health, Kangda College of Nanjing Medical University, Lianyungang, China
| | - Huanqiang Wang
- Department of Public Health, Kangda College of Nanjing Medical University, Lianyungang, China
| | - Jun Li
- Department of General Surgery, Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Yan Zhang
- Department of Public Health, Kangda College of Nanjing Medical University, Lianyungang, China
| |
Collapse
|
32
|
Zammit VA, Park SO. In Vivo Monitoring of Glycerolipid Metabolism in Animal Nutrition Biomodel-Fed Smart-Farm Eggs. Foods 2024; 13:722. [PMID: 38472835 DOI: 10.3390/foods13050722] [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: 01/17/2024] [Revised: 02/20/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
Although many studies have examined the biochemical metabolic pathways by which an egg (egg yolk) lowers blood lipid levels, data on the molecular biological mechanisms that regulate and induce the partitioning of hepatic glycerolipids are missing. The aim of this study was to investigate in vivo monitoring in four study groups using an animal nutrition biomodel fitted with a jugular-vein cannula after egg yolk intake: CON (control group, oral administration of 1.0 g of saline), T1 (oral administration of 1.0 g of pork belly fat), T2 (oral administration of 1.0 g of smart-farm egg yolk), and T3 (oral administration of T1 and T2 alternately every week). The eggs induced significant and reciprocal changes in incorporating 14C lipids into the total glycerolipids and releasing 14CO2, thereby regulating esterification and accelerating oxidation in vivo. The eggs increased phospholipid secretion from the liver into the blood and decreased triacylglycerol secretion by regulating the multiple cleavage of fatty acyl-CoA moieties' fluxes. In conclusion, the results of the current study reveal the novel fact that eggs can lower blood lipids by lowering triacylglycerol secretion in the biochemical metabolic pathway of hepatic glycerolipid partitioning while simultaneously increasing phospholipid secretion and 14CO2 emission.
Collapse
Affiliation(s)
- Victor A Zammit
- Metabolic Biochemistry, Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
| | - Sang O Park
- Institute of Animal Life Science, Kangwon National University, Chuncheon-si 24341, Gangwon State, Republic of Korea
| |
Collapse
|
33
|
Morvaridzadeh M, Zoubdane N, Heshmati J, Alami M, Berrougui H, Khalil A. High-Density Lipoprotein Metabolism and Function in Cardiovascular Diseases: What about Aging and Diet Effects? Nutrients 2024; 16:653. [PMID: 38474781 DOI: 10.3390/nu16050653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/20/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
Cardiovascular diseases (CVDs) have become the leading global cause of mortality, prompting a heightened focus on identifying precise indicators for their assessment and treatment. In this perspective, the plasma levels of HDL have emerged as a pivotal focus, given the demonstrable correlation between plasma levels and cardiovascular events, rendering them a noteworthy biomarker. However, it is crucial to acknowledge that HDLs, while intricate, are not presently a direct therapeutic target, necessitating a more nuanced understanding of their dynamic remodeling throughout their life cycle. HDLs exhibit several anti-atherosclerotic properties that define their functionality. This functionality of HDLs, which is independent of their concentration, may be impaired in certain risk factors for CVD. Moreover, because HDLs are dynamic parameters, in which HDL particles present different atheroprotective properties, it remains difficult to interpret the association between HDL level and CVD risk. Besides the antioxidant and anti-inflammatory activities of HDLs, their capacity to mediate cholesterol efflux, a key metric of HDL functionality, represents the main anti-atherosclerotic property of HDL. In this review, we will discuss the HDL components and HDL structure that may affect their functionality and we will review the mechanism by which HDL mediates cholesterol efflux. We will give a brief examination of the effects of aging and diet on HDL structure and function.
Collapse
Affiliation(s)
- Mojgan Morvaridzadeh
- Department of Medicine, Geriatric Service, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 4N4, Canada
| | - Nada Zoubdane
- Department of Medicine, Geriatric Service, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 4N4, Canada
| | - Javad Heshmati
- Department of Medicine, Geriatric Service, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 4N4, Canada
| | - Mehdi Alami
- Department of Medicine, Geriatric Service, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 4N4, Canada
| | - Hicham Berrougui
- Department of Medicine, Geriatric Service, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 4N4, Canada
| | - Abdelouahed Khalil
- Department of Medicine, Geriatric Service, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 4N4, Canada
| |
Collapse
|
34
|
Konaklieva MI, Plotkin BJ. Targeting host-specific metabolic pathways-opportunities and challenges for anti-infective therapy. Front Mol Biosci 2024; 11:1338567. [PMID: 38455763 PMCID: PMC10918472 DOI: 10.3389/fmolb.2024.1338567] [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: 11/14/2023] [Accepted: 01/24/2024] [Indexed: 03/09/2024] Open
Abstract
Microorganisms can takeover critical metabolic pathways in host cells to fuel their replication. This interaction provides an opportunity to target host metabolic pathways, in addition to the pathogen-specific ones, in the development of antimicrobials. Host-directed therapy (HDT) is an emerging strategy of anti-infective therapy, which targets host cell metabolism utilized by facultative and obligate intracellular pathogens for entry, replication, egress or persistence of infected host cells. This review provides an overview of the host lipid metabolism and links it to the challenges in the development of HDTs for viral and bacterial infections, where pathogens are using important for the host lipid enzymes, or producing their own analogous of lecithin-cholesterol acyltransferase (LCAT) and lipoprotein lipase (LPL) thus interfering with the human host's lipid metabolism.
