{"title":"The Effects of α-Linolenic Acid Intake on Skin and Blood Vessel Health and Subjective Fatigue in Middle-Aged Japanese Females: A Randomized, Double-Blind, Placebo-Controlled, Parallel-Group Comparative Trial.","authors":"Nanaka Ando, Naohisa Nosaka, Chie Arai, Kazuhiko Kato","doi":"10.1016/j.tjnut.2025.07.023","DOIUrl":"10.1016/j.tjnut.2025.07.023","url":null,"abstract":"<p><strong>Background: </strong>α-Linolenic acid (ALA) has antioxidant and anti-inflammatory effects. Previous studies have shown its effects on skin, blood vessels, and subjective fatigue; however, most were conducted in populations with low n-3 fatty acid intake.</p><p><strong>Objectives: </strong>We investigated the effects of ALA on skin, vascular health, and subjective fatigue in the Japanese female population, which has a relatively high n-3 fatty acid intake.</p><p><strong>Methods: </strong>This randomized, placebo-controlled, double-blind, parallel trial enrolled 120 Japanese females aged 45-65 y. Participants ingested 1 of the 3 edible oil blends (0.97, 1.36, or 2.13 g/d of ALA) or placebo for 12 wk. Skin condition, vascular health and subjective fatigue were measured. Dietary effects were analyzed using statistical methods that accounted for multiplicity.</p><p><strong>Results: </strong>No significant diet effect was observed for the primary outcome, TEWL (P = 0.428 and P = 0.329, respectively). For exploratory outcomes, a significant diet effect was observed in skin hydration (P = 0.020). Intake of 2.13 g/d of ALA showed a trend toward higher change values than the placebo intake at week 12 (P = 0.054). Brachial-ankle pulse wave velocity significantly decreased in all experimental oil groups compared with that in the control group (P = 0.008-0.068). Serum n-3 fatty acid concentrations, particularly ALA, were significantly higher in all experimental groups than those in the control group (P = 0.0002-0.035). No significant differences were observed between groups for subjective fatigue (visual analog scale: smallest P = 0.102; POMS2: smallest P = 0.15).</p><p><strong>Conclusions: </strong>Even in the Japanese population, which has a mean consumption of high amounts of n-3 fatty acids, it is shown that ALA intake can increase the concentration of n-3 fatty acids in blood serum and contribute to maintaining the health of blood vessels. In addition, in people of normal weight, ALA increased skin hydration and contributed to maintaining skin health. This study was registered at UMIN-CTR before the start of the study as UMIN-ID: UMIN000052677 (https://center6.umin.ac.jp/cgi-open-bin/ctr/ctr_view.cgi?recptno=R000060078).</p>","PeriodicalId":16620,"journal":{"name":"Journal of Nutrition","volume":" ","pages":""},"PeriodicalIF":3.8,"publicationDate":"2025-08-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144775661","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Ana M Palacios, Dominick J Lemas, Bridget E Young, Erik Parker, Stephanie Dickinson, Nandi Marshall, Kelly L Sullivan, Haley Wilt, Michelle I Cardel, David B Allison
{"title":"Associations among Human Milk Polyunsaturated Fatty Acids and Infant Sleep Patterns: A Cross-Sectional Study.","authors":"Ana M Palacios, Dominick J Lemas, Bridget E Young, Erik Parker, Stephanie Dickinson, Nandi Marshall, Kelly L Sullivan, Haley Wilt, Michelle I Cardel, David B Allison","doi":"10.1016/j.tjnut.2025.07.021","DOIUrl":"10.1016/j.tjnut.2025.07.021","url":null,"abstract":"<p><strong>Background: </strong>Infant sleep is critical for cognitive, emotional, and long-term health outcomes. Although diet-sleep relationships are established, limited research has explored how polyunsaturated fatty acids (PUFAs) in human milk (HM) relate to infant sleep.</p><p><strong>Objectives: </strong>This study aims to examine associations between PUFAs in HM and sleep patterns in 2-mo-old infants exclusively fed with HM.