Nothing Special   »   [go: up one dir, main page]

You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (923)

Search Parameters:
Keywords = hypercholesterolemia

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
22 pages, 12321 KiB  
Article
Impact of Kiwifruit Consumption on Cholesterol Metabolism in Rat Liver: A Gene Expression Analysis in Induced Hypercholesterolemia
by Abdolvahab Ebrahimpour Gorji, Anna Ciecierska, Hanna Leontowicz, Zahra Roudbari and Tomasz Sadkowski
Nutrients 2024, 16(23), 3999; https://doi.org/10.3390/nu16233999 - 22 Nov 2024
Abstract
Background/Objectives: Cholesterol is vital in various bodily functions, such as maintaining cell membranes, producing hormones, etc. However, imbalances, like hypercholesterolemia, can lead to diseases such as cancer, kidney disease, non-alcoholic fatty liver disease, and cardiovascular conditions. This study explores the impact of kiwifruit [...] Read more.
Background/Objectives: Cholesterol is vital in various bodily functions, such as maintaining cell membranes, producing hormones, etc. However, imbalances, like hypercholesterolemia, can lead to diseases such as cancer, kidney disease, non-alcoholic fatty liver disease, and cardiovascular conditions. This study explores the impact of kiwifruit consumption, specifically Actinidia arguta cultivar Geneva and Actinidia deliciosa cultivar Hayward, on cholesterol and lipid metabolism in rat liver. Methods: Rats were divided into groups: a 1% cholesterol control group (Ch), a 5% Geneva kiwifruit-supplemented group (ChGENE), and a 5% Hayward kiwifruit-supplemented group (ChHAYW). Gene expression was analyzed using Gene Spring v.14. Gene ontology, pathway analysis, miRNA, and transcription factor prediction were performed using DAVID, Reactome, and miRNet. In addition, we used Agilent Literature Search software to gain further insights. Results: Statistical analysis identified 72 genes in ChGENE-Ch and 2 genes in ChHAYW-Ch comparison. Key genes involved in cholesterol metabolism pathways, including PCSK9, SCD1, SLC27A5, HMGCR, and DHCR24, showed lower expression in the kiwifruit-supplemented groups. The genes mentioned above showed lower expression in the kiwifruit-supplemented group, probably contributing to the liver lipid level reduction. Further analysis identified miRNA-26a, miRNA-29a/b/c, miRNA-33a/b, and miRNA-155 targeting hub genes. Conclusions: Our findings suggest that dietary supplementation with kiwifruit, particularly the Geneva cultivar, reduces fat accumulation in the liver of rats with hypercholesterolemia, likely through downregulation of critical genes involved in cholesterol metabolism. These studies highlight the potential of kiwifruit as a part of a dietary strategy to manage cholesterol levels. Full article
(This article belongs to the Section Nutrition and Public Health)
16 pages, 952 KiB  
Perspective
Effective Strategies and a Ten-Point Plan for Cardio-Kidney-Metabolic Health in Croatia: An Expert Opinion
by Željko Reiner, Bojan Jelaković, Davor Miličić, Marija Bubaš, Ines Balint, Nikolina Bašić Jukić, Valerija Bralić Lang, Vili Beroš, Ivana Brkić Biloš, Silvija Canecki Varžić, Krunoslav Capak, Verica Kralj, Ana Ljubas, Branko Malojčić, Viktor Peršić, Ivana Portolan Pajić, Dario Rahelić, Alen Ružić, Tomislav Sokol, Ana Soldo and Ivan Pećinadd Show full author list remove Hide full author list
J. Clin. Med. 2024, 13(23), 7028; https://doi.org/10.3390/jcm13237028 - 21 Nov 2024
Viewed by 204
Abstract
Cardiovascular diseases (CVD) are the leading cause of morbidity and mortality worldwide, including in Croatia. Since most patients have multiple disorders and diseases caused largely by the same risk factors, and as it is essential to approach each patient as a person with [...] Read more.
Cardiovascular diseases (CVD) are the leading cause of morbidity and mortality worldwide, including in Croatia. Since most patients have multiple disorders and diseases caused largely by the same risk factors, and as it is essential to approach each patient as a person with all disorders, today, we are talking about a new paradigm—cardio-renal-metabolic (CKM) syndrome and cardio-renal-metabolic health, which necessarily includes brain health. Elevated systolic blood pressure, LDL cholesterol, smoking, obesity, diabetes, impaired renal function or chronic kidney disease, which all stem from insufficient physical activity, an unhealthy diet with excessive intake of table salt, and air pollution, are the leading causes of overall morbidity and mortality from CKM diseases, especially mortality from CVD. Experts from various fields key to CKM health have written this document with the aim of integrating it as part of the national plan for the prevention of chronic non-communicable diseases with a focus on CVD, which should become mandatory and be based on the existing guidelines of professional societies. Full article
(This article belongs to the Section Nephrology & Urology)
18 pages, 1707 KiB  
Article
Portuguese Lipid Study (e_LIPID)
by Joana Rita Chora, Ana Catarina Alves, Cibelle Mariano, Quitéria Rato, Marília Antunes and Mafalda Bourbon
J. Clin. Med. 2024, 13(22), 6965; https://doi.org/10.3390/jcm13226965 - 19 Nov 2024
Viewed by 236
Abstract
Background/Objectives: Incidence of cardiovascular disease (CVD) is increasing in low- and middle-income countries because of changing lifestyles. Since dyslipidaemia is a major independent cardiovascular risk factor, its correct identification is critical to implement specific interventions for CVD prevention. This study aimed to [...] Read more.
Background/Objectives: Incidence of cardiovascular disease (CVD) is increasing in low- and middle-income countries because of changing lifestyles. Since dyslipidaemia is a major independent cardiovascular risk factor, its correct identification is critical to implement specific interventions for CVD prevention. This study aimed to characterise the lipid profile of the Portuguese population. Methods: Overall, 1688 individuals from the general population (e_COR study, 2012–2014) were included. Population-specific percentiles for ten lipid biomarkers were estimated by bootstrapping methods to ensure national representativity. Statistical analyses were performed using RStudio. Results: The 50th percentile estimated for total cholesterol (TC), LDL-C, and non-HDL-C are similar to scientific societies recommended values for the general (low or moderate risk) population. National prevalence of having lipid parameters above recommended values was 64.6%, 66.9%, 51.3%, 68.9%, 17.8%, and 21.1% for TC, LDL-C, apoB, non-HDL-C, triglycerides, and Lp(a), respectively; these values are generally higher in men and increasing with age, except for Lp(a). A high prevalence of severe dyslipidaemia (>90th percentile) was identified, highest for small dense LDL-C (31.3%), apoB (30.4%), and LDL-C (30.3%). The national prevalence of CVD events was 5%. Three individuals were genetically identified with familial hypercholesterolemia, a high CVD risk condition. Conclusions: We provide for the first-time lipid biomarker percentiles for the general Portuguese population. Our results highlight that hypercholesterolemia is a neglected cardiovascular risk factor with over half of the population with TC, LDL-C, and apoB above recommended values. Since hypercholesterolemia is a modifiable risk factor, strategies to increase adherence to changes in lifestyle habits and medication need to be urgently discussed. Full article
(This article belongs to the Section Cardiovascular Medicine)
Show Figures