Collapse
Affiliation(s)
| | - Balbina J. Plotkin
- Department of Microbiology and Immunology, Midwestern University, Downers Grove, IL, United States
| |
Collapse
|
35
|
Sun J, Xie F, Wang J, Luo J, Chen T, Jiang Q, Xi Q, Liu GE, Zhang Y. Integrated meta-omics reveals the regulatory landscape involved in lipid metabolism between pig breeds. MICROBIOME 2024; 12:33. [PMID: 38374121 PMCID: PMC10877772 DOI: 10.1186/s40168-023-01743-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 12/19/2023] [Indexed: 02/21/2024]
Abstract
BACKGROUND Domesticated pigs serve as an ideal animal model for biomedical research and also provide the majority of meat for human consumption in China. Porcine intramuscular fat content associates with human health and diseases and is essential in pork quality. The molecular mechanisms controlling lipid metabolism and intramuscular fat accretion across tissues in pigs, and how these changes in response to pig breeds, remain largely unknown. RESULTS We surveyed the tissue-resident cell types of the porcine jejunum, colon, liver, and longissimus dorsi muscle between Lantang and Landrace breeds by single-cell RNA sequencing. Combining lipidomics and metagenomics approaches, we also characterized gene signatures and determined key discriminating markers of lipid digestibility, absorption, conversion, and deposition across tissues in two pig breeds. In Landrace, lean-meat swine mainly exhibited breed-specific advantages in lipid absorption and oxidation for energy supply in small and large intestinal epitheliums, nascent high-density lipoprotein synthesis for reverse cholesterol transport in enterocytes and hepatocytes, bile acid formation, and secretion for fat emulsification in hepatocytes, as well as intestinal-microbiota gene expression involved in lipid accumulation product. In Lantang, obese-meat swine showed a higher synthesis capacity of chylomicrons responsible for high serum triacylglycerol levels in small intestinal epitheliums, the predominant characteristics of lipid absorption in muscle tissue, and greater intramuscular adipcytogenesis potentials from muscular fibro-adipogenic progenitor subpopulation. CONCLUSIONS The findings enhanced our understanding of the cellular biology of lipid metabolism and opened new avenues to improve animal production and human diseases. Video Abstract.
Collapse
Affiliation(s)
- Jiajie Sun
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Fang Xie
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Jing Wang
- Institute of Animal Husbandry and Veterinary Medicine, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, China
| | - Junyi Luo
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Ting Chen
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Qingyan Jiang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Qianyun Xi
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China.
| | - George E Liu
- Animal Genomics and Improvement Laboratory, USDA-ARS, BARC-East, Beltsville, MD, 20705, USA.
| | - Yongliang Zhang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China.
| |
Collapse
|
36
|
Wu Z, Jiang Y, Guo Z, Li P, Zheng Y, Wang Y, Zhang H, Balmer L, Li X, Tao L, Zhang Q, Gao B, Guo X. Remnant cholesterol traits and risk of stroke: A multivariable Mendelian randomization study. PNAS NEXUS 2024; 3:pgae033. [PMID: 38380054 PMCID: PMC10877093 DOI: 10.1093/pnasnexus/pgae033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 01/09/2024] [Indexed: 02/22/2024]
Abstract
Observational epidemiological studies have reported a relationship between remnant cholesterol and stroke. However, the results are inconclusive, and causality remains unclear due to confounding or reverse causality. Our objective in this study was to investigate the causal relevance of remnant cholesterol and the risk of stroke and its subtypes using the Mendelian randomization (MR) approach. Genome-wide association studies (GWASs) including 115,082 European individuals (UK Biobank) were used to identify instruments for remnant cholesterol, including intermediate-density lipoprotein (IDL) cholesterol and very-low-density lipoprotein (VLDL) cholesterol. Summary-level data for total stroke, intracerebral hemorrhage, subarachnoid hemorrhage, ischemic stroke (IS), and IS subtypes were obtained from GWAS meta-analyses conducted by the MEGASTROKE consortium. Univariable and multivariable MR analyses were performed. The GWAS identified multiple single-nucleotide polymorphisms after clumping for remnant cholesterol (n = 52), IDL cholesterol (n = 62), and VLDL cholesterol (n = 67). Assessed individually using MR, remnant cholesterol (weighted median: odds ratio [OR] 1.32 per 1-SD higher trait; 95% CI: 1.04-1.67; P = 0.024) had effect estimates consistent with a higher risk of LAS-IS, driven by IDL cholesterol (OR 1.32; 95% CI: 1.04-1.68; P = 0.022). In multivariable MR, IDL cholesterol (OR 1.46; 95% CI: 1.10-1.93; P = 0.009) retained a robust effect on LAS-IS after controlling for VLDL cholesterol and high-density lipoprotein cholesterol. The MR analysis did not indicate causal associations between remnant cholesterol and other stroke subtypes. This study suggests that remnant cholesterol is causally associated with the risk of LAS-IS driven by IDL cholesterol.