</p><p><strong>Methods: </strong>This cross-sectional secondary analysis used data from a lactation cookie trial involving 131 parents of healthy, term infants. Participants provided a fasting HM sample and completed sociodemographic and Brief Infant Sleep Questionnaire surveys. Multivariate linear models were adjusted for infant sex assigned at birth, weight-for-length z-scores, maternal prepregnancy body mass index, and daily feedings.</p><p><strong>Results: </strong>Participants self-identified as 81.7% White, 13.3% Hispanic, with 25.9% reporting an annual income <US$45,000. Infants slept a mean of 14.67 h [standard deviation (SD) = 2.65]. PUFAs constituted 20.91% (SD = 4.47) of total milk fatty acids, with omega-3 and omega-6 comprising 1.75% (SD = 0.53) and 19.16% (SD = 4.09), respectively. Significant positive associations were observed between total sleep and omega-3 [B = 1.06, standard error (SE) = 0.47, P = 0.026], omega-6 (B = 0.14, SE = 0.06, P = 0.022), alpha-linolenic acid (ALA) (B = 1.10, SE = 0.53, P = 0.040), and linoleic acid (LA) (B = 0.15, SE = 0.06, P = 0.016). When sleep was partitioned into diurnal or nocturnal, greater omega-3 and omega-6, particularly ALA and LA, were significantly associated with increased diurnal sleep duration: (ALA, B = 1.11, SE = 0.39, P = 0.005; LA, B = 0.12, SE = 0.05, P = 0.009). No significant associations were observed with nocturnal sleep.</p><p><strong>Conclusions: </strong>Higher HM proportions of omega-3 and omega-6, particularly ALA and LA, were associated with greater diurnal sleep in exclusively HM-fed 2 month olds. Further research is needed to explore underlying mechanisms and long-term developmental impacts. This trial was registered at clinicaltrials.gov as NCT04805008.</p>","PeriodicalId":16620,"journal":{"name":"Journal of Nutrition","volume":" ","pages":""},"PeriodicalIF":3.8,"publicationDate":"2025-08-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144775659","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"L-Malic Acid Improves the Metabolic Health of Gestating Sows and Drives Skeletal Muscle Fiber Remodeling in the Offspring.","authors":"Haijun Sun, Qiyuan Miao, Boyang Wan, Jialong Liao, Niannian Tian, Zhuo Chang, Jingdong Yin, Xin Zhang","doi":"10.1016/j.tjnut.2025.07.018","DOIUrl":"10.1016/j.tjnut.2025.07.018","url":null,"abstract":"<p><strong>Background: </strong>L-malic acid (MA) consumption in sows during late gestation could improve glucose metabolism and increase myogenin expression in the skeletal muscle of the offspring.</p><p><strong>Objectives: </strong>This study aimed to investigate the impact of maternal MA intake during the critical period of skeletal muscle development on muscle development and metabolic traits in offspring.</p><p><strong>Methods: </strong>At day 30 of gestation, 45 Landrace × Yorkshire sows were divided into 3 groups and received either a corn-soybean basal diet (control) or the basal diet supplemented with 1% or 2% MA complex. Sows' serum was collected at day 50 and 90 of gestation. Levels of inflammation markers and myokines were measured in piglet serum at birth and at weaning (21 day of age). The longissimus dorsi muscle was also collected to assess skeletal muscle growth and development. Statistical analyses included t-tests, 1-way or 2-way analysis of variance.</p><p><strong>Results: </strong>Dietary MA supplementation reduced inflammation and oxidative stress in sows. MA increased serum insulin-like growth factor 1 by 18.51% (P < 0.01) and decreased transforming growth factor β1 concentrations by 42.77% (P < 0.01) in sows, particularly at day 50 of gestation. Notably, maternal MA reduced inflammation and altered myokine secretion. It also increased paired box 7 expression in piglet skeletal muscle by 74.29% at day 1 (P < 0.05), and induced muscle fiber-type transition at day 21. This is evidenced by increased slow myosin heavy chain protein concentrations (P < 0.05) and decreased myosin heavy chain-IIx messenger ribonucleic acid (RNA) concentrations (P < 0.05). RNA-sequential analysis revealed that maternal MA consumption affected the expression of genes involved in amino acid and fatty acid metabolism in the skeletal muscle of piglets at day 1, while inhibiting the Hippo signaling pathway.