Figure 1

Figure 1
<p>Prevalence and 95% CI of lipid parameters above the recommended values for low and moderate risk in the Portuguese population by sex (<b>A</b>) and age (<b>B</b>). ApoB, apolipoprotein B; CI, confidence interval; LDL-C, low-density lipoprotein cholesterol; Lp(a), lipoprotein (a); non-HDL-C, non-high-density lipoprotein cholesterol; TC, total cholesterol; TG, triglycerides.</p>
Full article ">Figure 2
<p>Prevalence and 95% CI of dyslipidaemia (lipid parameters above P90 or below P10) in the Portuguese population by sex (<b>A</b>) and age (<b>B</b>). ApoA1, apolipoprotein A1; ApoB, apolipoprotein B; CI, confidence interval; HDL, high-density lipoprotein cholesterol; LDL-C, low-density lipoprotein cholesterol; Lp(a), lipoprotein (a); non-HDL-C, non-high-density lipoprotein cholesterol; sdLDL-C, small dense low-density lipoprotein cholesterol; TC, total cholesterol; TG, triglycerides; VLDL, very-low-density lipoprotein cholesterol.</p>
Full article ">Figure 3
<p>Odds ratio for having a previous CVD event, given the lipid profile. * This analysis was performed on all individuals (vs only individuals not under statins). ApoB, apolipoprotein B; LDL-C, low-density lipoprotein cholesterol; Lp(a), lipoprotein (a); non-HDL-C, non-high-density lipoprotein cholesterol; TC, total cholesterol; TG, triglycerides.</p>
Full article ">
15 pages, 452 KiB  
Article
Prediction Models for Elevated Cardiac Biomarkers from Previous Risk Factors and During the COVID-19 Pandemic in Residents of Trujillo City, Peru
by Joao Caballero-Vidal, Jorge Luis Díaz-Ortega, Irma Luz Yupari-Azabache, Luz Angélica Castro-Caracholi and Juan M. Alva Sevilla
Diagnostics 2024, 14(22), 2503; https://doi.org/10.3390/diagnostics14222503 - 8 Nov 2024
Viewed by 579
Abstract
Cardiac troponin serum concentration is a marker of myocardial injury, but NT-pro BNP is a marker of myocardial insufficiency. The purpose of this study was to determine binary logistic regression models to verify the possible association of cardiovascular risk indicators, pre-pandemic history, the [...] Read more.
Cardiac troponin serum concentration is a marker of myocardial injury, but NT-pro BNP is a marker of myocardial insufficiency. The purpose of this study was to determine binary logistic regression models to verify the possible association of cardiovascular risk indicators, pre-pandemic history, the number of times participants were infected with SARS-CoV-2, and vaccination against these biomarkers. A total of 281 residents of Trujillo city (Peru) participated between September and December 2023. We found a high prevalence of abdominal obesity of 55.2%; glycemia > 100 m/dL in 53%; hypercholesterolemia in 49.8%; low HDL in 71.9%; and LDL > 100 mg/dL in 78.6%. A total of 97.5% were vaccinated against COVID-19, and 92.2% had three or more doses. Also, 2.5% had cTnI > 0.05 ng/mL, and 3.3% had NT-proBNP > 125 pg/mL. The number of COVID-19 infections versus cTnI > 0.05 ng/mL presented an OR = 3.513 (p = 0.003), while for NT-proBNP > 125 pg/mL, the number of comorbidities presented an OR = 2.185 (p = 0.025) and LDL an OR = 0.209 (p = 0.025). A regression model was obtained in which there is an association between a higher number of COVID-19 infections and elevated cTnI values and a model implying an association of the number of comorbidities and LDL with the NT-proBNP level in a direct and inverse manner, respectively. Both models contribute to the prevention of cardiac damage. Full article
(This article belongs to the Section Clinical Laboratory Medicine)
Show Figures

Figure 1

Figure 1
<p>Flow diagram of subject inclusion and exclusion.</p>
Full article ">
11 pages, 1040 KiB  
Case Report
Targeted NGS Revealed Pathogenic Mutation in a 13-Year-Old Patient with Homozygous Familial Hypercholesterolemia: A Case Report
by Ayaulym E. Chamoieva, Zhanel Z. Mirmanova, Madina R. Zhalbinova, Saule E. Rakhimova, Asset Z. Daniyarov, Ulykbek Y. Kairov, Almira I. Baigalkanova, Murat A. Mukarov, Makhabbat S. Bekbossynova and Ainur R. Akilzhanova
Int. J. Mol. Sci. 2024, 25(22), 11882; https://doi.org/10.3390/ijms252211882 - 5 Nov 2024
Viewed by 460
Abstract
Familial hypercholesterolemia is an autosomal hereditary disease defined by an increased level of low-density lipoprotein cholesterol (LDL-C), which predisposes significant risks for premature cardiovascular disorders. We present a family trio study: proband, a 13-year-old Kazakh girl with homozygous familial hypercholesterolemia (HoFH) and her [...] Read more.
Familial hypercholesterolemia is an autosomal hereditary disease defined by an increased level of low-density lipoprotein cholesterol (LDL-C), which predisposes significant risks for premature cardiovascular disorders. We present a family trio study: proband, a 13-year-old Kazakh girl with homozygous familial hypercholesterolemia (HoFH) and her parents. HoFH is much more rare and severe than a heterozygous form of the disorder. HoFH patients generally present with LDL-C levels exceeding 13 mmol/L, resulting in early and life-threatening cardiovascular events within the first decades of life. In cases of neglected treatment, young patients have a risk of death from coronary diseases before the age of 30. The aim of this research was to identify genetic mutations in the affected patient and her parents. Genetic testing was necessary due to highly elevated LDL-C levels and the presence of multiple xanthomas. Targeted next-generation sequencing (NGS) was performed in this study using the Illumina TruSight cardio panel, which targets 174 genes related to cardiac disorders. The girl was diagnosed with HoFH based on the results of genetic testing. A biallelic mutation was observed in exon 3 of the low-density lipoprotein receptor (LDLR): c. 295 G>A (p.Glu99Lys). Sanger sequencing confirmed that the mutant gene was inherited from both parents. After confirming the genetic diagnosis of HoFH, the patient was treated with LDL apheresis and statins. This case report is the first study of HoFH in a pediatric patient from the Central Asian region. Globally, it emphasizes the need for increased clinical awareness among healthcare providers, as early detection and intervention are important for improving outcomes, particularly in pediatric patients with this rare genetic disorder. Full article
(This article belongs to the Section Molecular Genetics and Genomics)
Show Figures

Figure 1

Figure 1
<p>Xanthomas in a 13-year-old girl: (<b>A</b>) finger xanthomas, (<b>B</b>) Achilles tendon xanthomas.</p>
Full article ">Figure 2
<p>Sanger sequencing and co-segregation analysis in the family trio. The results of c.295 G&gt;A (p.Glu99Lys) mutation in <span class="html-italic">LDLR</span>: (<b>A</b>) 13-year-old girl (proband), (<b>B</b>) father, (<b>C</b>) mother. The arrows indicate peaks corresponding to a specific nucleotide (A, T, C, or G) change. Y–nucleotide code, indicating the presence of either C or T.</p>
Full article ">Figure 3
<p>The pedigree of the family (<b>a</b>): III 2—proband, homozygous carrier of c. 295 G&gt;A (p.Glu99Lys); II 1—father, heterozygous carrier of c. 295 G&gt;A (p.Glu99Lys); II 2—mother, heterozygous carrier of c. 295 G&gt;A (p.Glu99Lys); and the family members’ phenotypes (<b>b</b>).</p>
Full article ">
15 pages, 3256 KiB  
Article
The MARVIN Hypothesis: Linking Unhealthy Lifestyles to Intracranial Aneurysm Rupture Risk and Clinical Prognosis
by Vanessa M. Swiatek, Igor Fischer, Rajiv Khajuria, Amir Amini, Hannah Steinkusch, Ali Rashidi, Klaus-Peter Stein, Claudia A. Dumitru, I. Erol Sandalcioglu and Belal Neyazi
Medicina 2024, 60(11), 1813; https://doi.org/10.3390/medicina60111813 - 4 Nov 2024
Viewed by 511
Abstract
Background and Objectives: The rising incidence of modifiable lifestyle risk factors and cardiovascular diseases, driven by poor diet, inactivity, excessive alcohol use, and smoking, may influence the development and rupture of intracranial aneurysms (IA). This study aimed to examine the impact of [...] Read more.
Background and Objectives: The rising incidence of modifiable lifestyle risk factors and cardiovascular diseases, driven by poor diet, inactivity, excessive alcohol use, and smoking, may influence the development and rupture of intracranial aneurysms (IA). This study aimed to examine the impact of lifestyle-related and cardiovascular risk factors on IA rupture and patient outcomes. Materials and Methods: We developed the “MARVIN” (Metabolic and Adverse Risk Factors and Vices Influencing Intracranial Aneurysms) model and conducted a retrospective analysis of 303 patients with 517 IAs, treated between 2007 and 2020. Of these, 225 patients were analyzed for rupture status and 221 for clinical outcomes. The analysis focused on hypertension, diabetes, hypercholesterolemia, vascular diseases, nicotine and alcohol abuse, obesity, aneurysm rupture status, and clinical outcomes. Logistic regression was used to evaluate the impact of these risk factors. Results: Among those with risk factors, 24.9% (56/225) and 25.3% (56/221) had one, 32.0% (72/225) and 30.8% (68/221) had two, 20.0% (45/225) and 20.4% (45/221) had three, 12.0% (27/225) and 12.2% (27/221) had four, 4.0% (9/225) and 4.1% (9/221) had five, 0.9% (2/225) had six in both groups, and 0.4% (1/225) and 0.5% (1/221) had seven risk factors, respectively. Strong relationships were found between lifestyle-related vascular risk factors, indicating multiple comorbidities in patients with unhealthy habits. Smokers with ruptured aneurysms had higher WFNS (World Federation of Neurosurgical Societies) scores, but nicotine abuse did not affect long-term outcomes. The most significant predictors for poor outcomes were WFNS score and age, while age and a history of vascular diseases were protective against rupture. Despite the high prevalence of modifiable risk factors, they did not significantly influence rupture risk. Conclusions: The findings suggest a need for multifactorial risk assessment strategies in managing IA patients. Future studies with larger cohorts are required to confirm these results and better understand IA progression. Full article
(This article belongs to the Section Neurology)
Show Figures