Collapse
Affiliation(s)
- Zhiyuan Wu
- Beijing Municipal Key Laboratory of Clinical Epidemiology, School of Public Health, Capital Medical University, Beijing 100069, China
- Centre for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA 6027, Australia
| | - Yue Jiang
- Beijing Municipal Key Laboratory of Clinical Epidemiology, School of Public Health, Capital Medical University, Beijing 100069, China
| | - Zheng Guo
- Centre for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA 6027, Australia
| | - Pingan Li
- Beijing Municipal Key Laboratory of Clinical Epidemiology, School of Public Health, Capital Medical University, Beijing 100069, China
| | - Yulu Zheng
- Centre for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA 6027, Australia
| | - Yutao Wang
- Centre of Xunshu, Shanghai Fufan Information Technology Co., Ltd, Shanghai 200433, China
| | - Haiping Zhang
- Beijing Municipal Key Laboratory of Clinical Epidemiology, School of Public Health, Capital Medical University, Beijing 100069, China
| | - Lois Balmer
- Centre for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA 6027, Australia
| | - Xingang Li
- Centre for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA 6027, Australia
| | - Lixin Tao
- Beijing Municipal Key Laboratory of Clinical Epidemiology, School of Public Health, Capital Medical University, Beijing 100069, China
| | - Qi Zhang
- Department of Informatics, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Bo Gao
- Beijing Municipal Key Laboratory of Clinical Epidemiology, School of Public Health, Capital Medical University, Beijing 100069, China
| | - Xiuhua Guo
- Beijing Municipal Key Laboratory of Clinical Epidemiology, School of Public Health, Capital Medical University, Beijing 100069, China
| |
Collapse
|
37
|
El-Darzi N, Mast N, Li Y, Pikuleva IA. APOB100 transgenic mice exemplify how the systemic circulation content may affect the retina without altering retinal cholesterol input. Cell Mol Life Sci 2024; 81:52. [PMID: 38253888 PMCID: PMC10803575 DOI: 10.1007/s00018-023-05056-4] [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: 09/08/2023] [Revised: 10/24/2023] [Accepted: 11/17/2023] [Indexed: 01/24/2024]
Abstract
Apolipoprotein B (APOB) is a constituent of unique lipoprotein particles (LPPs) produced in the retinal pigment epithelium (RPE), which separates the neural retina from Bruch's membrane (BrM) and choroidal circulation. These LPPs accumulate with age in BrM and contribute to the development of age-related macular degeneration, a major blinding disease. The APOB100 transgenic expression in mice, which unlike humans lack the full-length APOB100, leads to lipid deposits in BrM. Herein, we further characterized APOB100 transgenic mice. We imaged mouse retina in vivo and assessed chorioretinal lipid distribution, retinal sterol levels, retinal cholesterol input, and serum content as well as tracked indocyanine green-bound LPPs in mouse plasma and retina after an intraperitoneal injection. Retinal function and differentially expressed proteins were also investigated. APOB100 transgenic mice had increased serum LDL content and an additional higher density HDL subpopulation; their retinal cholesterol levels (initially decreased) became normal with age. The LPP cycling between the RPE and choroidal circulation was increased. Yet, LPP trafficking from the RPE to the neural retina was limited, and total retinal cholesterol input did not change. There were lipid deposits in the RPE and BrM, and retinal function was impaired. Retinal proteomics provided mechanistic insights. Collectively, our data suggested that the serum LDL/HDL ratio may not affect retinal pathways of cholesterol input as serum LPP load is mainly handled by the RPE, which offloads LPP excess to the choroidal circulation rather than neural retina. Different HDL subpopulations should be considered in studies linking serum LPPs and age-related macular degeneration.
Collapse
Affiliation(s)
- Nicole El-Darzi
- Department of Ophthalmology and Visual Science, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Natalia Mast
- Department of Ophthalmology and Visual Science, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Yong Li
- Department of Ophthalmology and Visual Science, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Irina A Pikuleva
- Department of Ophthalmology and Visual Science, Case Western Reserve University, Cleveland, OH, 44106, USA.
| |
Collapse
|
38
|
Yang CR, Lin WJ, Shen PC, Liao PY, Dai YC, Hung YC, Lai HC, Mehmood S, Cheng WC, Ma WL. Phenotypic and metabolomic characteristics of mouse models of metabolic associated steatohepatitis. Biomark Res 2024; 12:6. [PMID: 38195587 PMCID: PMC10777576 DOI: 10.1186/s40364-023-00555-9] [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: 09/30/2023] [Accepted: 12/29/2023] [Indexed: 01/11/2024] Open
Abstract
BACKGROUND Metabolic associated steatohepatitis (MASH) is metabolic disease that may progress to cirrhosis and hepatocellular carcinoma. Mouse models of diet-induced MASH, which is characterized by the high levels of fats, sugars, and cholesterol in diets, are commonly used in research. However, mouse models accurately reflecting the progression of MASH in humans remain to be established. Studies have explored the potential use of serological metabolites as biomarkers of MASH severity in relation to human MASH. METHODS We performed a comparative analysis of three mouse models of diet-induced MASH in terms of phenotypic and metabolomic characteristics; MASH was induced using different diets: a high-fat diet; a Western diet; and a high-fat, high-cholesterol diet. Liver cirrhosis was diagnosed using standard clinical approaches (e.g., METAVIR score, hyaluronan level, and collagen deposition level). Mouse serum samples were subjected to nuclear magnetic resonance spectroscopy-based metabolomic profiling followed by bioinformatic analyses. Metabolomic analysis of a retrospective cohort of patients with hepatocellular carcinoma was performed; the corresponding cirrhosis scores were also evaluated. RESULTS Using clinically relevant quantitative diagnostic methods, the severity of MASH was evaluated. Regarding metabolomics, the number of lipoprotein metabolites increased with both diet and MASH progression. Notably, the levels of very low-density lipoprotein (VLDL) and low-density lipoprotein (LDL) significantly increased with fibrosis progression. During the development of diet-induced MASH in mice, the strongest upregulation of expression was noted for VLDL receptor. Metabolomic analysis of a retrospective cohort of patients with cirrhosis indicated lipoproteins (e.g., VLDL and LDL) as predominant biomarkers of cirrhosis. CONCLUSIONS Our findings provide insight into the pathophysiology and metabolomics of experimental MASH and its relevance to human MASH. The observed upregulation of lipoprotein expression reveals a feedforward mechanism for MASH development that may be targeted for the development of noninvasive diagnosis.