</p><p><strong>Conclusions: </strong>These findings suggest that maternal MA consumption is a potential strategy for remodeling skeletal muscle function and improving metabolic health in offspring. This porcine model will provide translational insights into maternal nutrition interventions for human skeletal muscle development.</p>","PeriodicalId":16620,"journal":{"name":"Journal of Nutrition","volume":" ","pages":""},"PeriodicalIF":3.8,"publicationDate":"2025-07-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144768709","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Shan Sun, Xiangzhu Zhu, Xiang Huang, Chang Yu, Timothy Su, Harvey J Murff, Reid M Ness, M Andrea Azcarate-Peril, Martha J Shrubsole, Qi Dai
{"title":"The Association of Gut Microbiota With TRPM7 Genotype, Colorectal Polyps, and Magnesium.","authors":"Shan Sun, Xiangzhu Zhu, Xiang Huang, Chang Yu, Timothy Su, Harvey J Murff, Reid M Ness, M Andrea Azcarate-Peril, Martha J Shrubsole, Qi Dai","doi":"10.1016/j.tjnut.2025.07.015","DOIUrl":"10.1016/j.tjnut.2025.07.015","url":null,"abstract":"<p><strong>Background: </strong>We previously reported that individuals with the transient receptor potential melastatin 7 (TRPM7) GA/AA genotype and consumed diets high in Ca:Mg ratio had increased risk of colorectal polyps.</p><p><strong>Objectives: </strong>The aim was to identify whether the gut microbiota plays a role in the association of TRPM7 genotype, Ca:Mg intake ratio, and risk of colorectal polyps.</p><p><strong>Methods: </strong>We analyzed the gut microbiota of 240 participants in a double-blind 2 × 2 factorial (TRPM7 genotype and Ca:Mg intake ratios) randomized trial by sequencing the stool, rectal swab, and rectal mucosa tissue samples of each participant.</p><p><strong>Results: </strong>The gut microbiota of participants with the GA genotype significantly differed from those with the GG genotype in all 3 sample types, with an altered abundance of Prevotella and Bacteroides in swab samples. Prevotella in rectal mucosa and Bacteroides in swab were associated with an increased risk of metachronous colorectal polyps. Optimizing high-diet Ca:Mg ratios to 2.3 through Mg supplementation resulted in a reduced abundance of Prevotella in rectal swabs and Bacteroides in stool samples. We identified multiple microbes in all 3 sample types linked to risk of metachronous colorectal polyps.</p><p><strong>Conclusions: </strong>Our findings indicate that the gut microbiota in stool, rectal swab, and mucosa are associated with risk of metachronous colorectal polyps, and diet changes could modify the abundance of TRPM7-related microbes. This study was registered at clinicaltrials.gov as NCT01105169 (https://clinicaltrials.gov/study/NCT01105169).</p>","PeriodicalId":16620,"journal":{"name":"Journal of Nutrition","volume":" ","pages":""},"PeriodicalIF":3.8,"publicationDate":"2025-07-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144765011","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Physiologically Based Pharmacokinetic Modeling of Vitamin B-12 Incorporating Mechanistic Absorption: An Example Application for Intake Estimation During Pregnancy.","authors":"Mian Zhang, Lisa M Almond, Hannah M Jones","doi":"10.1016/j.tjnut.2025.07.019","DOIUrl":"10.1016/j.tjnut.2025.07.019","url":null,"abstract":"<p><strong>Background: </strong>Vitamin B-12 (cobalamin) is an essential micronutrient required for neurologic function, hematopoiesis, and DNA synthesis. Oral absorption of vitamin B-12 occurs through an intrinsic factor (IF)-dependent pathway at physiological intake amounts supplemented by passive diffusion at pharmacologic doses. Vitamin B-12 deficiency is a global public health concern, with certain populations such as older adults and pregnant women at higher risk. Despite adherence to intake recommendations, a substantial proportion of pregnant women exhibit suboptimal vitamin B-12 status. A pharmacokinetic model may improve the evaluation of intake requirements across different populations.