Figure 1

Figure 1
<p>Presentation of the patient cohort according to the inclusion criteria of the study. Of 303 patients with IA, 226 had complete information on the lifestyle-associated risk factors recorded. Information on rupture status was available for 225 patients and on clinical outcomes for 221.</p>
Full article ">Figure 2
<p>Part (<b>A</b>) of the figure shows the distribution of the number of risk factors for the rupture and outcome analyses, with the rupture analysis shown in dark blue and the outcome analysis in light blue. Part (<b>B</b>) of the figure schematically shows the concept of “MARVIN”, which stands for “Metabolic and Adverse Risk Factors and Vices influencing Intracranial aneurysms” and shows the accumulation of the established seven risk factors in patients who lead an unhealthy lifestyle.</p>
Full article ">Figure 3
<p>This figure illustrates the associations between various lifestyle-associated risk factors and hypertension, tested using the χ<sup>2</sup> test: (<b>A</b>) shows the association between hypertension and nicotine abuse, (<b>B</b>) between hypercholesterolemia and hypertension, (<b>C</b>) between obesity and hypertension, (<b>D</b>) between a history of vascular diseases and hypertension, and (<b>E</b>) between diabetes and hypertension.</p>
Full article ">Figure 4
<p>This figure illustrates the associations tested using the χ<sup>2</sup> test between (<b>A</b>) alcohol and nicotine abuse, (<b>B</b>) diabetes and obesity, and (<b>C</b>) history of vascular diseases and hypercholesterolemia.</p>
Full article ">Figure 5
<p>This figure illustrates the associations between age and various risk factors examined using a <span class="html-italic">t</span>-test. Part (<b>A</b>) depicts the association between older age and the increased likelihood of hypertension, diabetes, and a history of vascular diseases. Part (<b>B</b>) shows the association between younger age and a higher prevalence of nicotine abuse. Asterisks denote the level of statistical significance: * = <span class="html-italic">p</span> &lt; 0.05, ** = <span class="html-italic">p</span> &lt; 0.01, and *** = <span class="html-italic">p</span> &lt; 0.001. Blue dots represent the mean values.</p>
Full article ">Figure 6
<p>This figure illustrates the association between nicotine abuse and clinical outcomes in patients with ruptured IA, examined by a <span class="html-italic">t</span>-test. Part (<b>A</b>) shows that smokers presented with significantly higher WFNS scores at admission, indicating more severe initial clinical conditions. However, Part (<b>B</b>) demonstrates that nicotine abuse did not associate with a worse outcome at discharge, despite the more severe initial presentation. Asterisks denote the level of statistical significance: * = <span class="html-italic">p</span> &lt; 0.05. Blue dots represent the mean values.</p>
Full article ">Figure 7
<p>This figure illustrated the prediction of clinical outcome trichotomized in favorable outcome, disability, or death (proportional odds logistic regression). Here, the only significant predictors for a worse outcome were the WFNS score and age. The two are combined into a composite score, as given by the regression model. The x-axis represents the composite score, which is calculated using the formula provided below the axis.</p>
Full article ">Figure 8
<p>The prediction analysis of rupture, depicted in this figure, showed protective effects of higher age and history of vascular diseases against rupture (logistic regression). Pr(rupture) represents the conditional probability of rupture, based on the patient’s age and history of vascular diseases.</p>
Full article ">
25 pages, 3677 KiB  
Article
Determination of Recovery by Total Restitution or Compensation Using Multifrequency Vestibular Tests and Subjective Functional Scales in a Human Model of Vestibular Neuritis
by Enrico Armato, Georges Dumas, Flavio Perottino, Matthieu Casteran and Philippe Perrin
Audiol. Res. 2024, 14(6), 958-982; https://doi.org/10.3390/audiolres14060080 - 4 Nov 2024
Viewed by 514
Abstract
Background: Vestibular Neuritis (VN) can induce unilateral acute vestibular syndrome (AVS). This study aimed to identify predictive factors of recovery from vestibular neuritis considering total restitution and/or compensation. Methods: In this longitudinal study, 40 patients were included. The initial assessment, performed within 36 [...] Read more.
Background: Vestibular Neuritis (VN) can induce unilateral acute vestibular syndrome (AVS). This study aimed to identify predictive factors of recovery from vestibular neuritis considering total restitution and/or compensation. Methods: In this longitudinal study, 40 patients were included. The initial assessment, performed within 36 to 72 h from the onset (T0), included medical history taking (general and specific), including screening for cardiovascular risk factors (CVRFs), and a battery of diagnostic vestibular tests, comprising the bithermal caloric test (BCT), video head impulse test (VHIT), and skull vibration-induced nystagmus (SVIN) test. All patients also completed a Dizziness Handicap Inventory (DHI). All assessments were repeated 90 ± 15 days later (T3). Subjective compensation criteria were based on the DHI total score, and objective compensation criteria were based on laboratory test results. Four groups of patients (A, B, C, D) were delineated by combining patients with normal vs. abnormal vestibular tests and patients with normal vs. abnormal DHI. Results: CVRFs (but not age or body mass index (BMI)) were associated with a poorer recovery of symptoms. The BCT (lateral semicircular canal paresis %), VHIT (lateral semicircular canal gain), and SVINT (nystagmus slow phase velocity) recovered to normal values in 20%, 20%, and 27% of patients, respectively, at T3. Conclusions: Vascular risk factors (hypercholesterolemia) are correlated with patients who do not recover their symptoms via either total restitution or compensation. There was no significant difference between high- and low-frequency vestibular tests in patients recovering from their symptoms. Some patients with objective recovery may continue to have persistent subjective symptoms. Full article
Show Figures