Collapse
Affiliation(s)
- Cian-Ru Yang
- Program for Health Science and Industry, Graduate Institute of Biomedical Sciences, and Department of Medicine, and Tumor Biology Center, School of Medicine, China Medical University, Taichung, Taiwan
- Department of Medical Research, Department of Gynecology and Obstetrics, and Department of Gastroenterology, China Medical University Hospital, Taichung, Taiwan
| | - Wen-Jen Lin
- Program for Health Science and Industry, Graduate Institute of Biomedical Sciences, and Department of Medicine, and Tumor Biology Center, School of Medicine, China Medical University, Taichung, Taiwan
- Department of Medical Research, Department of Gynecology and Obstetrics, and Department of Gastroenterology, China Medical University Hospital, Taichung, Taiwan
| | - Pei-Chun Shen
- Program for Health Science and Industry, Graduate Institute of Biomedical Sciences, and Department of Medicine, and Tumor Biology Center, School of Medicine, China Medical University, Taichung, Taiwan
- Department of Medical Research, Department of Gynecology and Obstetrics, and Department of Gastroenterology, China Medical University Hospital, Taichung, Taiwan
| | - Pei-Yin Liao
- Program for Health Science and Industry, Graduate Institute of Biomedical Sciences, and Department of Medicine, and Tumor Biology Center, School of Medicine, China Medical University, Taichung, Taiwan
- Department of Medical Research, Department of Gynecology and Obstetrics, and Department of Gastroenterology, China Medical University Hospital, Taichung, Taiwan
| | - Yuan-Chang Dai
- Department of Pathology, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chia-Yi City, Taiwan
| | - Yao-Ching Hung
- Department of Gynecology and Obstetrics, Asia University Hospital, Taichung, Taiwan
| | - Hsueh-Chou Lai
- Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
- Center for Digestive Medicine, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Shiraz Mehmood
- Program for Health Science and Industry, Graduate Institute of Biomedical Sciences, and Department of Medicine, and Tumor Biology Center, School of Medicine, China Medical University, Taichung, Taiwan
| | - Wei-Chung Cheng
- Program for Health Science and Industry, Graduate Institute of Biomedical Sciences, and Department of Medicine, and Tumor Biology Center, School of Medicine, China Medical University, Taichung, Taiwan.
| | - Wen-Lung Ma
- Program for Health Science and Industry, Graduate Institute of Biomedical Sciences, and Department of Medicine, and Tumor Biology Center, School of Medicine, China Medical University, Taichung, Taiwan.
- Department of Medical Research, Department of Gynecology and Obstetrics, and Department of Gastroenterology, China Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
39
|
Liu Y, Zhao Y, Liu Q, Li B, Daniel PV, Chen B, Wu Z. Effects of apolipoprotein H downregulation on lipid metabolism, fatty liver disease, and gut microbiota dysbiosis. J Lipid Res 2024; 65:100483. [PMID: 38101620 PMCID: PMC10818206 DOI: 10.1016/j.jlr.2023.100483] [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: 10/03/2023] [Revised: 11/27/2023] [Accepted: 12/04/2023] [Indexed: 12/17/2023] Open
Abstract
Apolipoprotein H (APOH) downregulation can cause hepatic steatosis and gut microbiota dysbiosis. However, the mechanism by which APOH-regulated lipid metabolism contributes to metabolic dysfunction-associated steatotic liver disease (MASLD) remains undetermined. Herein, we aim to explore the regulatory effect of APOH, mediated through various pathways, on metabolic homeostasis and MASLD pathogenesis. We analyzed serum marker levels, liver histopathology, and cholesterol metabolism-related gene expression in global ApoH-/- C57BL/6 male mice. We used RNA sequencing and metabolomic techniques to investigate the association between liver metabolism and bacterial composition. Fifty-two differentially expressed genes were identified between ApoH-/- and WT mice. The mRNA levels of de novo lipogenesis genes were highly upregulated in ApoH-/- mice than in WT mice. Fatty acid, glycerophospholipid, sterol lipid, and triglyceride levels were elevated, while hyodeoxycholic acid levels were significantly reduced in the liver tissues of ApoH-/- mice than in those of WT mice. Microbial beta diversity was lower in ApoH-/- mice than in WT mice, and gut microbiota metabolic functions were activated in ApoH-/- mice. Moreover, ApoH transcripts were downregulated in patients with MASLD, and APOH-related differential genes were enriched in lipid metabolism. Open-source transcript-level data from human metabolic dysfunction-associated steatohepatitis livers reinforced a significant association between metabolic dysfunction-associated steatohepatitis and APOH downregulation. In conclusion, our studies demonstrated that APOH downregulation aggravates fatty liver and induces gut microbiota dysbiosis by dysregulating bile acids. Our findings offer a novel perspective on APOH-mediated lipid metabolic dysbiosis and provide a valuable framework for deciphering the role of APOH in fatty liver disease.
Collapse
Affiliation(s)
- Yaming Liu
- Department of Gastroenterology and Hepatology, Xiamen University Zhongshan Hospital, Xiamen, FJ, China; Department of Digestive Diseases, School of Medicine, Xiamen University, Xiamen, FJ, China.
| | - Yiqun Zhao
- Department of Gastroenterology and Hepatology, Xiamen University Zhongshan Hospital, Xiamen, FJ, China; Department of Digestive Diseases, School of Medicine, Xiamen University, Xiamen, FJ, China
| | - Qiusong Liu
- Department of Tumor and Vascular Interventional Radiology, Xiamen University Zhongshan Hospital, Xiamen, FJ, China
| | - Binbin Li
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - P Vineeth Daniel
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Binbin Chen
- Department of Digestive Diseases, School of Medicine, Xiamen University, Xiamen, FJ, China
| | - Zeyi Wu
- Department of Computer Science and Engineering, University of California, San Diego, CA, USA
| |
Collapse
|
40
|
Ou-Yang YN, Deng FF, Wang YJ, Chen M, Yang PF, Yang Z, Tian Z. High-salt diet induces dyslipidemia through the SREBP2/PCSK9 pathway in dahl salt-sensitive rats. Biochimie 2024; 216:34-45. [PMID: 37844755 DOI: 10.1016/j.biochi.2023.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 09/24/2023] [Accepted: 10/02/2023] [Indexed: 10/18/2023]
Abstract
A high-salt diet is known to increase serum cholesterol levels; however, the underlying mechanism of salt-induced dyslipidemia in patients with salt-sensitivity remains poorly understood. We aimed to investigate whether high-salt diet (HSD) can induce dyslipidemia and elucidate the underlying mechanism of salt-induced dyslipidemia in Dahl salt-sensitive (SS) rats. Metabolomic and biochemical analyses revealed that the consumption of an HSD (8 % NaCl) significantly increased the serum levels of total cholesterol (TC) and low-density lipoprotein cholesterol (LDL-C) in SS rats. The enzyme-linked immunosorbent assay demonstrated an increase in circulating proprotein convertase subtilisin/kexin type 9 (PCSK9) levels, accompanied by a decrease in hepatic low-density lipoprotein receptor (LDLR) levels due to HSD consumption. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and Western blot analysis revealed that HSD consumption activated sterol regulatory element-binding protein-2 (SREBP2) expression in the liver and kidney, resulting in upregulation of PCSK9 at the transcriptional level in the liver and at the translational level in the kidney, ultimately increasing circulating PCSK9 levels. The combined effects of HSD on the liver and kidney contributed to the development of hypercholesterolemia. Furthermore, an in vitro assay confirmed that high-salt exposure led to an increase in the protein expression of SREBP2 and PCSK9 secretion, thereby reducing low-density lipoprotein (LDL) uptake. This study, for the first time, shows that an HSD induces dyslipidemia through activation of the SREBP2/PCSK9 pathway, providing new insights into the prevention and treatment of dyslipidemia in patients with salt sensitivity.
Collapse
Affiliation(s)
- Ya-Nan Ou-Yang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China.
| | - Fen-Fen Deng
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China.
| | - Yun-Jia Wang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China.
| | - Meng Chen
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China.
| | - Peng-Fei Yang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China.
| | - Zhe Yang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China.
| | - Zhongmin Tian
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China.
| |
Collapse
|
41
|
Wang L, Wang Z, Chen Y, Cao J. Effects of monochromatic light on hepatic glycogen and lipid synthesis in broilers. Poult Sci 2024; 103:103193. [PMID: 37931402 PMCID: PMC10654228 DOI: 10.1016/j.psj.2023.103193] [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: 08/01/2023] [Revised: 10/06/2023] [Accepted: 10/11/2023] [Indexed: 11/08/2023] Open
Abstract
Animal growth is closely related to glycolipid metabolism, and the liver is the main organ for glycogen storage and fat synthesis in birds, but whether monochromatic light affects glycogen and lipid synthesis in the liver is unclear. Therefore, in this study, a total of 96 Arbor Acre (AA) broilers at posthatching d 0 (P0) were raised under 4 kinds of light-emitting diode (LED) lights, white light (WL), red light (RL), green light (GL), and blue light (BL), to posthatching d 21 (P21) and 35 (P35). The results showed that the liver, abdominal fat, and abdominal fat indices gradually increased with increasing age under monochromatic light treatments. The liver glycogen and triglyceride (TG) contents also showed an increasing trend. Furthermore, compared with those at P21, the mRNA levels of glycogen synthase (GS), glycogen synthase kinase-3β (GSK-3β), and protein kinase B (AKT1) in the liver were increased in the WL and RL groups at P35, and the mRNA levels of acetyl-CoA carboxylase (ACC) and apolipoprotein B (APOB) increased in all groups at P35. At the same time, the total antioxidant capacity (T-AOC) and liver superoxide dismutase (SOD) contents increased in all groups at P35 compared with those at P21. In addition, at P21, compared with WL, GL and BL promoted the serum glucose (GLU) and TG contents by increasing the mRNA levels of GS, GSK-3β, glucose-6-phosphatase (G6PC), ACC, and fatty acid synthase (FAS), but no effect on the proliferative ability and damage of hepatocytes. At P35, RL promoted the hepatic glycogen and TG contents by increasing GSK-3β, AKT1, ACC, and APOB mRNA levels, and the serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels were increased than in the WL group. These results suggest that the effects of light color on liver glycogen and lipid synthesis in broilers changed with age, and also provide a theoretical guidance for scientific use of color of light information to improve productive performance in broilers.
Collapse
Affiliation(s)
- Lu Wang
- Laboratory of Anatomy of Domestic Animal, National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Zixu Wang
- Laboratory of Anatomy of Domestic Animal, National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Yaoxing Chen
- Laboratory of Anatomy of Domestic Animal, National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Jing Cao
- Laboratory of Anatomy of Domestic Animal, National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
42
|
Grzelak N, Kaczmarek D, Mrówczyński W. Comparison of the effects of BDNF/TRKB signalling on metabolic biomarkers in the liver of sedentary and trained rats with normal and knockout BDNF genotypes. Front Physiol 2023; 14:1268648. [PMID: 38152248 PMCID: PMC10751318 DOI: 10.3389/fphys.2023.1268648] [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: 07/28/2023] [Accepted: 12/04/2023] [Indexed: 12/29/2023] Open
Abstract
Introduction: The effect of brain-derived neurotrophic factor (BDNF) on the modulation of metabolic processes in the liver is poorly understood. Therefore, the aim of this study was to investigate whether hepatic concentrations or activities of metabolic biomarkers depend on altered BDNF/TrkB content in the liver, resulting from different BDNF genotypes of rats. In addition, it was assessed whether 5-week moderate endurance training modifies the levels of BDNF/Trk-B signaling and studied hepatic markers. Methods: Experiments were performed on wild-type and heterozygous BDNF knockout (HET, SD-Bdnf) rats, which were divided into four groups: control with normal genotype (Bdnf+/+), control with BDNF knockout genotype (Bdnf+/-), trained with normal genotype (Bdnf+/+T) and trained with BDNF knockout genotype (Bdnf +/-T). BDNF/TrkB concentrations as well as selected metabolic biomarkers including lipids-total cholesterol (CHOL), low-density lipoprotein (LDL), triglycerides (TG); enzymes-alanine aminotransferase (ALAT), aspartate aminotransferase (ASAT), gamma-glutamyl transferase (GGT), lactate dehydrogenase (LDH), alkaline phosphatase (ALP); hormones-insulin (INS) and leptin (LEPT) as well as interleukin-6 (IL-6) as regeneration indicator were measured directly in liver homogenates. Results and Discussion: The study showed that Bdnf+/- rats exhibited reduced BDNF/TrkB signaling (BDNF, p < 0.0001; Trk-B, p = 0.0005), altered lipid levels (CHOL, p < 0.0001; LDL, p < 0.0001; TG, p = 0.0006) and reduced hepatic ALAT (p = 0.0004) and GGT (p < 0.0001) activity, which may contribute to hepatic steatosis and obesity, as well as indicate impairment of specific metabolic pathways in the liver. Interestingly, endurance training did not alter hepatic BDNF and TrkB content, but improved ALAT (p = 0.0366) and ASAT (p = 0.0191) activities and increased hepatic IL-6 (p = 0.0422) levels in Bdnf +/- rats, suggesting enhanced liver regeneration in animals with BDNF allele loss.
Collapse
Affiliation(s)
- Norbert Grzelak
- Department of Neurobiology, Poznań University of Physical Education, Poznań, Poland
| | - Dominik Kaczmarek
- Department of Physiology and Biochemistry, Poznań University of Physical Education, Poznań, Poland
| | | |
Collapse
|
43
|
Wiedmer SK, Riekkola ML. Field-flow fractionation - an excellent tool for fractionation, isolation and/or purification of biomacromolecules. J Chromatogr A 2023; 1712:464492. [PMID: 37944435 DOI: 10.1016/j.chroma.2023.464492] [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: 10/17/2023] [Revised: 10/30/2023] [Accepted: 11/03/2023] [Indexed: 11/12/2023]
Abstract
Field-flow fractionation (FFF) with its several variants, has developed into a mature methodology. The scope of the FFF investigations has expanded, covering both a wide range of basic studies and especially a wide range of analytical applications. Special attention of this review is given to the achievements of FFF with reference to recent applications in the fractionation, isolation, and purification of biomacromolecules, and from which especially those of (in alphabetical order) bacteria, cells, extracellular vesicles, liposomes, lipoproteins, nucleic acids, and viruses and virus-like particles. In evaluating the major approaches and trends demonstrated since 2012, the most significant biomacromolecule applications are compiled in tables. It is also evident that asymmetrical flow field-flow fractionation is by far the most dominant technique in the studies. The industry has also shown current interest in FFF and adopted it in some sophisticated fields. FFF, in combination with appropriate detectors, handles biomacromolecules in open channel in a gentle way due to the lack of shear forces and unwanted interactions caused by the stationary phase present in chromatography. In addition, in isolation and purification of biomacromolecules quite high yields can be achieved under optimal conditions.
Collapse
Affiliation(s)
- Susanne K Wiedmer
- Department of Chemistry, POB 55, 00014 University of Helsinki, Finland
| | | |
Collapse
|
44
|
Ardiana M, Fadila AN, Zuhra Z, Kusuma NM, Surya Erlangga Rurus ME, Oceandy D. Non-coding RNA therapeutics in cardiovascular diseases and risk factors: Systematic review. Noncoding RNA Res 2023; 8:487-506. [PMID: 37483458 PMCID: PMC10362275 DOI: 10.1016/j.ncrna.2023.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 06/08/2023] [Accepted: 06/19/2023] [Indexed: 07/25/2023] Open
Abstract
At present, RNA-based therapy which includes therapies using non-coding RNAs (ncRNAs), antisense oligonucleotides (ASOs), and aptamers are gaining widespread attention as possible ways to target genes in various cardiovascular diseases (CVDs), thereby serving as a promising therapeutic approach for CVDs and risk factors management. However, data are primarily in an early stage. A systematic review was carried out using literature from several databases (Pubmed, Cochrane, Scopus, and DOAJR) following the PRISMA guidelines. Of the 64 articles reviewed, 39 papers were included in this review with three main types of RNAs: aptamers, antisense oligonucleotides (ASOs), and small-interfering RNA (siRNA). All studies were human clinical trials. RNA-based therapies were demonstrated to be efficacious in treating various CVDs and controlling cardiovascular risk factors. They are generally safe and well-tolerated. However, data are still in the early stage and warrant further investigation.
Collapse
Affiliation(s)
- Meity Ardiana
- Department of Cardiology and Vascular Medicine, Dr.Soetomo General Hospital, Surabaya, Indonesia
- Faculty of Medicine, Airlangga University, Surabaya, Indonesia
| | - Asiyah Nurul Fadila
- Department of Cardiology and Vascular Medicine, Dr.Soetomo General Hospital, Surabaya, Indonesia
- Faculty of Medicine, Airlangga University, Surabaya, Indonesia
| | - Zakirah Zuhra
- Department of Cardiology and Vascular Medicine, Dr.Soetomo General Hospital, Surabaya, Indonesia
- Faculty of Medicine, Airlangga University, Surabaya, Indonesia
| | | | | | - Delvac Oceandy
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
45
|
Yang Y, Huangfu L, Li H, Yang D. Research progress of hyperthermia in tumor therapy by influencing metabolic reprogramming of tumor cells. Int J Hyperthermia 2023; 40:2270654. [PMID: 37871910 DOI: 10.1080/02656736.2023.2270654] [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: 04/25/2023] [Accepted: 10/09/2023] [Indexed: 10/25/2023] Open
Abstract
Cellular metabolic reprogramming is an important feature of malignant tumors. Metabolic reprogramming causes changes in the levels or types of specific metabolites inside and outside the cell, which affects tumorigenesis and progression by influencing gene expression, the cellular state, and the tumor microenvironment. During tumorigenesis, a series of changes in the glucose metabolism, fatty acid metabolism, amino acid metabolism, and cholesterol metabolism of tumor cells occur, which are involved in the process of cellular carcinogenesis and constitute part of the underlying mechanisms of tumor formation. Hyperthermia, as one of the main therapeutic tools for malignant tumors, has obvious effects on tumor cell metabolism. In this paper, we will combine the latest research progress in the field of cellular metabolic reprogramming and focus on the current experimental research and clinical treatment of hyperthermia in cellular metabolic reprogramming to discuss the feasibility of cellular metabolic reprogramming-related mechanisms guiding hyperthermia in malignant tumor treatment, so as to provide more ideas for hyperthermia to treat malignant tumors through the direction of cellular metabolic reprogramming.
Collapse
Affiliation(s)
- Yuchuan Yang
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| | - Linkuan Huangfu
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| | - Huizhen Li
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| | - Daoke Yang
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| |
Collapse
|
46
|
Vignoli A, Miolo G, Tenori L, Buonadonna A, Lombardi D, Steffan A, Scalone S, Luchinat C, Corona G. Novel metabolomics-biohumoral biomarkers model for predicting survival of metastatic soft-tissue sarcomas. iScience 2023; 26:107678. [PMID: 37752948 PMCID: PMC10518687 DOI: 10.1016/j.isci.2023.107678] [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: 03/22/2023] [Revised: 06/23/2023] [Accepted: 08/14/2023] [Indexed: 09/28/2023] Open
Abstract
Soft tissue sarcomas (STSs) are rare malignant tumors that are difficult to prognosticate using currently available instruments. Omics sciences could provide more accurate and individualized survival predictions for patients with metastatic STS. In this pilot, hypothesis-generating study, we integrated clinicopathological variables with proton nuclear magnetic resonance (1H NMR) plasma metabolomic and lipoproteomic profiles, capturing both tumor and host characteristics, to identify novel prognostic biomarkers of 2-year survival. Forty-five metastatic STS (mSTS) patients with prevalent leiomyosarcoma and liposarcoma histotypes receiving trabectedin treatment were enrolled. A score combining acetate, triglycerides low-density lipoprotein (LDL)-2, and red blood cell count was developed, and it predicts 2-year survival with optimal results in the present cohort (84.4% sensitivity, 84.6% specificity). This score is statistically significant and independent of other prognostic factors such as age, sex, tumor grading, tumor histotype, frailty status, and therapy administered. A nomogram based on these 3 biomarkers has been developed to inform the clinical use of the present findings.
Collapse
Affiliation(s)
- Alessia Vignoli
- Magnetic Resonance Center (CERM) and Department of Chemistry “Ugo Schiff”, University of Florence, 50019 Sesto Fiorentino, Italy
| | - Gianmaria Miolo
- Medical Oncology and Cancer Prevention Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Leonardo Tenori
- Magnetic Resonance Center (CERM) and Department of Chemistry “Ugo Schiff”, University of Florence, 50019 Sesto Fiorentino, Italy
- Consorzio Interuniversitario Risonanze Magnetiche MetalloProteine (CIRMMP), 50019 Sesto Fiorentino, Italy
| | - Angela Buonadonna
- Medical Oncology and Cancer Prevention Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Davide Lombardi
- Medical Oncology and Cancer Prevention Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Agostino Steffan
- Immunopathology and Cancer Biomarkers Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Simona Scalone
- Medical Oncology and Cancer Prevention Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Claudio Luchinat
- Consorzio Interuniversitario Risonanze Magnetiche MetalloProteine (CIRMMP), 50019 Sesto Fiorentino, Italy
- GiottoBiotech s.r.l, Sesto Fiorentino, Italy
| | - Giuseppe Corona
- Immunopathology and Cancer Biomarkers Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| |
Collapse
|
47
|
Huang HC, Lee PN, Huang WC, Yang HY. Partial Replacement of Diet with Dehulled Adlay Ameliorates Hepatic Steatosis, Inflammation, Oxidative Stress, and Gut Dysbiosis in Rats with Nonalcoholic Fatty Liver Disease. Nutrients 2023; 15:4375. [PMID: 37892450 PMCID: PMC10610228 DOI: 10.3390/nu15204375] [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: 09/02/2023] [Revised: 09/27/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
The prevalence of nonalcoholic fatty liver disease (NAFLD) has been increasing worldwide, and the average age at NAFLD diagnosis has been decreasing. Although some components of adlay can ameliorate lipid metabolism, oxidative stress, inflammatory response, and gut microbiota, few studies have explored the effects of the dietary intake of intact dehulled adlay on liver diseases. Therefore, in this study, we investigated the effects of the dietary intake of dehulled adlay on NAFLD progression and explored the potential underlying mechanisms. Rats were randomized into a control group; a high-fat, high-sucrose diet (60% total energy derived from fat and 9.4% from sucrose)-induced NAFLD group (N); or a high-fat, high-sucrose diet with dehulled adlay group (received the same amounts of dietary fiber and total energy as did the N group). The experimental duration was 16 weeks. The diet containing dehulled adlay mitigated hepatic fat accumulation, proinflammatory cytokine levels, and oxidative stress by regulating the AMPK-Nrf2-NLRP3 inflammasome pathway and ferroptosis. Additionally, the dietary intake of dehulled adlay modulated the composition of the gut microbiota. In conclusion, a diet containing dehulled adlay may decelerate the progression of NAFLD by ameliorating hepatic steatosis, inflammation, oxidative stress, and gut dysbiosis.
Collapse
Affiliation(s)
- Hsuan-Chih Huang
- Department of Nutritional Science, Fu Jen Catholic University, No. 510, Zhongzheng Rd., Xinzhuang District, New Taipei City 24205, Taiwan
| | - Pei-Ni Lee
- Department of Nutrition, Taipei Hospital, Ministry of Health and Welfare, No. 127, Siyuan Rd., Xinzhuang District, New Taipei City 24250, Taiwan;
| | - Wen-Chih Huang
- Department of Anatomical Pathology, Taipei Institute of Pathology, No. 146, Sec. 3, Chongqing N. Rd., Datong District, Taipei City 10374, Taiwan
| | - Hsin-Yi Yang
- Department of Nutritional Science, Fu Jen Catholic University, No. 510, Zhongzheng Rd., Xinzhuang District, New Taipei City 24205, Taiwan
| |
Collapse
|
48
|
Wang Y, Liu T, Cai Y, Liu W, Guo J. SIRT6's function in controlling the metabolism of lipids and glucose in diabetic nephropathy. Front Endocrinol (Lausanne) 2023; 14:1244705. [PMID: 37876546 PMCID: PMC10591331 DOI: 10.3389/fendo.2023.1244705] [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: 06/23/2023] [Accepted: 09/21/2023] [Indexed: 10/26/2023] Open
Abstract
Diabetic nephropathy (DN) is a complication of diabetes mellitus (DM) and the main cause of excess mortality in patients with type 2 DM. The pathogenesis and progression of DN are closely associated with disorders of glucose and lipid metabolism. As a member of the sirtuin family, SIRT6 has deacetylation, defatty-acylation, and adenosine diphosphate-ribosylation enzyme activities as well as anti-aging and anticancer activities. SIRT6 plays an important role in glucose and lipid metabolism and signaling, especially in DN. SIRT6 improves glucose and lipid metabolism by controlling glycolysis and gluconeogenesis, affecting insulin secretion and transmission and regulating lipid decomposition, transport, and synthesis. Targeting SIRT6 may provide a new therapeutic strategy for DN by improving glucose and lipid metabolism. This review elaborates on the important role of SIRT6 in glucose and lipid metabolism, discusses the potential of SIRT6 as a therapeutic target to improve glucose and lipid metabolism and alleviate DN occurrence and progression of DN, and describes the prospects for future research.
Collapse
Affiliation(s)
- Ying Wang
- Country Renal Research Institution of Beijing University of Chinese Medicine, Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Tongtong Liu
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuzi Cai
- Country Renal Research Institution of Beijing University of Chinese Medicine, Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Weijing Liu
- Country Renal Research Institution of Beijing University of Chinese Medicine, Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Jing Guo
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
49
|
Conlon DM, Welty FK, Reyes-Soffer G, Amengual J. Sex-Specific Differences in Lipoprotein Production and Clearance. Arterioscler Thromb Vasc Biol 2023; 43:1617-1625. [PMID: 37409532 PMCID: PMC10527393 DOI: 10.1161/atvbaha.122.318247] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 06/19/2023] [Indexed: 07/07/2023]
Abstract
Therapeutic approaches to reduce atherogenic lipid and lipoprotein levels remain the most effective and assessable strategies to prevent and treat cardiovascular disease. The discovery of novel research targets linked to pathways associated with cardiovascular disease development has enhanced our ability to decrease disease burden; however, residual cardiovascular disease risks remain. Advancements in genetics and personalized medicine are essential to understand some of the factors driving residual risk. Biological sex is among the most relevant factors affecting plasma lipid and lipoprotein profiles, playing a pivotal role in the development of cardiovascular disease. This minireview summarizes the most recent preclinical and clinical studies covering the effect of sex on plasma lipid and lipoprotein levels. We highlight the recent advances in the mechanisms regulating hepatic lipoprotein production and clearance as potential drivers of disease presentation. We focus on using sex as a biological variable in studying circulating lipid and lipoprotein levels.
Collapse
Affiliation(s)
| | | | - Gissette Reyes-Soffer
- Department of Medicine, Division of Preventive Medicine and Nutrition, Columbia University College of Physicians and Surgeons
| | - Jaume Amengual
- Department of Food Science and Human Nutrition and Division of Nutritional Sciences. University of Illinois Urbana Champaign
| |
Collapse
|
50
|
Bao C, Wu T, Zhu S, Wang X, Zhang Y, Wang X, Yang L, He C. Regulation of cholesterol homeostasis in osteoporosis mechanisms and therapeutics. Clin Sci (Lond) 2023; 137:1131-1143. [PMID: 37553962 DOI: 10.1042/cs20220752] [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: 11/04/2022] [Revised: 06/27/2023] [Accepted: 07/03/2023] [Indexed: 08/10/2023]
Abstract
Osteoporosis is a metabolic bone disease that affects hundreds of millions of people worldwide and is characterized by excessive loss of bone protein and mineral content. The incidence and mortality of osteoporosis increase with age, creating a significant medical and economic burden globally. The importance of cholesterol levels has been reported in the development of diseases including osteoporosis. It is important to note that key enzymes and molecules involved in cholesterol homeostasis are closely related to bone formation. Excessive cholesterol may cause osteoporosis, cholesterol and its metabolites affect bone homeostasis by regulating the proliferation and stimulation of osteoblasts and osteoclasts. Therefore, antagonism of elevated cholesterol levels may be a potential strategy to prevent osteoporosis. There is sufficient evidence to support the use of bisphosphonates and statin drugs for osteoporosis in the clinic. Therefore, in view of the aggravation of the aging problem, we summarize the intracellular mechanism of cholesterol homeostasis and its relationship with osteoporosis (including cholesterol and cholesterol oxidation products (COPs) in osteoporosis). Furthermore, the current clinical cholesterol-lowering drugs for osteoporosis were also summarized, as are new and promising therapies (cell-based therapies (e.g., stem cells) and biomaterial-delivered target drug therapies for osteoporosis as well).
Collapse
Affiliation(s)
- Chuncha Bao
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, People's Republic of China
- Key Laboratory of Rehabilitation Medicine, Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Tao Wu
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, People's Republic of China
- Key Laboratory of Rehabilitation Medicine, Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Siyi Zhu
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, People's Republic of China
- Key Laboratory of Rehabilitation Medicine, Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Xiaoyi Wang
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, People's Republic of China
- Key Laboratory of Rehabilitation Medicine, Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Yujia Zhang
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, People's Republic of China
- Key Laboratory of Rehabilitation Medicine, Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Xiangxiu Wang
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, People's Republic of China
- Key Laboratory of Rehabilitation Medicine, Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Lin Yang
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, People's Republic of China
- Key Laboratory of Rehabilitation Medicine, Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Chengqi He
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, People's Republic of China
- Key Laboratory of Rehabilitation Medicine, Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| |
Collapse
|