</p><p><strong>Objectives: </strong>This study aimed to develop a physiologically based pharmacokinetic (PBPK) model of vitamin B-12 absorption and apply it to evaluate intake requirements under altered physiological conditions.</p><p><strong>Methods: </strong>A PBPK model incorporated key absorption mechanisms, including vitamin B-12-IF binding, cubilin-mediated uptake, and passive diffusion. The model was applied to simulate vitamin B-12 absorption across a range of intake doses and to predict plasma vitamin B-12 concentrations under reduced IF secretion and increased systemic clearance reflective of pregnancy-related changes.</p><p><strong>Results: </strong>The model successfully reproduced the nonlinear absorption behavior of vitamin B-12 and its dependence on IF and cubilin. Simulated absorption percentages following single oral doses of 1, 2, and 1000 μg (83%, 63%, and 1%, respectively) were within the expected range. Simulations with unlimited cubilin availability indicated that cubilin-mediated uptake is the rate-limiting step in IF-dependent absorption. In pregnancy simulations, a daily intake of 150 μg, divided into three 50-μg doses at 6-h intervals, was required to restore plasma vitamin B-12 concentrations depleted by pregnancy to an adequate concentration of >300 pmol/L, whereas the same amount given as a single daily dose did not achieve this concentration.</p><p><strong>Conclusions: </strong>This study demonstrates the utility of PBPK modeling in providing mechanistic understanding of vitamin B-12 absorption and supporting intake recommendations for populations with altered physiological states.</p>","PeriodicalId":16620,"journal":{"name":"Journal of Nutrition","volume":" ","pages":""},"PeriodicalIF":3.8,"publicationDate":"2025-07-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144765010","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Associations of Fish Oil Supplementation With Risks of Osteoarthritis and Arthroplasty: A Large Population-Based Prospective Cohort Study.","authors":"Zhilan Li, Yancong Chen, Yinyan Gao, Boya Xu, Vivian Yawei Guo, Weiru Zhang, Irene Xy Wu","doi":"10.1016/j.tjnut.2025.07.017","DOIUrl":"10.1016/j.tjnut.2025.07.017","url":null,"abstract":"<p><strong>Background: </strong>Fish oil, rich in ω-3 (n-3) PUFAs, has been considered beneficial for human health; however, mixed effects on osteoarthritis (OA) and arthroplasty risk were reported.</p><p><strong>Objectives: </strong>This study aimed to examine the association between fish oil supplementation and incident OA and arthroplasty in a large population-based prospective cohort.</p><p><strong>Methods: </strong>In total, 255,853 participants from the UK Biobank were included and divided into fish oil users and nonusers according to their answers to a questionnaire at baseline. The association between fish oil use and different sites of OA, knee arthroplasty, and hip arthroplasty incidence was assessed using weighted Cox regression models.</p><p><strong>Results: </strong>During a median of 10.90, 11.44, and 11.47 y of follow-up, 33,290, 7734, and 6376 participants developed all OA, experienced hip arthroplasty, or experienced knee arthroplasty, respectively. After adjustment for multiple covariates, fish oil supplementation was associated with significantly increased risks of all OA [hazard ratio (HR): 1.18; 95% CI: 1.14, 1.23], multisite OA (HR: 1.15; 95% CI: 1.05, 1.25), knee OA (HR: 1.25; 95% CI: 1.19, 1.32), hip OA (HR: 1.22; 95% CI: 1.13, 1.31), hand OA (HR: 1.19; 95% CI: 1.06, 1.34), hip arthroplasty (HR: 1.25; 95% CI: 1.16, 1.35), and knee arthroplasty (HR: 1.37; 95% CI: 1.27, 1.49), whereas no significant association was found between fish oil supplementation and spine OA (HR: 1.05; 95% CI: 0.94, 1.18). Subgroup analyses showed the association between fish oil use and risks of OA and arthroplasty was modified by age and BMI (P-interaction < 0.05).</p><p><strong>Conclusions: </strong>Fish oil supplementation is positively associated with risks of OA and arthroplasty. Our findings provide new evidence highlighting the importance of cautious use of fish oil supplements and the need for informed dietary strategies for OA prevention. Further research is warranted to understand the underlying mechanisms.</p>","PeriodicalId":16620,"journal":{"name":"Journal of Nutrition","volume":" ","pages":""},"PeriodicalIF":3.8,"publicationDate":"2025-07-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144765009","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Creatine Supplementation for Cognition: A Critical Perspective on Promise, Proof, and Public Perception.","authors":"Igor Eckert","doi":"10.1016/j.tjnut.2025.07.016","DOIUrl":"10.1016/j.tjnut.2025.07.016","url":null,"abstract":"<p><p>Creatine is widely regarded as a safe and effective supplement for physical performance but has recently gained attention as a potential cognitive enhancer. Public and commercial enthusiasm for this use has surged, spiking sales, prescriptions and social media engagement despite a body of evidence that, paradoxically, haved remained considerably limited. This article critically examines the growing disconnect between marketing narratives and scientific rigor in the discourse around creatine and brain health. It also explores how biases, both cognitive and structural, shapes how the evidence is interpreted and communicated. Rather than dismissing creatine's potential, the essay argues for greater critical appraisal and transparency in how health claims are evaluated and disseminated, to ensure that conversations remain anchored to what the data can actually support.</p>","PeriodicalId":16620,"journal":{"name":"Journal of Nutrition","volume":" ","pages":""},"PeriodicalIF":3.8,"publicationDate":"2025-07-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144760374","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Karen E Christensen, Marie-Lou Faquette, Aaron T Gebert, Vafa Keser, Teodoro Bottiglieri, Rima Rozen
{"title":"Contrasting Effects of Phosphatidylcholine and Betaine Supplementation on Embryonic Development in a Mouse Model of the MTHFD1 R653Q Variant.","authors":"Karen E Christensen, Marie-Lou Faquette, Aaron T Gebert, Vafa Keser, Teodoro Bottiglieri, Rima Rozen","doi":"10.1016/j.tjnut.2025.07.013","DOIUrl":"10.1016/j.tjnut.2025.07.013","url":null,"abstract":"<p><strong>Background: </strong>Folate and choline are interconnected nutrients important for embryonic development. Genetic variants in folate enzymes, such as MTHFD1 R653Q (c.1958 G>A, rs2236225), may increase dietary choline requirements.</p><p><strong>Objectives: </strong>To evaluate the impact of phosphatidylcholine or betaine supplementation on embryonic development and maternal choline metabolism in the Mthfd1S<sup>+/-</sup> mouse model for the MTHFD1 653QQ genotype.</p><p><strong>Methods: </strong>Female Mthfd1S<sup>+/+</sup> (wild type [WT]) and Mthfd1S<sup>+/-</sup> (heterozygous [HET]) mice were fed amino acid-defined control (Ctrl), phosphatidylcholine-supplemented (PtdCho), or betaine-supplemented (BTN) diets for 4-6 wk before mating and during pregnancy (18-33 mice/group). Ctrl contained 2.5 g choline bitartrate/kg diet (based on AIN-93G). Supplemented diets contained additional phosphatidylcholine or betaine to obtain the molar equivalent of 3 times the free choline of Ctrl. Embryos were examined for anatomical developmental delays and morphological defects at embryonic day 10.5. One-carbon metabolites (homocysteine, choline, betaine, methionine, cystathionine, S-adenosylmethionine, S-adenosylhomocysteine) were measured in maternal plasma and liver by liquid chromatography-mass spectrometry (LC-MS/MS). The MTHFR protein in maternal liver was assessed by Western blot. Data were analyzed by binary logistic regression or 2-way analysis of variance.</p><p><strong>Results: </strong>PtdCho increased developmental delays, compared with Ctrl, in litters of WT (170%) and HET (125%) mothers (P<sub>diet</sub> < 0.05). BTN decreased delays 53% compared with Ctrl in HET mother's litters (P<sub>diet (HET)</sub> < 0.05). Defects decreased 71% in BTN-fed HET mother's litters compared to WT (P < 0.05). BTN increased total pregnancy losses 360% (P<sub>diet</sub> < 0.01); PtdCho had no effect. Both diets decreased maternal plasma homocysteine (30-50%, P<sub>diet</sub> < 0.05), increased liver BTN (220-290%, P<sub>diet</sub> < 0.0001), and decreased S-adenosylmethionine/S-adenosylhomocysteine (WT: 20-30%, HET: 48-61%, P<sub>diet</sub> < 0.05). MTHFR protein expression decreased 25% in PtdCho maternal liver (P<sub>diet</sub> < 0.01).</p><p><strong>Conclusions: </strong>Phosphatidylcholine and betaine supplementation had contrasting effects on reproductive outcomes. PtdCho increased developmental delays. BTN reduced delays and defects in the Mthfd1S<sup>+/-</sup> mouse model, but may impair the establishment of pregnancy. The timing, form, and dosage of choline supplementation may be important and require further investigation.</p>","PeriodicalId":16620,"journal":{"name":"Journal of Nutrition","volume":" ","pages":""},"PeriodicalIF":3.8,"publicationDate":"2025-07-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144718051","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Fredric W. Hill, Ph.D. (1918-2003)","authors":"D’Ann Finley, Robert B Rucker","doi":"10.1016/j.tjnut.2025.07.001","DOIUrl":"10.1016/j.tjnut.2025.07.001","url":null,"abstract":"","PeriodicalId":16620,"journal":{"name":"Journal of Nutrition","volume":"155 9","pages":"Pages 3134-3136"},"PeriodicalIF":3.8,"publicationDate":"2025-07-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144731862","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Blueberry Bioactives in Metabolic Syndrome","authors":"Kexin Liu, Bing Chen","doi":"10.1016/j.tjnut.2025.07.014","DOIUrl":"10.1016/j.tjnut.2025.07.014","url":null,"abstract":"<div><div>Metabolic syndrome (MetS) is a special disorder with abnormal lipid levels, excessive body fat, high blood pressure, and insulin resistance. It is commonly associated with chronic diseases like diabetes, obesity, and cardiovascular diseases (CVDs). Blueberries are rich in bioactive substances, including flavonoids and phenolic acids. These substances have garnered interest for their potential to improve health and manage MetS. Beyond the health benefits, blueberries may offer potential applications in the food industry. This review provides an in-depth analysis of the bioactive components of blueberries and their roles in mitigating various aspects of MetS, including hyperlipidemia, obesity, nonalcoholic fatty liver disease (NAFLD), hyperglycemia, diabetes, hypertension, and CVDs. Future research should focus on exploring the clinical applications of compounds derived from blueberries and their synergistic effects with other therapies to develop more effective approaches for preventing and treating MetS and related conditions.</div></div>","PeriodicalId":16620,"journal":{"name":"Journal of Nutrition","volume":"155 9","pages":"Pages 2779-2798"},"PeriodicalIF":3.8,"publicationDate":"2025-07-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144707832","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}