Figure 1

Figure 1
<p>Flow chart diagram according to the PRISMA 2020 guidelines. AUVP: acute unilateral vestibulopathy.</p>
Full article ">Figure 2
<p>The currently known frequency spectrum of the vestibular system. Graph summarizing the complementarity of vestibular tests, sorted by stimulation frequency. The bithermal caloric test (BCT) is representative of very low frequencies, the video head impulse test (VHIT) of high frequencies, and the skull vibration-induced nystagmus test (SVINT) of very high frequencies (VLFs: very low frequencies; LFs: low frequencies; MFs: middle frequencies; HFs: high frequencies; VHFs: very high frequencies; Calo: caloric test; Sweep: multifrequency sinusoidal test; DVAP: Dynamic Visual Acuity Performance; HST: head shaking test; VAT: vestibular autorotation test; HIT: head impulse test; EMG: electromyography; oVEMP: ocular vestibular evoked myogenic potentials; cVEMP: cervical vestibular evoked myogenic potentials) [<a href="#B44-audiolres-14-00080" class="html-bibr">44</a>,<a href="#B45-audiolres-14-00080" class="html-bibr">45</a>,<a href="#B46-audiolres-14-00080" class="html-bibr">46</a>].</p>
Full article ">Figure 3
<p>Graphical representation by means and standard deviations for the 40-patients group at T0 (black) and T3—after 3 months (grey) for (<b>a</b>) Dizziness Handicap Inventory (y-axis = total score); (<b>b</b>) bithermal caloric test (y-axis = percentage of canal paresis); (<b>c</b>) skull vibration-induced nystagmus test (y-axis = nystagmus slow phase velocity); (<b>d</b>) anterior semicircular canal video head impulse test (y-axis = video head impulse test gain); (<b>e</b>) lateral semicircular canal video head impulse test (y-axis = video head impulse test gain); and (<b>f</b>) the head shaking test (y-axis = nystagmus slow phase velocity). The dashed lines represent the boundary between normal and pathological values. The red arrow represents the extent of pathological values, and the green arrow represents the extent of normal values. Dizziness Handicap Inventory (DHI), bithermal caloric test (BCT), skull vibration-induced nystagmus test (SVINT), head impulse test (VHIT), head shaking test (HST).</p>
Full article ">Figure 4
<p>Bithermal caloric test: Lateral semicircular canal paresis percentage (y-axis = percentage of canal paresis) at T0 and T3. (<b>a</b>) A + D (patients without symptoms at T3, red line) and B + C (patients with symptoms at T3, blue line) groups; (<b>b</b>) A + B (patients with no vestibular laboratory test dysfunction at T3, orange line), and C + D (patients with vestibular laboratory test dysfunction at T3, green line) groups. The last result suggests that when the initial BCT at T0 shows a hypofunction &gt; 90%, the recovery of caloric function is very poor, and the lesion is permanent. Conversely, for a BTC hypofunction &lt; 80% at T0, recovery is possible at T3 and can be complete in some cases. Significance is expressed as follows: * = <span class="html-italic">p</span> &lt; 0.05; ** = <span class="html-italic">p</span> &lt; 0.01; *** = <span class="html-italic">p</span> &lt; 0.001; and ns = <span class="html-italic">p</span> &gt; 0.05.</p>
Full article ">Figure 5
<p>Video head impulse test gain averages for left and right vestibular neuritis at T0 and T3 (y-axis = video head impulse test gain) for the A + D (patients without symptoms, red line) and B + C (patients with symptoms, blue line) groups. (<b>a</b>) lateral semicircular canal and (<b>b</b>) anterior semicircular canal. Significance is expressed as follows: * = <span class="html-italic">p</span> &lt; 0.05; ** = <span class="html-italic">p</span> &lt; 0.01; and ns = <span class="html-italic">p</span> &gt; 0.05.</p>
Full article ">Figure 6
<p>Evolution of video head impulse test gain averages (y-axis = video head impulse test gain) on the affected side for left and right vestibular neuritis at T0 and T3 for the A + B (patients with no vestibular laboratory test dysfunction, orange plot) and C + D (patients with vestibular laboratory test dysfunction, green plot) groups. (<b>a</b>) lateral semicircular canal and (<b>b</b>) anterior semicircular canal. Significance is expressed as follows:; *** = <span class="html-italic">p</span> &lt; 0.001; and ns = <span class="html-italic">p</span> &gt; 0.05.</p>
Full article ">Figure 7
<p>Skull vibration-induced nystagmus slow phase velocity averages (y-axis = skull vibration-induced nystagmus slow phase velocity) for the right and left mastoids at T0 and T3 for the A + D (patients without symptoms, red line) and B + C (patients with symptoms, blue line) groups. Significance is expressed as follows: * = <span class="html-italic">p</span> &lt; 0.05; *** = <span class="html-italic">p</span> &lt; 0.001; and ns = <span class="html-italic">p</span> &gt; 0.05.</p>
Full article ">Figure 8
<p>Comparison of skull vibration-induced nystagmus slow phase velocity averages (y-axis = skull vibration-induced nystagmus slow phase velocity) for the right and left mastoids at T0 and T3 for the A + B (patients with no vestibular laboratory test dysfunction, orange line) and C + D groups (patients with vestibular laboratory test dysfunction, green line) groups. Significance is expressed as follows; *** = <span class="html-italic">p</span> &lt; 0.001; and ns = <span class="html-italic">p</span> &gt; 0.05.</p>
Full article ">Figure 9
<p>Vestibular neuritis evolution diagram. On the left: The onset of vestibular neuritis is parameterized through Dizziness Handicap Inventory (DHI), spontaneous nystagmus (Ny), bithermal caloric test (BCT), video head impulse test (VHIT), and skull vibration induced nystagmus test (SVINT) (tests are ordered by labyrinthine analysis frequency from lowest to highest). In the center: Development of vestibular neuritis from the normalization of all tests with normal (A group) or pathological (B group) DHI to the persistence of pathological tests with ineffective objective laboratory test compensation parameters and pathological DHI (C group) or pathological tests with effective objective laboratory test compensation parameters and normal DHI (D group). On the right: Symptom profile of the various evolutive groups, i.e., asymptomatic (A group), chronic dizziness (B and C groups), and pauci- or asymptomatic (D group). Slow phase velocity (SPV), vestibular oculomotor reflex (VOR), corrective saccades (CS), vibration induced nystagmus (VIN).</p>
Full article ">
7 pages, 411 KiB  
Brief Report
Nonalcoholic Fatty Liver Disease Risk and Proprotein Convertase Subtilisin Kexin 9 in Familial Hypercholesterolemia Under Statin Treatment
by Masato Hamasaki, Naoki Sakane and Kazuhiko Kotani
Nutrients 2024, 16(21), 3686; https://doi.org/10.3390/nu16213686 - 29 Oct 2024
Viewed by 596
Abstract
Background/Objectives: Fatty acids are involved in some hepatic disorders. The proprotein convertase subtilisin kexin 9 (PCSK9) inhibits the uptake of low-density lipoproteins (LDLs), which contain lipids, into the liver and may thus be associated with nonalcoholic fatty liver disease (NAFLD), a cardiovascular disorder [...] Read more.
Background/Objectives: Fatty acids are involved in some hepatic disorders. The proprotein convertase subtilisin kexin 9 (PCSK9) inhibits the uptake of low-density lipoproteins (LDLs), which contain lipids, into the liver and may thus be associated with nonalcoholic fatty liver disease (NAFLD), a cardiovascular disorder (CVD) risk. Statins reduce blood LDL–cholesterol (LDL-C) levels and CVD risk and can attenuate the development of NAFLD while increasing blood PCSK9 levels. Methods: We investigated the correlation between PCSK9 and liver conditions in patients with familial hypercholesterolemia (FH), a CVD risk population with elevated blood LDL-C levels, under statin treatment. Blood tests for lipids, PCSK9, and liver function (aspartate aminotransferase [AST] and alanine aminotransferase [ALT]) were performed in patients with FH taking statins (n = 25, mean age = 57 years, 12% of males). The ALT:AST ratio was used as a marker of NAFLD risk. Results: The mean LDL-C level was 3.38 mmol/L, and the median PCSK9 level was 312 ng/mL. The median ALT:AST ratio was 0.88. A significant negative correlation was observed between the PCSK9 and ALT:AST ratio (β = −0.67, p < 0.05). Conclusions: Their negative correlation might give a hypothetical insight into the effect of statin treatment on the development of NAFLD, in relation to PCSK9 behavior, in patients with FH. Full article
Show Figures

Figure 1

Figure 1
<p>Correlation between the PCSK9 and ALT:AST ratio in patients with FH under statin treatment. ALT, alanine aminotransferase; AST, aspartate aminotransferase; PCSK9, proprotein convertase subtilisin/kexin type 9. The PCSK9 levels and ALT:AST ratio were log-transformed. Pearson’s correlation coefficient (<span class="html-italic">r</span>) was = −0.45 (<span class="html-italic">p</span> = 0.02).</p>
Full article ">
18 pages, 918 KiB  
Review
Bempedoic Acid, the First-in-Class Oral ATP Citrate Lyase Inhibitor with Hypocholesterolemic Activity: Clinical Pharmacology and Drug–Drug Interactions
by Nicola Ferri, Elisa Colombo and Alberto Corsini
Pharmaceutics 2024, 16(11), 1371; https://doi.org/10.3390/pharmaceutics16111371 - 26 Oct 2024
Viewed by 533
Abstract
Bempedoic acid is a new drug that improves the control of cholesterol levels, either as monotherapy or in combination with existing lipid-lowering therapies, and shows clinical efficacy in cardiovascular disease patients. Thus, patients with comorbidities and under multiple therapies may be eligible for [...] Read more.
Bempedoic acid is a new drug that improves the control of cholesterol levels, either as monotherapy or in combination with existing lipid-lowering therapies, and shows clinical efficacy in cardiovascular disease patients. Thus, patients with comorbidities and under multiple therapies may be eligible for bempedoic acid, thus facing the potential problem of drug–drug interactions (DDIs). Bempedoic acid is a prodrug administered orally at a fixed daily dose of 180 mg. The dicarboxylic acid is enzymatically activated by conjugation with coenzyme A (CoA) to form the pharmacologically active thioester (bempedoic acid–CoA). This process is catalyzed by very-long-chain acyl-CoA synthetase 1 (ACSVL1), expressed almost exclusively at the hepatic level. Bempedoic acid–CoA is a potent and selective inhibitor of ATP citrate lyase (ACL), a key enzyme in the biosynthetic pathway of cholesterol and fatty acids. The drug reduces low-density lipoprotein–cholesterol (LDL-C) (20–25%), non-high-density lipoprotein–cholesterol (HDL-C) (19%), apolipoprotein B (apoB) (15%), and total cholesterol (16%) in patients with hypercholesterolemia or mixed dyslipidemia. The drug has a favorable pharmacokinetics profile. Bempedoic acid and its metabolites are not substrates or inhibitors/inducers of cytochrome P450 (CYP450) involved in drug metabolism. On the other hand, bempedoic acid–glucuronide is a substrate for organic anion transporter 3 (OAT3). Bempedoic acid and its glucuronide are weak inhibitors of the OAT2, OAT3, and organic anion-transporting polypeptide 1B1 (OATP1B1) and 1B3 (OATP1B3). Thus, bempedoic acid could inhibit (perpetrator) the hepatic uptake of OATP1B1/3 substrate drugs and the renal elimination of OAT2 and OAT3 substrates and could suffer (victim) the effect of OAT3 transporter inhibitors, reducing its renal elimination. Based on these pharmacological characteristics, here, we describe the potential DDIs of bempedoic acid with concomitant medications and the possible clinical implications. Full article
(This article belongs to the Section Pharmacokinetics and Pharmacodynamics)
Show Figures

Figure 1

Figure 1
<p>Schematic representation of the mechanism of action of bempedoic acid compared to statins, ezetimibe, and anti-PCSK9 therapies. Statins, by inhibiting the hydroxy-methyl-glutaryl–CoA (HMG-CoA) reductase, reduce cholesterol biosynthesis in the liver, determining the activation of the transcription factor SREBP2, which drives the expression of the LDL receptor. Statins also induce the expression of PCSK9, which can degrade the LDL receptor and thus partially reduce the hypocholesterolemic effect of statins. Monoclonal antibodies (evolocumab and alirocumab) bind and inhibit PCSK9, while inclisiran reduces its synthesis, interfering with its mRNA. Bempedoic acid acts by inhibiting the ACLY, and thus reduces cholesterol biosynthesis, which activates a similar cellular response to that observed with statins. Finally, ezetimibe interacts with the intestinal cholesterol transporter NPC1L1, reducing its absorption from the diet.</p>
Full article ">Figure 2
<p>Main metabolites of bempedoic acid. Bempedoic acid and its keto metabolite ECT-15228 are both substrates of the UGT2B7 enzyme, which forms the two glucuronide derivatives. The enzyme ACSVL1 also converted the bempedoic acid and ECT-15228 into their active metabolites. UGT-2B7: UDP-Glucuronosyltransferase-2B7; ACSVL1: very-long-chain acyl–CoA synthetase 1. Bempedoic acid–CoA and ETC-15228–CoA are the active metabolites formed selectively in the liver.</p>
Full article ">
11 pages, 2246 KiB  
Article
Phenotype in Individuals with Heterozygous Rare Variants in LIPC Encoding Hepatic Lipase
by Erin O. Jacob, Jian Wang, Adam D. McIntyre and Robert A. Hegele
Int. J. Mol. Sci. 2024, 25(21), 11445; https://doi.org/10.3390/ijms252111445 - 24 Oct 2024
Viewed by 546
Abstract
Hepatic lipase deficiency is a rare genetic condition caused by biallelic loss-of-function variants in the LIPC gene encoding hepatic lipase. These variants reduce or abolish the protein’s lipolytic activity, resulting in elevated plasma lipids. The condition is classified as autosomal recessive, since dyslipidemia [...] Read more.
Hepatic lipase deficiency is a rare genetic condition caused by biallelic loss-of-function variants in the LIPC gene encoding hepatic lipase. These variants reduce or abolish the protein’s lipolytic activity, resulting in elevated plasma lipids. The condition is classified as autosomal recessive, since dyslipidemia is inconsistently observed in heterozygous patients with only one LIPC variant. However, this has been concluded from historical studies encompassing a few families and having very small sample sizes. Here, we conduct a retrospective chart review of 46 heterozygous subjects, each harboring one rare pathogenic LIPC variant. We compare plasma levels of total cholesterol, low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), triglycerides, and apolipoprotein B to those of matched controls without LIPC variants. Variant pathogenicity is classified according to the guidelines of the American College of Medical Genetics and Genomics. We observe that levels of total cholesterol, LDL-C, and triglycerides are significantly elevated in the LIPC variant heterozygotes, but HDL-C and apolipoprotein B are not. When filtering solely with respect to pathogenic or likely pathogenic variants, all lipid variables emerge as significantly elevated compared to controls. Thus, hepatic lipase deficiency may not necessarily be a recessive condition, but perhaps semi-dominant since individuals with one variant are phenotypically distinct from the controls. These hypothesis-generating findings regarding LIPC genetic variations observed in a clinical cohort should be evaluated in larger populations and databases. Full article
(This article belongs to the Special Issue Apolipoproteins and Lipoproteins in Health and Disease, 3rd Edition)
Show Figures

Figure 1

Figure 1
<p>Lipid and lipoprotein levels of patients with <span class="html-italic">LIPC</span> variants compared to population controls. The plasma levels of total cholesterol (TC) (<b>A</b>), low-density lipoprotein cholesterol (LDL-C) (<b>B</b>), high-density lipoprotein cholesterol (HDL-C) (<b>C</b>), and triglycerides (TG) (<b>D</b>) were measured in patients with <span class="html-italic">LIPC</span> variants. Each patient’s levels were converted to percentile values, with the average percentile of the cohort displayed above (vertical line). The reference percentile values were obtained from the LRC prevalence study (North America). The distributions were formed using data from 50–55-year-old women, corresponding to the average age and sex of the cohort.</p>
Full article ">Figure 2
<p>Lipid levels of patients with <span class="html-italic">LIPC</span> variants compared to normal controls. Plasma levels of total cholesterol (TC) (green), low-density lipoprotein cholesterol (LDL-C) (purple), high-density lipoprotein cholesterol (HDL-C) (orange), triglycerides (log(TG)) (blue), and apolipoprotein (apo) B (pink) were compared between patients with <span class="html-italic">LIPC</span> variants (<span class="html-italic">n</span> = 46) and normal controls (<span class="html-italic">n</span> = 72). The darker color corresponds to the <span class="html-italic">LIPC</span> cohort, the lighter to the control. Unpaired one-tailed <span class="html-italic">t</span>-tests were used to compare the lipid levels between cohorts: ‘*’ denotes a <span class="html-italic">p</span>-value &lt; 0.05 and ‘**’ denotes a <span class="html-italic">p</span>-value &lt; 0.0001. The one-tailed <span class="html-italic">p</span>-value between <span class="html-italic">LIPC</span> and control groups is 1.5 × 10<sup>−6</sup> for TC, 0.019 for LDL-C, 0.11 for HDL-C, 5.4 × 10<sup>−9</sup> for log(TG), and 0.20 for Apo B.</p>
Full article ">Figure 3
<p>Lipid levels of patients with pathogenic <span class="html-italic">LIPC</span> variants compared to normal controls. Plasma levels of total cholesterol (TC) (green), low-density lipoprotein cholesterol (LDL-C) (purple), high-density lipoprotein cholesterol (HDL-C) (orange), triglycerides (log(TG)) (blue), and apolipoprotein B (Apo B) (pink) were compared among patients with pathogenic or likely pathogenic <span class="html-italic">LIPC</span> variants (<span class="html-italic">n</span> = 13) and normal controls (<span class="html-italic">n</span> = 72). The darker color corresponds to the <span class="html-italic">LIPC</span> cohort, the lighter to the control. Unpaired one-tailed <span class="html-italic">t</span>-tests were used to compare the lipid levels between cohorts, ‘*’ denotes a <span class="html-italic">p</span>-value &lt; 0.05 and ‘**’ denotes a <span class="html-italic">p</span>-value &lt; 0.0001: TC <span class="html-italic">p</span> = 5.7 × 10<sup>−5</sup>, LDL-C <span class="html-italic">p</span> = 0.011, HDL-C <span class="html-italic">p</span> = 0.039, log(TG) <span class="html-italic">p</span> = 5.9 × 10<sup>−3</sup>, and Apo B <span class="html-italic">p</span> = 0.005.</p>
Full article ">Figure 4
<p>Lipid levels of patients with benign <span class="html-italic">LIPC</span> variants compared to normal controls. Plasma levels of total cholesterol (TC) (green), low-density lipoprotein cholesterol (LDL-C) (purple), high-density lipoprotein cholesterol (HDL-C) (orange), triglycerides (log(TG)) (blue), and apolipoprotein B (Apo B) (pink) were compared among patients with benign or likely benign <span class="html-italic">LIPC</span> variants (<span class="html-italic">n</span> = 4) and normal controls (<span class="html-italic">n</span> = 72). The darker color corresponds to the <span class="html-italic">LIPC</span> cohort, the lighter to the control. Unpaired one-tailed <span class="html-italic">t</span>-tests were used to compare the lipid levels between cohorts, ‘*’ denotes a <span class="html-italic">p</span>-value &lt; 0.05. The one-tailed <span class="html-italic">p</span>-value between the <span class="html-italic">LIPC</span> group and the control group is 0.159 for TC, 0.313 for LDL-C, 0.385 for HDL-C, 0.048 for log(TG), and 0.25 for Apo B.</p>
Full article ">Figure 5
<p>Lipid levels of patients with AI-predicted pathogenic <span class="html-italic">LIPC</span> variants compared to normal controls. Plasma levels of total cholesterol (TC) (green), low-density lipoprotein cholesterol (LDL-C) (purple), high-density lipoprotein cholesterol (HDL-C) (orange), triglycerides (log(TG)) (blue), and apolipoprotein B (Apo B) (pink) were compared among patients with pathogenic or likely pathogenic <span class="html-italic">LIPC</span> variants (<span class="html-italic">n</span> = 10) and normal controls (<span class="html-italic">n</span> = 72). The pathogenic classification was obtained using the AI predictive software VARITY (v1.0). All samples scored 0.823 or higher, with scores closer to 1.00 indicating a higher likelihood of pathogenicity. The darker color corresponds to the <span class="html-italic">LIPC</span> cohort, the lighter to the control. Unpaired one-tailed <span class="html-italic">t</span>-tests were used to compare the lipid levels between cohorts, ‘*’ denotes a <span class="html-italic">p</span>-value &lt; 0.05: TC <span class="html-italic">p</span> = 1.9 × 10<sup>−3</sup>, LDL-C <span class="html-italic">p</span> = 0.061, HDL-C <span class="html-italic">p</span> = 0.075, log(TG) <span class="html-italic">p</span> = 6.0 × 10<sup>−3</sup>, and Apo B <span class="html-italic">p</span> = 0.31.</p>
Full article ">Figure 6
<p>Lipid levels of patients with <span class="html-italic">LIPC</span> and familial hypercholesterolemia variants. Plasma lipid levels were compared among patients with both <span class="html-italic">LIPC</span> and familial hypercholesterolemia (FH) variants (<span class="html-italic">n</span> = 7) (purple) and patients with only <span class="html-italic">LIPC</span> variants (<span class="html-italic">n</span> = 46) (pink) or only FH variants (<span class="html-italic">n</span> = 135) (blue). (<b>A</b>) Total cholesterol (TC). The <span class="html-italic">p</span>-value between the <span class="html-italic">LIPC</span> and FH + <span class="html-italic">LIPC</span> groups is <span class="html-italic">p</span> = 0.003. (<b>B</b>) Low-density lipoprotein cholesterol (LDL-C). The one-tailed <span class="html-italic">p</span>-value between <span class="html-italic">LIPC</span> and FH + <span class="html-italic">LIPC</span> is <span class="html-italic">p</span> = 0.002. (<b>C</b>) High-density lipoprotein cholesterol (HDL-C). (<b>D</b>) Log(triglycerides) (log(TG)). ‘*’ denotes a <span class="html-italic">p</span>-value &lt; 0.05. All other comparisons had <span class="html-italic">p</span>-values greater than 0.1.</p>
Full article ">
18 pages, 4142 KiB  
Article
Antarctic Krill Euphausia superba Oil Supplementation Attenuates Hypercholesterolemia, Fatty Liver, and Oxidative Stress in Diet-Induced Obese Mice
by Jun-Hui Choi, Se-Eun Park and Seung Kim
Nutrients 2024, 16(21), 3614; https://doi.org/10.3390/nu16213614 - 24 Oct 2024
Viewed by 881
Abstract
Background: Several Previous studies indicate that consuming krill oil may aid in reducing hypercholesterolemia and improving cholesterol metabolism. Therefore, our study was designed to investigate the effectiveness of Antarctic krill oil (Euphausia superba) (ESKO) in combating obesity and lowering fat/lipid/cholesterol levels. [...] Read more.
Background: Several Previous studies indicate that consuming krill oil may aid in reducing hypercholesterolemia and improving cholesterol metabolism. Therefore, our study was designed to investigate the effectiveness of Antarctic krill oil (Euphausia superba) (ESKO) in combating obesity and lowering fat/lipid/cholesterol levels. Methods: The study aimed to investigate the molecular docking model targeting 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR) using ESKO-derived eicosapentaenoic acid (EPA), docosahexaenoic acid (DHA), and astaxanthin. In this study, histological alterations in the liver of the obesity model (ICR male mouse), obesity-related or antioxidant markers in both liver and serum, the molecular mechanisms in HepG2 cells and liver tissue, and HMGCR activity were analyzed. Results: Our findings revealed that a high-fat diet (HFD) significantly led to increased oxidative stress, obesity-related indicators, and cardiovascular-associated risk indices. However, ESKO effectively mitigated HFD-induced oxidative stress, fat accumulation, and the suppression of low-density lipoprotein receptor (LDLR) or activation of related molecular pathways. This was achieved through improvements in metabolic parameters, including CD36/liver X receptor α (LXRα)/sterol regulatory element-binding protein 1c (SREBP1c), proprotein convertase subtilsin/kexin type 9 (PCSK-9), and HMGCR, ultimately ameliorating HFD-induced hypercholesterolemia and obesity. Conclusions: These beneficial findings indicate that ESKO might have significant potential for preventing and treating obesity-related disorders. Full article
(This article belongs to the Special Issue Nutrition and Dietary Intake in Liver-Related Diseases)
Show Figures

Figure 1

Figure 1
<p>Effect of ESKO on cell viability (<b>A</b>,<b>B</b>), lipid (<b>C</b>), triglyceride (<b>D</b>) level, and HMGCR activity (<b>E</b>) in HepG2 cells. MTT assay showing the effect of ESKO and palmitic acid on HepG2 cells viability. Cells were incubated with ESKO at different concentrations (0–1000 μg/mL) or palmitic acid (0–1000 μM) for 24 h and cell viability was analyzed by MTT reduction assay. Each value is the mean ± SD of triplicate measurements. # <span class="html-italic">p</span> &lt; 0.01, compared with Control group, * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01, compared with palmitic acid only-treated group. ESKO, <span class="html-italic">Euphausia superba</span> krill oil; HMGCR, 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR).</p>
Full article ">Figure 2
<p>Effect of ESKO on CD36 (<b>A</b>,<b>B</b>), LXRα (<b>C</b>), SREBP1C (<b>D</b>), LDLR (<b>E</b>), and PCSK9 (<b>F</b>) signaling and HMGCR activity (<b>G</b>) in HepG2 cells. Each value is the mean ± SD of triplicate measurements. # <span class="html-italic">p</span> &lt; 0.01, compared with Control group, * <span class="html-italic">p</span> &lt; 0.01, compared with palmitic acid only-treated group as negative control (NC). PA, palmitic acid; ESKO, <span class="html-italic">Euphausia superba</span> krill oil; LXRα, liver X receptor α; SREBP1c, sterol regulatory element-binding protein 1c; LDLR, low-density lipoprotein receptor; PCSK9, proprotein convertase subtilsin/kexin type 9; HMGCR, 3-hydroxy-3-methylglutaryl-CoA reductase.</p>
Full article ">Figure 3
<p>Effect of ESKO on body weight (<b>A</b>,<b>C</b>,<b>D</b>) and feed intake (<b>B</b>,<b>E</b>) in DIO mouse models. Each value is the mean ± SD (<span class="html-italic">n</span> = 6). # <span class="html-italic">p</span> &lt; 0.05 and ## <span class="html-italic">p</span> &lt; 0.01, compared with Control group. Control, non-treated normal group; Model, diet-induced obesity (DIO) model group; Feno, 200 mg/kg fenofibrate-administrated DIO group, ESKO, 400 mg/kg <span class="html-italic">Euphausia superba</span> krill oil-administrated DIO group.</p>
Full article ">Figure 4
<p>Histopathological analysis of livers and epididymal fats from DIO models. Effects of ESKO and fenofibrate on hepatic (<b>A</b>,<b>B</b>) or epididymal fats (<b>C</b>,<b>D</b>) in DIO model was analyzed with H&amp;E, microscope, ImageJ, or fat weight. Each value is the mean ± SD (<span class="html-italic">n</span> = 6). # <span class="html-italic">p</span> &lt; 0.01, compared with Control group, * <span class="html-italic">p</span> &lt; 0.01, compared with DIO model group. Control, non-treated normal group; Model, diet-induced obesity (DIO) model group; Feno, 200 mg/kg fenofibrate-administrated DIO group, ESKO, 400 mg/kg <span class="html-italic">Euphausia superba</span> krill oil-administrated DIO group.</p>
Full article ">Figure 5
<p>Effect of ESKO on CD36 (<b>A</b>,<b>B</b>), LXRα (<b>C</b>), SREBP1C (<b>D</b>), LDLR (<b>E</b>), and PCSK9 (<b>F</b>) signaling and HMGCR activity (<b>G</b>) in livers from DIO model. Each value is the mean ± SD (<span class="html-italic">n</span> = 6). # <span class="html-italic">p</span> &lt; 0.01, compared with Control group, * <span class="html-italic">p</span> &lt; 0.01, compared with DIO Model group. Control, non-treated normal group; Model, diet-induced obesity (DIO) Model group; Feno, 200 mg/kg fenofibrate-administrated DIO group, ESKO, 400 mg/kg <span class="html-italic">Euphausia superba</span> krill oil-administrated DIO group. LXRα, liver X receptor α; SREBP1c, sterol regulatory element-binding protein 1c; LDLR, low-density lipoprotein receptor; PCSK9, proprotein convertase subtilsin/kexin type 9; HMGCR, 3-hydroxy-3-methylglutaryl-CoA reductase.</p>
Full article ">
14 pages, 3545 KiB  
Article
Lactobacillus Kefir M20 Adaptation to Bile Salts: A Novel Pathway for Cholesterol Reduction
by Changlu Ma, Qichen Liu, Shuwen Zhang, Ailing Qu, Qing Liu, Jiaping Lv and Xiaoyang Pang
Foods 2024, 13(21), 3380; https://doi.org/10.3390/foods13213380 - 24 Oct 2024
Viewed by 591
Abstract
(1) Background: This study investigated the impact of in vitro adaptations to acid and bile stress on the cholesterol-lowering activity of the probiotic Lactobacillus kefir M20. (2) Methods: Lactobacillus kefir M20 was extracted from fermented dairy products in Xinjiang, China, and isolated using [...] Read more.
(1) Background: This study investigated the impact of in vitro adaptations to acid and bile stress on the cholesterol-lowering activity of the probiotic Lactobacillus kefir M20. (2) Methods: Lactobacillus kefir M20 was extracted from fermented dairy products in Xinjiang, China, and isolated using MRS medium. The lactic acid bacteria were cultured for stress resistance to acid and bile salts and then gavaged into mice for animal experiments. (3) Results: The adaptation to bile stress treatment resulted in a notable enhancement of the cholesterol-lowering capacity of Lactobacillus kefir M20, with reductions of 16.5% and 33.1% in total and non-HDL cholesterol, respectively, compared to the untreated strain. Furthermore, the daily fecal total bile acid excretion was 9.2, 5.4 and 5.0 times higher in the M20-BSA group compared to the HC, M20 and M20-ASA groups, respectively. (4) Conclusions: This study suggests that targeted probiotics have the potential for application in the next generation of functional foods and probiotic formulations aimed at combating hypercholesterolemia. Full article
Show Figures

Figure 1

Figure 1
<p>Effects of the acid and bile stress adaption treatments on acid tolerance (<b>A</b>), bile tolerance (<b>B</b>) and adhesion ability (<b>C</b>) of the strain. Data are expressed as the mean ± SD (<span class="html-italic">n</span> = 3). Means not sharing a common letter are significantly (<span class="html-italic">p</span> &lt; 0.05) different from each other.</p>
Full article ">Figure 2
<p>The effects of acid and bile stress acclimation treatments on bile salt hydrolase-specific activity (<b>A</b>) and BSH gene expression (<b>B</b>) of the strain are presented herewith. The data are presented as the mean ± standard deviation (SD) (<span class="html-italic">n</span> = 3). Significant differences were observed between means that did not share the same letter (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 3
<p>Effects of the acid and bile stress adaption treatments on compositions of membrane 14:0 (<b>A</b>), 16:0 (<b>B</b>), 16:1 (<b>C</b>), 18:0 (<b>D</b>), 18:1 (<b>E</b>), 18:2 (<b>F</b>), Δ19:0 (<b>G</b>) and other fatty acids (<b>H</b>) of the strain. Fatty acid methyl esters are designated by the number of carbon atoms to the left of the colon and the number of double bonds to the right. Δ, cyclopropane ring. Data are expressed as the mean ± SD (<span class="html-italic">n</span> = 3). Means not sharing a common letter are significantly (<span class="html-italic">p</span> &lt; 0.05) different from each other.</p>
Full article ">Figure 4
<p>Serum total cholesterol (<b>A</b>), HDL cholesterol (<b>B</b>) and non-HDL cholesterol (<b>C</b>) levels in different groups of hamsters. Abbreviation: HDL, high-density lipoprotein. Data are expressed as the mean ± SD (<span class="html-italic">n</span> = 8). Means not sharing a common letter are significantly (<span class="html-italic">p</span> &lt; 0.05) different from each other.</p>
Full article ">Figure 5
<p>Hepatic total cholesterol (<b>A</b>), free cholesterol (<b>B</b>), and esterified cholesterol (<b>C</b>) levels in different groups of hamsters. Data are expressed as the mean ± SD (<span class="html-italic">n</span> = 8). Means not sharing a common letter are significantly (<span class="html-italic">p</span> &lt; 0.05) different from each other.</p>
Full article ">Figure 6
<p>Daily fecal bile acid excretion levels in different groups of hamsters. Data are expressed as the mean ± SD (<span class="html-italic">n</span> = 8). Means not sharing a common letter are significantly (<span class="html-italic">p</span> &lt; 0.05) different from each other.</p>
Full article ">
18 pages, 3742 KiB  
Article
Comparative Study of Cutaneous Squamous Cell Carcinogenesis in Different Hairless Murine Models
by Georgios Gkikas, Dimitrios Katsiris, Andreas Vitsos, Anna Gioran, Dimitra Ieronymaki, Maria Kostaki, Georgios Ladopoulos, Vaya Ioannidou, Elisavet Theodoraki, Niki Chondrogianni, Ioannis Sfiniadakis, Georgios T. Papaioannou and Michail Christou Rallis
Cancers 2024, 16(20), 3546; https://doi.org/10.3390/cancers16203546 - 21 Oct 2024
Viewed by 656
Abstract
Background: In recent decades, a significant global increase in the incidence of non-melanoma skin cancer has been observed. To explore the pathogenesis of and potential therapeutic approaches for squamous cell carcinoma, various in vivo studies using mouse models have been conducted. However, [...] Read more.
Background: In recent decades, a significant global increase in the incidence of non-melanoma skin cancer has been observed. To explore the pathogenesis of and potential therapeutic approaches for squamous cell carcinoma, various in vivo studies using mouse models have been conducted. However, investigations comparing different hairless mouse models, with or without melanin, as well as models with hypercholesterolemia and immunosuppression, in terms of their ability to induce squamous cell carcinoma have yet to be undertaken. Methods: Four mouse strains, namely SKH-hr1, SKH-hr2, SKH-hr2+ApoE, and immunodeficient Nude (Foxn1 knockout), were exposed to UVA and UVB radiation three times per week, initially to 1 Minimal Erythemal Dose (MED), incrementally increased weekly to a maximum dose of 3 MED. Clinical evaluation, photodocumentation, and biophysical parameters were monitored, along with proteasome protein activity and histopathological assessments. Results: The SKH-hr1 model primarily developed actinic keratosis without significant progression to invasive squamous cell carcinoma (SCC), while the SKH-hr2 and SKH-hr2+ApoE models exhibited a higher likelihood and intensity of papilloma and aggressive SCC formation, with the latter showing upregulated proteasome activity. Histopathological analysis confirmed the presence of poorly differentiated, invasive SCCs in the SKH-hr2 and SKH-hr2+ApoE models, contrasting with the less aggressive SCCs in the Nude mice and the mixed lesions observed in the SKH-hr1 mice. Conclusions: The SKH-hr2+ApoE and SKH-hr2 mice were identified as the most suitable for further exploration of squamous cell carcinogenesis. In contrast, the SKH-hr1 mice were found to be the least suitable, even though they are albino. Notably, proteasome analysis revealed a potential role of proteasome activity in squamous cell carcinogenesis. Full article
(This article belongs to the Section Methods and Technologies Development)
Show Figures

Figure 1

Figure 1
<p>Evolution of carcinogenesis over time. Representative images of the four irradiated mouse models (SKH-hr1, SKH-hr2, SKH-hr2+ApoE, and Nude) at various time points. Differences are evident as early as the first month across all the groups, with increased melanin observed in the skin of the SKH-hr2 and SKH-hr2+ApoE models by the third month. Papilloma formation began after the third month, and tumor development was noted after the seventh month in the SKH-hr2, SKH-hr2+ApoE, and Nude models. In contrast, the SKH-hr1 mice primarily exhibited actinic keratosis toward the study’s end, with minimal papilloma and tumor formation.</p>
Full article ">Figure 2
<p>Cumulative counts of papillomas and tumors over time. (<b>A</b>) Cumulative number of papillomas per mouse strain. (<b>B</b>) Cumulative number of tumors per mouse strain. (<b>C</b>) Cumulative number of animals exhibiting carcinogenesis per mouse strain (<span class="html-italic">n</span> = 10 SKH-hr1, 10 SKH-hr2, 10 SKH-hr2+ApoE, and 9 Nude mice).</p>
Full article ">Figure 3
<p>Histopathological assessment of squamous cell carcinoma. (<b>A</b>) SCC exhibiting numerous keratin pearls (indicated by black arrows), extending from the epidermis into the dermis. (<b>B</b>) Aggressively, poorly differentiated SCC characterized by the presence of abundant keratin pearls (marked by black arrows). (<b>C</b>) Actinic keratosis is noted, alongside an evolving SCC in situ that remains non-invasive and has yet to breach the basement membrane. (<b>D</b>) Aggressive SCC displaying significant mitotic activity and the presence of enlarged, atypical nuclei. To the right of the image, a well-differentiated region of the SCC features numerous keratin pearls (black arrows), in contrast to the left side, which shows a poorly differentiated region of the SCC with cohesive cell clusters (blue arrows). Panels (<b>A</b>–<b>D</b>) correspond to Nude and SKH-hr2+ApoE, SKH-hr1, and SKH-hr2 mice, respectively. (Magnification: ×100).</p>
Full article ">Figure 4
<p>Graphical representation of the alterations in the skin parameters per month. (<b>A</b>) Transepidermal water loss. (<b>B</b>) Hydration. (<b>C</b>) Melanin. (<b>D</b>) Skin thickness. Each mouse model is depicted with a different color (blue for SKH-hr1, red for SKH-hr2, green for SKH-hr2+ApoE, orange for Nude).</p>
Full article ">Figure 5
<p>Proteasome status in the SKH-hr2 and SKH-hr2+ApoE mice: (<b>A</b>) chymotrypsin-like (CT-L), (<b>B</b>) caspase-like (C-L) and (<b>C</b>) trypsin-like (T-L) activities measured in normal (N) and tumorous (T) skin of hr2 (left) and hr2-ApoE (right) animals. Tissues from (<b>A</b>) 5 (N) -8 (T) hr2 animals and 9 (N) -11 (T) hr2-ApoE animals, (<b>B</b>) 6 (N) -8 (T) hr2 animals and 9 (N and T) hr2-ApoE animals, and (<b>C</b>) 3 (N) -4 (T) hr2 animals and 4 (N and T) hr2-ApoE animals were analyzed (paired <span class="html-italic">t</span>-test for (<b>A</b>,<b>B</b>), Wilcoxon test for (<b>C</b>), * <span class="html-italic">p</span> value &lt; 0.05, ns, non-significant). (<b>D</b>) Representative immunoblots and (<b>E</b>) Band densitometry for the β1 and β2 proteasome subunits, depicting the mean band density ± SEM. The braces indicate that the normal (N) and tumorous (T) tissue came from the same animal. The β1 and β2 protein levels were normalized to gels stained with Coomassie Brilliant Blue. Tissues from 5–8 animals per group were analyzed (unpaired <span class="html-italic">t</span>-test, ns, non-significant). The original Western blot figures can be found in <a href="#app1-cancers-16-03546" class="html-app">Supplemental Materials</a>.</p>
Full article ">Figure 6
<p>Sebum measurements across different murine models over time. While no statistically significant differences in the skin sebum levels were detected across the groups, the SKH-hr2+ApoE mice consistently exhibited higher average sebum concentrations compared to the other models.</p>
Full article ">
14 pages, 1526 KiB  
Article
GC-MS and PCA Analysis of Fatty Acid Profile in Various Ilex Species
by Anna Zwyrzykowska-Wodzińska, Bogdan Jarosz, Piotr Okińczyc, Jakub Szperlik, Przemysław Bąbelewski, Zdeněk Zadák, Anna Jankowska-Mąkosa and Damian Knecht
Molecules 2024, 29(20), 4833; https://doi.org/10.3390/molecules29204833 - 12 Oct 2024
Viewed by 561
Abstract
Natural compounds are important source of desired biological activity which helps to improve nutritional status and brings many health benefits. Ilex paraguariensis St. Hill. which belongs to the family Aquifoliaceae is a plant rich in bioactive substances (polyphenols, saponins, alkaloids) with therapeutic potential [...] Read more.
Natural compounds are important source of desired biological activity which helps to improve nutritional status and brings many health benefits. Ilex paraguariensis St. Hill. which belongs to the family Aquifoliaceae is a plant rich in bioactive substances (polyphenols, saponins, alkaloids) with therapeutic potential including hepatic and digestive disorders, arthritis, rheumatism, and other inflammatory diseases, obesity, hypertension, hypercholesterolemia. In terms of phytochemical research I. paraguariensis has been the subject of most intensive investigations among Ilex species. Therefore, we concentrated on other available Ilex varieties and focused on the content of fatty acids of these shrubs. The fatty acid compounds present in Ilex sp. samples were analyzed by GC-MS. 27 different fatty acids were identified in the extracts. The results showed that many constituents with significant commercial or medicinal importance were present in high concentrations. The primary component in all samples was α linolenic acid(18:3 Δ9,12,15). Differences of this component concentration were observed between cultivars and extensively analyzed by PCA, one- way ANOVA and Kruskal-Wallis ANOVA. Significant correlations between compound concentrations were reported. Full article
(This article belongs to the Special Issue Chemical and Biological Research on Bioactive Natural Products)
Show Figures

Figure 1

Figure 1
<p>Heat map of single component presence in fatty acid profiles of <span class="html-italic">Ilex</span> spp. leaves. US FA—sum of unsaturated fatty acid; SA FA—sum of saturated FA; %; FA—% of fatty acid in profile.</p>
Full article ">Figure 2
<p>Projection of the variable on the factor plane.</p>
Full article ">Figure 3
<p>Projection of the cases on the factor plane.</p>
Full article ">Figure 4
<p>Correlation matrix R<sup>2</sup> heat map.</p>
Full article ">
16 pages, 444 KiB  
Article
Cardiovascular Health and Diet Quality among Vegetarians, Vegans and Omnivores: Insights from a Large Urban Population in Poland
by Oliwia Grygorczuk, Martyna Mrozik, Anna Lipert, Sylwia Kamińska, Adam Białas, Wojciech Drygas, Ewa Rębowska, Stanisław Łęgocki, