Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (4,434)

Search Parameters:
Keywords = regulation of lipid metabolism

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
40 pages, 4949 KB  
Review
Is Obesity a Modifiable Risk Factor in Multiple Sclerosis? Mechanistic Insights into Neuroinflammation and Oxidative Damage
by Fani-Niki Varra, Olga Pagonopoulou, Michail Varras, Viktoria-Konstantina Varra and Panagiotis Theodosis-Nobelos
Pathophysiology 2026, 33(1), 5; https://doi.org/10.3390/pathophysiology33010005 - 13 Jan 2026
Abstract
Introduction: Multiple sclerosis (MS) is a chronic autoimmune inflammatory disorder of the central nervous system (CNS) that leads to demyelination of CNS neurons and is influenced by genetic, environmental, and lifestyle factors, including diet and obesity. Methods: This review aims to analyze at [...] Read more.
Introduction: Multiple sclerosis (MS) is a chronic autoimmune inflammatory disorder of the central nervous system (CNS) that leads to demyelination of CNS neurons and is influenced by genetic, environmental, and lifestyle factors, including diet and obesity. Methods: This review aims to analyze at the molecular level the relationship between obesity, as a chronic inflammatory condition, and the pathophysiology of MS, as a chronic autoimmune inflammatory disease, in order to understand the complex links between obesity and MS through a search of the PubMed and Google Scholar databases. Discussion: Chronic inflammation and OS are interconnected processes, causing a toxic state, which contributes to the development of CNS neuroinflammation and neuronal damage, resulting in neuronal demyelination and the onset of MS. Adipose tissue is a complex endocrine organ; in addition to being a lipid storage organ, it secretes cytokines and adipokines, which are involved in the regulation of hormones, metabolism, inflammation, and whole-body homeostasis. Obesity triggers chronic low-grade inflammation, disruption of the blood–brain barrier (BBB) and brain metabolism, infiltration of the CNS by immune cells, production of ROS, and generation of oxidative stress (OS). Anti-inflammatory and pro-inflammatory adipokines are also implicated in MS and obesity. Conclusions: Obesity affects MS through common underlying mechanisms and seems to be a modifiable risk factor. Antioxidant and anti-inflammatory compounds with multi-functional characteristics could be additional tools to slow the progression of MS and its promotion through obesity while also offering potential treatment options for both conditions via their multi-targeting characteristics. Full article
32 pages, 1950 KB  
Article
Association of Circulating Irisin with Insulin Resistance and Metabolic Risk Markers in Prediabetic and Newly Diagnosed Type 2 Diabetes Patients
by Daniela Denisa Mitroi Sakizlian, Lidia Boldeanu, Diana Clenciu, Adina Mitrea, Ionela Mihaela Vladu, Alina Elena Ciobanu Plasiciuc, Mohamed-Zakaria Assani and Daniela Ciobanu
Int. J. Mol. Sci. 2026, 27(2), 787; https://doi.org/10.3390/ijms27020787 - 13 Jan 2026
Abstract
Circulating irisin, a myokine implicated in energy expenditure and adipose tissue regulation, has been increasingly studied as a potential biomarker of metabolic dysfunction. This study evaluated the relationship between serum irisin and metabolic indices, including the atherogenic index of plasma (AIP), the lipid [...] Read more.
Circulating irisin, a myokine implicated in energy expenditure and adipose tissue regulation, has been increasingly studied as a potential biomarker of metabolic dysfunction. This study evaluated the relationship between serum irisin and metabolic indices, including the atherogenic index of plasma (AIP), the lipid accumulation product (LAP), and hypertriglyceridemic-waist (HTGW) phenotype in individuals with prediabetes (PreDM) and newly diagnosed type 2 diabetes mellitus (T2DM). A total of 138 participants (48 PreDM, 90 T2DM) were assessed for anthropometric, glycemic, and lipid parameters. Serum irisin levels were measured by enzyme-linked immunosorbent assay (ELISA) and correlated with insulin resistance indices (Homeostatic Model Assessment of Insulin Resistance (HOMA-IR), Quantitative Insulin Sensitivity Check Index (QUICKI)), glycemic control (glycosylated hemoglobin A1c (HbA1c)), and composite lipid markers (total triglycerides-to-high-density lipoprotein cholesterol (TG/HDL-C)). Group differences were evaluated using non-parametric tests; two-way ANOVA assessed interactions between phenotypes and markers; multiple linear regression (MLR) and logistic regression models explored independent associations with metabolic indices and HTGW; receiver operating characteristic (ROC) analyses compared global and stratified model performance. Serum irisin was significantly lower in T2DM than in PreDM (median 140.4 vs. 230.7 ng/mL, p < 0.0001). Irisin levels remained comparable between males and females in both groups. Post hoc analysis shows that lipid indices and irisin primarily distinguish HTGW phenotypes, especially in T2DM. In both groups, irisin correlated inversely with HOMA-IR, AIP, and TG/HDL-C, and positively with QUICKI, indicating a possible compensatory role in early insulin resistance. MLR analyses revealed no independent relationship between irisin and either AIP or LAP in PreDM, while in T2DM, waist circumference remained the strongest negative predictor of irisin. Logistic regression identified age, male sex, and HbA1c as independent predictors of the HTGW phenotype, while irisin contributed modestly to overall model discrimination. ROC curves demonstrated good discriminative performance (AUC = 0.806 for global; 0.794 for PreDM; 0.813 for T2DM), suggesting comparable predictive accuracy across glycemic stages. In conclusion, irisin levels decline from prediabetes to overt diabetes and are inversely linked to lipid accumulation and insulin resistance but do not independently predict the HTGW phenotype. These findings support irisin’s role as an integrative indicator of metabolic stress rather than a stand-alone biomarker. Incorporating irisin into multi-parameter metabolic panels may enhance early detection of cardiometabolic risk in dysglycemic populations. Full article
(This article belongs to the Special Issue Molecular Diagnosis and Treatments of Diabetes Mellitus: 2nd Edition)
Show Figures

Figure 1

15 pages, 772 KB  
Article
High Ratio of Dietary Palmitic Acid to DHA + EPA Induces Glucose Metabolic Disorder Through Endocrine and Transcriptional Regulation in Large Yellow Croaker (Larimichthys crocea)
by Qi Wang, Huaicheng Ge, Zhixiang Gu, Hao Chen, Hua Mu, Kangsen Mai and Wenbing Zhang
Metabolites 2026, 16(1), 72; https://doi.org/10.3390/metabo16010072 - 13 Jan 2026
Abstract
Background/Objectives: Replacing fish oil with vegetable oil is an important measure for aquaculture to relieve the pressure of fish oil, but it is also easy to cause the growth decline and metabolic disorder of farmed animals, mainly due to the change in [...] Read more.
Background/Objectives: Replacing fish oil with vegetable oil is an important measure for aquaculture to relieve the pressure of fish oil, but it is also easy to cause the growth decline and metabolic disorder of farmed animals, mainly due to the change in dietary fatty acids. This study investigated the regulatory effects of dietary fatty acid composition on glucose metabolism in large yellow croaker (Larimichthys crocea) with an initial weight of 30.51 ± 0.16 g. Methods: Three isonitrogenous (~43% crude protein) and isolipid (~11% crude lipid) diets were formulated as follows: control (CON, DHA/EPA-rich oil as primary lipid), moderate palmitic acid (MPA, 50% of DHA+EPA-rich oil was replaced by glyceryl palmitate), and high palmitic acid (HPA, 100% of DHA+EPA-rich oil was replaced by glyceryl palmitate). Results: After 10 weeks of feeding, the HPA significantly reduced the liver/muscle glycogen contents, increased the liver lipid content, decreased the serum leptin/insulin level, and increased the adiponectin level. The levels of DHA and EPA in liver were decreased significantly. Transcriptionally, HPA upregulated hepatic glucokinase (gk, glycolysis) but down-regulated glycogen synthase (gys) and insulin/irs2 (insulin pathway) while inhibiting muscle ampk and leptin receptor (lepr). Conclusions: This study showed that high dietary PA/(DHA + EPA) impacted glycolipid homeostasis through endocrine and transcriptional regulation, leading to increased crude lipid and decreased glycogen levels, which provides a theoretical basis for scientific aquatic feed fatty acid formulation. Full article
(This article belongs to the Special Issue Nutrition, Metabolism and Physiology in Aquatic Animals)
Show Figures

Figure 1

17 pages, 3779 KB  
Article
Cycloastragenol Improves Fatty Acid Metabolism Through NHR-49/FAT-7 Suppression and Potent AAK-2 Activation in Caenorhabditis elegans Obesity Model
by Liliya V. Mihaylova, Martina S. Savova, Monika N. Todorova, Valeria Tonova, Biser K. Binev and Milen I. Georgiev
Int. J. Mol. Sci. 2026, 27(2), 772; https://doi.org/10.3390/ijms27020772 - 13 Jan 2026
Abstract
Obesity is among the top contributing factors for non-communicable chronic disease development and has attained menacing global proportions, affecting approximately one of eight adults. Phytochemicals that support energy metabolism and prevent obesity development have been the subject of intense research endeavors over the [...] Read more.
Obesity is among the top contributing factors for non-communicable chronic disease development and has attained menacing global proportions, affecting approximately one of eight adults. Phytochemicals that support energy metabolism and prevent obesity development have been the subject of intense research endeavors over the past several decades. Cycloastragenol is a natural triterpenoid compound and aglycon of astragaloside IV, known for activating telomerase and mitigating cellular aging. Here, we aim to characterize the effect of cycloastragenol on lipid metabolism in a glucose-induced obesity model in Caenorhabditis elegans. We assessed the changes in the body length, width, and area in C. elegans maintained under elevated glucose through automated WormLab system. Lipid accumulation in the presence of either cycloastragenol (100 μM) or orlistat (12 μM), used as a positive anti-obesity control drug, was quantified through Nile Red fluorescent staining. Furthermore, we evaluated the changes in key energy metabolism molecular players in GFP-reporter transgenic strains. Our results revealed that cycloastragenol treatment decreased mean body area and reduced lipid accumulation in the C. elegans glucose-induced model. The mechanistic data indicated that cycloastragenol suppresses the nuclear hormone receptor family member NHR-49 and the delta(9)-fatty-acid desaturase 7 (FAT-7) enzyme, and activates the 5′-AMP-activated protein kinase catalytic subunit alpha-2 (AAK-2) and the protein skinhead 1 (SKN-1) signaling. Collectively, our findings highlight that cycloastragenol reprograms lipid metabolism by down-regulating the insulin-like receptor (daf-2)/phosphatidylinositol 3-kinase (age-1)/NHR-49 signaling while simultaneously enhancing the activity of the AAK-2/NAD-dependent protein deacetylase (SIR-2.1) pathway. The anti-obesogenic potential of cycloastragenol rationalizes further validation in the context of metabolic diseases and obesity management. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Obesity and Metabolic Diseases)
Show Figures

Figure 1

19 pages, 1167 KB  
Article
Metabolic and Orexin-A Responses to Ketogenic Diet and Intermittent Fasting: A 12-Month Randomized Trial in Adults with Obesity
by Antonietta Monda, Maria Casillo, Salvatore Allocca, Fiorenzo Moscatelli, Marco La Marra, Vincenzo Monda, Girolamo Di Maio, Paride Vasco, Marcellino Monda, Rita Polito, Giovanni Messina and Antonietta Messina
Nutrients 2026, 18(2), 238; https://doi.org/10.3390/nu18020238 - 12 Jan 2026
Abstract
Background/Objectives: Intermittent fasting and ketogenic dietary approaches are increasingly investigated for their potential metabolic benefits in obesity. However, their long-term neuroendocrine effects—particularly those involving Orexin-A, a peptide implicated in energy regulation—remain poorly understood. The objective of this study was to compare the long-term [...] Read more.
Background/Objectives: Intermittent fasting and ketogenic dietary approaches are increasingly investigated for their potential metabolic benefits in obesity. However, their long-term neuroendocrine effects—particularly those involving Orexin-A, a peptide implicated in energy regulation—remain poorly understood. The objective of this study was to compare the long-term metabolic, inflammatory, and orexinergic responses to different dietary strategies in adults with obesity. Methods: In this 12-month randomized, three-arm trial, 30 adults with obesity (BMI ≥ 30 kg/m2) were randomly assigned (1:1:1) to a hypocaloric ketogenic diet (KD), a 16:8 time-restricted eating regimen (TRF16:8), or a 5:2 intermittent fasting protocol (ADF5:2). Anthropometric parameters, body composition, fasting glucose, lipid profile, inflammatory cytokines (CRP, IL-6, TNF-α, IL-10), and plasma Orexin-A levels were assessed at baseline and every 3 months. Dietary adherence was monitored through structured logs and monthly assessments. Statistical analyses included repeated-measures models with sensitivity analyses adjusted for age and sex. Results: All participants completed the intervention. The ketogenic diet produced the largest sustained reductions in BMI, fat mass, fasting glucose, and total cholesterol over 12 months. TRF16:8 elicited more rapid early metabolic improvements and showed the most consistent longitudinal increase in Orexin-A levels. The ADF5:2 protocol resulted in moderate improvements across outcomes. In all groups, increases in Orexin-A were associated with markers of improved metabolic flexibility and reduced inflammation; however, mediation analyses were exploratory and non-causal. Between-group differences remained significant for fat mass, glucose, and Orexin-A trajectories after correction for multiple comparisons. Conclusions: The ketogenic diet was associated with the most pronounced long-term metabolic improvements, whereas 16:8 time-restricted eating yielded faster early responses and the most stable enhancement in Orexin-A levels. These findings indicate distinct metabolic and neuroendocrine adaptation profiles across dietary strategies. Given the small sample size, results should be interpreted cautiously, and larger trials are warranted to clarify the role of Orexin-A as a potential biomarker of dietary response in obesity. Full article
(This article belongs to the Special Issue The Ketogenic Diet: Effects on Human Metabolism and Brain Health)
Show Figures

Figure 1

15 pages, 11950 KB  
Article
Integrated Multi-Omics Analysis Reveals Dysregulated Lipid Metabolism as a Novel Mechanism in Androgenetic Alopecia
by Xiao-Shuang Yang, Liyang Duan, Yu-Jie Miao, Zhongfa Lu and Ru Dai
Biomedicines 2026, 14(1), 160; https://doi.org/10.3390/biomedicines14010160 - 12 Jan 2026
Abstract
Background and Aims: Androgenetic alopecia (AGA) represents the most prevalent multifactorial condition leading to hair loss, necessitating an enhanced molecular understanding. The aim of this study is to present the analysis integrating protein, mRNA and miRNA between frontal and occipital regions of patients [...] Read more.
Background and Aims: Androgenetic alopecia (AGA) represents the most prevalent multifactorial condition leading to hair loss, necessitating an enhanced molecular understanding. The aim of this study is to present the analysis integrating protein, mRNA and miRNA between frontal and occipital regions of patients with androgenetic alopecia (AGA) and to identify potential mechanism. Methods and Results: Paired frontal and occipital scalps from four male donors with AGA were collected for transcriptomic and proteomics analyses. The molecular and protein characteristics of AGA were demonstrated by a comprehensive bioinformatics approach. Additionally, immunofluorescence (IF) and dual-luciferase reporter (DLR) assays were employed to confirm the analytical findings. A total of 758 differentially expressed proteins (DEPs), 1802 differentially expressed mRNAs (DERs) and 61 differentially expressed miRNAs (DEmiRNAs) were identified. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses revealed significant enrichments in lipid metabolism, especially those involving PPAR signaling. Co-expression analyses further supported the association of up-regulated genes with lipid metabolism. A protein–protein interaction network analysis, supplemented by KEGG enrichment and the MCE algorithm, pinpointed four candidate genes: DBI, ACAA1, IDH1 and PEX3. IF confirmed significant upregulation of ACAA1 and PEX3 in scalp tissues with AGA, while IDH1 was downregulated and DBI without significant changes. A competing endogenous RNA network indicated that hsa-miR-1343-3p targets ACAA1 and hsa-miR-3609_R-2 targets IDH1, which were confirmed by DLR assays. Conclusions: This study provides preliminary evidence that hsa-miR-1343-3p-mediated regulation of ACAA1 contributes to AGA pathogenesis, suggesting a link between AGA and lipid metabolism. Full article
(This article belongs to the Section Endocrinology and Metabolism Research)
Show Figures

Figure 1

31 pages, 2128 KB  
Review
RNA-Binding Proteins in Adipose Biology: From Mechanistic Understanding to Therapeutic Opportunities
by Ghida Dairi, Maria Al Ibrahim, Saeed Al Mahri, Khalid Al-Regaiey, Shuja Shafi Malik and Sameer Mohammad
Int. J. Mol. Sci. 2026, 27(2), 756; https://doi.org/10.3390/ijms27020756 - 12 Jan 2026
Abstract
Obesity, defined by excessive body fat accumulation, is strongly associated with dysfunction of adipose tissue, a major regulator of whole-body energy balance and metabolic health. Dysfunctional adipose tissue is characterized by altered adipokine secretion, impaired insulin sensitivity, and chronic low-grade inflammation, all of [...] Read more.
Obesity, defined by excessive body fat accumulation, is strongly associated with dysfunction of adipose tissue, a major regulator of whole-body energy balance and metabolic health. Dysfunctional adipose tissue is characterized by altered adipokine secretion, impaired insulin sensitivity, and chronic low-grade inflammation, all of which contribute to obesity-related comorbidities such as type 2 diabetes, cardiovascular disease, and certain cancers. Understanding how obesity disrupts adipose tissue biology is essential for developing strategies to mitigate these metabolic risks. In recent years, RNA-binding proteins (RBPs) have emerged as important regulators of energy metabolism. By controlling post-transcriptional gene expression, RBPs influence RNA stability, localization, and translation, thereby shaping key cellular processes. Dysregulation of specific RBPs has been implicated in obesity and metabolic disorders, with several shown to affect adipogenesis, lipid handling, thermogenesis, and insulin sensitivity across different adipose depots. Their ability to direct the fate of transcripts involved in metabolic homeostasis positions RBPs as critical nodes linking adipose dysfunction to systemic disease. This review provides a mechanistic overview of RBP functions in adipose biology, highlights how their dysregulation can reinforce metabolic dysfunction, and identifies gaps and future directions for exploring RBPs and their RNA networks as potential therapeutic targets for obesity and related metabolic diseases. Full article
Show Figures

Graphical abstract

13 pages, 733 KB  
Review
G Protein-Coupled Receptors in Irritable Bowel Syndrome: Mechanisms and Therapeutic Opportunities
by Zhenya Zhu, Ziyu Liu, Yate He, Xiaorui He, Wei Zheng and Mizu Jiang
Int. J. Mol. Sci. 2026, 27(2), 752; https://doi.org/10.3390/ijms27020752 - 12 Jan 2026
Abstract
Irritable bowel syndrome (IBS) is a functional gastrointestinal disorder characterized by abdominal pain, altered motility, and visceral hypersensitivity. Emerging evidence implicates G protein-coupled receptors (GPCRs) as key integrators of microbial, immune, endocrine, and neural signals in IBS pathophysiology. This review summarizes recent advances [...] Read more.
Irritable bowel syndrome (IBS) is a functional gastrointestinal disorder characterized by abdominal pain, altered motility, and visceral hypersensitivity. Emerging evidence implicates G protein-coupled receptors (GPCRs) as key integrators of microbial, immune, endocrine, and neural signals in IBS pathophysiology. This review summarizes recent advances in understanding how GPCRs mediate gut immune regulation, microbiota–host crosstalk, metabolic signaling, and pain processing in IBS. Recent studies show that microbial metabolites (e.g., short-chain fatty acids, biogenic amines, and lipid mediators) signal through GPCRs on immune cells, epithelia, and neurons to influence intestinal homeostasis. On immune cells and neurons, GPCRs also mediate signals from external substances (such as fats, sugars, histamine, etc.) to regulate immune and neural functions. And there are challenges and future directions in targeting GPCRs for IBS, including patient heterogeneity and the complexity of host–microbiome interactions. This review provides a mechanistic framework for GPCR-based therapies in IBS. Full article
(This article belongs to the Special Issue Emerging Roles of the Gut-Brain Axis (GBA) in Health and Disease)
Show Figures

Figure 1

24 pages, 2708 KB  
Review
Berberine: A Negentropic Modulator for Multi-System Coordination
by Xiaolian Tian, Qingbo Chen, Yingying He, Yangyang Cheng, Mengyu Zhao, Yuanbin Li, Meng Yu, Jiandong Jiang and Lulu Wang
Int. J. Mol. Sci. 2026, 27(2), 747; https://doi.org/10.3390/ijms27020747 - 12 Jan 2026
Abstract
Berberine (BBR), a protoberberine alkaloid with a long history of medicinal use, has consistently demonstrated benefits in glucose–lipid metabolism and inflammatory balance across both preclinical and human studies. These diverse effects are not mediated by a single molecular target but by BBR’s capacity [...] Read more.
Berberine (BBR), a protoberberine alkaloid with a long history of medicinal use, has consistently demonstrated benefits in glucose–lipid metabolism and inflammatory balance across both preclinical and human studies. These diverse effects are not mediated by a single molecular target but by BBR’s capacity to restore network coordination among metabolic, immune, and microbial systems. At the core of this regulation is an AMP-activated Protein Kinase (AMPK)-centered mechanistic hub, integrating signals from insulin and nutrient sensing, Sirtuin 1/3 (SIRT1/3)-mediated mitochondrial adaptation, and inflammatory pathways such as nuclear Factor Kappa-light-chain-enhancer of Activated B cells (NF-κB) and NOD-, LRR- and Pyrin Domain-containing Protein 3 (NLRP3). This hub is dynamically regulated by system-level inputs from the gut, mitochondria, and epigenome, which in turn strengthen intestinal barrier function, reshape microbial and bile-acid metabolites, improve redox balance, and potentially reverse the epigenetic imprint of metabolic stress. These interactions propagate through multi-organ axes, linking the gut, liver, adipose, and vascular systems, thus aligning local metabolic adjustments with systemic homeostasis. Within this framework, BBR functions as a negentropic modulator, reducing metabolic entropy by fostering a coordinated balance among these interconnected systems, thereby restoring physiological order. Combination strategies, such as pairing BBR with metformin, Sodium-Glucose Cotransporter 2 (SGLT2) inhibitors, and agents targeting the microbiome or inflammation, have shown enhanced efficacy and substantial translational potential. Berberine ursodeoxycholate (HTD1801), an ionic-salt derivative of BBR currently in Phase III trials and directly compared with dapagliflozin, exemplifies the therapeutic promise of such approaches. Within the hub–axis paradigm, BBR emerges as a systems-level modulator that recouples energy, immune, and microbial circuits to drive multi-organ remodeling. Full article
(This article belongs to the Special Issue Role of Natural Compounds in Human Health and Disease)
Show Figures

Figure 1

19 pages, 931 KB  
Review
Plant-Forward Dietary Approaches to Reduce the Risk of Cardiometabolic Disease Among Hispanic/Latinx Adults Living in the United States: A Narrative Review
by Franze De La Calle, Joanna Bagienska and Jeannette M. Beasley
Nutrients 2026, 18(2), 220; https://doi.org/10.3390/nu18020220 - 10 Jan 2026
Viewed by 132
Abstract
Background: Cardiometabolic risk (CMR), including obesity, dyslipidemia, hypertension, and impaired glucose regulation, disproportionately affects Hispanic/Latinx adults in the United States (U.S.). Although plant-forward dietary patterns are established as cardioprotective, less is known about how dietary patterns within Hispanic/Latinx subgroups relate to CMR. [...] Read more.
Background: Cardiometabolic risk (CMR), including obesity, dyslipidemia, hypertension, and impaired glucose regulation, disproportionately affects Hispanic/Latinx adults in the United States (U.S.). Although plant-forward dietary patterns are established as cardioprotective, less is known about how dietary patterns within Hispanic/Latinx subgroups relate to CMR. Methods: A narrative review was conducted of observational studies among U.S. Hispanic/Latinx adults (≥18 years) examining defined dietary patterns (a priori, a posteriori, or hybrid) in relation to CMR outcomes (e.g., BMI, waist circumference, blood pressure, glucose, lipids). Risk of bias was assessed using an adapted version of the Newcastle–Ottawa Scale. Results: Ten studies met the inclusion criteria, including Seventh-day Adventist Latinx, Puerto Rican adults, Mexican American adults, Hispanic women, and a national Hispanic cohort. Plant-forward dietary patterns were associated with lower BMI and waist circumference, lower triglycerides and fasting glucose, and higher HDL-C. In contrast, energy-dense patterns characterized by refined grains, added sugars, processed meats, fried foods, solid fats, and sugar-sweetened beverages were associated with greater adiposity, poorer lipid profiles, and higher blood pressure. Traditional rice-and-beans–based patterns observed in Puerto Rican and Mexican American groups were associated with central adiposity and higher metabolic syndrome prevalence, despite modestly higher intakes of fruits, vegetables, and fiber. Study quality ranged from good (n = 4) to very good (n = 6). Conclusions: Across Hispanic/Latinx subgroups, plant-forward dietary patterns were associated with favorable cardiometabolic profiles, whereas refined and animal-based patterns aligned with higher CMR. Given the predominance of cross-sectional evidence, these findings should be interpreted as associative rather than causal. Culturally grounded dietary counseling, along with additional longitudinal and intervention studies, is needed to support cardiometabolic health in these populations. Full article
Show Figures

Figure 1

16 pages, 3064 KB  
Article
Curcumin Mitigates Fumonisin B1-Induced Ovarian Toxicity in Peak-Laying Ducks via Hormone Metabolic Protection and Enhanced Reproductive Resilience
by Lihua Wang, Rui Liang, Qingyun Cao, Zhiwei Hou, Ali Mujtaba Shah, Qiuyi Deng, Xue Li, Jinze Li, Jiaqing Chen, Lukuyu A. Bernard, Muhammad Kashif Saleemi, Lin Yang and Wence Wang
Toxins 2026, 18(1), 34; https://doi.org/10.3390/toxins18010034 - 9 Jan 2026
Viewed by 94
Abstract
The objective of this study was to evaluate the protective effect of curcumin (Cur) on reproductive toxicity induced by fumonisin B1 (FB1) in laying ducks during the peak egg-laying period. A total of seventy-two 50-week-old Cherry Valley ducks were randomly [...] Read more.
The objective of this study was to evaluate the protective effect of curcumin (Cur) on reproductive toxicity induced by fumonisin B1 (FB1) in laying ducks during the peak egg-laying period. A total of seventy-two 50-week-old Cherry Valley ducks were randomly assigned to four groups: control, FB1 (30 mg/kg), Cur (200 mg/kg), and Cur + FB1 (200 mg/kg + 30 mg/kg). The experiment lasted for 35 days. Our results showed that cur supplementation effectively restored the reductions in final body weight (p = 0.005) and oviduct length (p = 0.020) induced by FB1 exposure. Residual FB1 concentrations in serum, liver, and ovaries were markedly increased in the FB1-treated group, while Cur significantly decreased the FB1 residual in duck liver (p < 0.05). Meanwhile, Cur supplementation markedly counteracted the FB1-induced reductions in serum total protein, albumin, triglycerides, and high-density lipoprotein induced by FB1 exposure. Cur supplementation effectively regulated FB1-induced oxidative stress, inflammation, and endocrine disruption. Specifically, Cur lowered FB1-induced malondialdehyde levels (p < 0.010), attenuated interleukin-1β increase (p = 0.083), and reversed the reduction in immunoglobulin G levels. FB increased the levels of hormones associated with duck reproduction, including estradiol, follicle-stimulating hormone, and luteinizing hormone; in contrast, curcumin supplementation decreased the levels of these hormones (p < 0.010). Histopathological analysis revealed that Cur significantly alleviated the inflammation and necrosis in the liver, kidneys, ovaries, and oviducts induced by FB1. In conclusion, dietary Cur supplementation effectively alleviated FB1-induced reproductive toxicity in laying ducks by enhancing antioxidant capacity, improving lipid metabolism, and restoring hormonal homeostasis. Full article
Show Figures

Figure 1

26 pages, 1242 KB  
Review
The Role of Astaxanthin as an Antioxidant and Anti-Inflammatory Agent in Human Health: A Systematic Review
by Giuseppina Malcangi, Angelo Michele Inchingolo, Lucia Casamassima, Irma Trilli, Laura Ferrante, Marialuisa Longo, Francesco Inchingolo, Grazia Marinelli, Andrea Palermo, Gianna Dipalma and Alessio Danilo Inchingolo
Int. J. Mol. Sci. 2026, 27(2), 700; https://doi.org/10.3390/ijms27020700 - 9 Jan 2026
Viewed by 120
Abstract
This systematic review aimed to summarize the effects of astaxanthin (ASX) supplementation on oxidative stress, inflammation, and metabolic regulation in human studies. A systematic search was conducted in Scopus, Web of Science (WOS), and PubMed for articles published between 2020 and 2025. Fifteen [...] Read more.
This systematic review aimed to summarize the effects of astaxanthin (ASX) supplementation on oxidative stress, inflammation, and metabolic regulation in human studies. A systematic search was conducted in Scopus, Web of Science (WOS), and PubMed for articles published between 2020 and 2025. Fifteen studies involving human participants were included, while in vitro and animal studies were excluded. ASX consistently reduced pro-inflammatory cytokines (IL-6, TNF-α, TGF-β1) and oxidative stress indices while increasing antioxidant capacity (SOD, TAC). Combined ASX and exercise interventions improved body composition, lipid profiles, insulin sensitivity, and immune recovery. In women with Polycystic Ovary Syndrome (PCOS) or endometriosis, ASX downregulated endoplasmic reticulum stress–related apoptotic pathways and improved oocyte and embryo quality. Cardiometabolic and respiratory outcomes showed improved endothelial function and reduced disease severity. Astaxanthin demonstrates broad antioxidant and anti-inflammatory properties, supporting its role as a promising adjunctive therapy for metabolic, reproductive, and cardiovascular health. Further well-designed clinical trials are needed to confirm optimal dosing and mechanisms of action. Full article
18 pages, 953 KB  
Review
The Role of Low CD36 Expression in the Development of Non-Small Cell Lung Cancer and Its Potential for Therapy
by Ran Wu, Xiaohong Xu, Danju Luo, Junhua Wu, Xiaona Chang, Chenggong Ma, Bo Huang, Jun Fan and Xiu Nie
Cancers 2026, 18(2), 217; https://doi.org/10.3390/cancers18020217 - 9 Jan 2026
Viewed by 73
Abstract
Lung cancer remains one of the most prevalent and lethal malignancies worldwide. NSCLC, which constitutes approximately 85% of cases, continues to exhibit a poor prognosis despite advancements in therapeutic interventions, underscoring the urgent necessity to elucidate its molecular mechanisms and identify novel therapeutic [...] Read more.
Lung cancer remains one of the most prevalent and lethal malignancies worldwide. NSCLC, which constitutes approximately 85% of cases, continues to exhibit a poor prognosis despite advancements in therapeutic interventions, underscoring the urgent necessity to elucidate its molecular mechanisms and identify novel therapeutic targets. CD36, a multifunctional transmembrane glycoprotein, is integral to lipid uptake, immune recognition, inflammatory regulation, molecular adhesion, and apoptosis. Increasing evidence implicates CD36 in the progression of various cancers. In the context of lung cancer, CD36 facilitates tumorigenesis through multiple pathways, including the remodeling of tumor cell lipid metabolism, reprogramming of tumor-associated macrophages, and modulation of immune cell functions such as those of Tregs and CD8+ T cells. Additionally, CD36 is intricately linked with the function of cancer-associated fibroblasts and the remodeling of the tumor stromal microvasculature. This systematic review synthesizes the mechanisms by which CD36 contributes to NSCLC proliferation, migration, epithelial–mesenchymal transition, and modulation of the tumor microenvironment. Furthermore, we explore emerging therapeutic strategies that target CD36. Regulating CD36 expression effectively intervenes in the malignant behavior of NSCLC, underscoring its potential as a promising therapeutic target and prognostic marker. Full article
(This article belongs to the Section Cancer Therapy)
22 pages, 1557 KB  
Review
Hawthorn (Crataegus monogyna Jacq.): A Review of Therapeutic Potential and Applications
by Jagoda Kępińska-Pacelik and Wioletta Biel
Molecules 2026, 31(2), 226; https://doi.org/10.3390/molecules31020226 - 9 Jan 2026
Viewed by 278
Abstract
Hawthorn (Crataegus monogyna Jacq.) is a medicinal and nutritional plant widely recognized for its rich phytochemical composition and diverse health-promoting properties. The fruit, leaves, and flowers contain significant amounts of polyphenols, flavonoids, flavonols, phenolic acids and dye compounds with antioxidant properties that [...] Read more.
Hawthorn (Crataegus monogyna Jacq.) is a medicinal and nutritional plant widely recognized for its rich phytochemical composition and diverse health-promoting properties. The fruit, leaves, and flowers contain significant amounts of polyphenols, flavonoids, flavonols, phenolic acids and dye compounds with antioxidant properties that contribute to its strong antioxidant capacity. Numerous studies have demonstrated hawthorn’s beneficial effects on cardiovascular health, including regulation of blood pressure, lipid metabolism, and cardiac function. Additionally, hawthorn exhibits anti-inflammatory, antimicrobial, hypolipidemic, and antidiabetic properties, supporting its role in the prevention and management of chronic diseases. Its potential as a functional food ingredient and natural health supplement is increasingly recognized. However, further clinical trials and standardization of bioactive components are needed to confirm its efficacy, safety, and optimal dosage. Overall, hawthorn represents a valuable natural resource for promoting human health and well-being through diet and phytotherapy. Therefore, the aim of this study is to present—based on the scientific literature—the antioxidant properties of hawthorn and to assess the possibility of using this plant as a functional ingredient. Full article
Show Figures

Figure 1

17 pages, 3619 KB  
Article
Nobiletin Attenuates Inflammation and Modulates Lipid Metabolism in an In Vitro Model of Intestinal Failure-Associated Liver Disease
by Marta Belka, Aleksandra Gostyńska-Stawna, Karina Sommerfeld-Klatta, Maciej Stawny and Violetta Krajka-Kuźniak
Pharmaceutics 2026, 18(1), 87; https://doi.org/10.3390/pharmaceutics18010087 - 9 Jan 2026
Viewed by 136
Abstract
Background: Intestinal failure-associated liver disease (IFALD) is a serious complication in patients receiving parenteral nutrition, often exacerbated by inflammation, lipid overload, and oxidative stress. Nobiletin (NOB), a polymethoxylated flavone, is known for its anti-inflammatory and lipid-regulating properties. Methods: We employed an [...] Read more.
Background: Intestinal failure-associated liver disease (IFALD) is a serious complication in patients receiving parenteral nutrition, often exacerbated by inflammation, lipid overload, and oxidative stress. Nobiletin (NOB), a polymethoxylated flavone, is known for its anti-inflammatory and lipid-regulating properties. Methods: We employed an in vitro model using THLE-2 human hepatocytes and primary human cholangiocytes exposed to Intralipid (INT) and lipopolysaccharide (LPS) to simulate IFALD conditions. NOB was tested at non-toxic concentrations (10 and 25 µM) to assess its protective effects. MTT viability assays, multiplex bead-based immunoassays (MAGPIX), RT-qPCR, and Western blotting were used to evaluate changes in inflammation markers, gene expression, and protein signaling. Moreover, ALT and AST activities were used to assess hepatocellular injury. Results: NOB maintained high cell viability in THLE-2 hepatocytes and cholangiocytes, confirming its low cytotoxicity. NOB normalized ALT and AST activities in both tested cell lines, but the effect reached statistical significance only for ALT in cholangiocytes. Under IFALD-like conditions (LPS+INT), NOB significantly preserved metabolic activity in both cell types. In THLE-2 and cholangiocytes, NOB markedly reduced the phosphorylation of pro-inflammatory proteins JNK, NF-κB, and STAT3, indicating a broad inhibition of inflammatory signaling. Moreover, in THLE-2 cells, NOB upregulated lipid metabolism-related genes (PRKAA2, CYP7A1, and ABCA1) and decreased oxidative stress, thereby enhancing the nuclear translocation of Nrf2 and increasing SOD1 level, which supports the activation of antioxidant defenses. Conclusions: NOB exhibits hepatoprotective properties under IFALD-like conditions in vitro, likely through modulation of inflammation-related signaling and lipid metabolism pathways. Full article
Show Figures

Graphical abstract

Back to TopTop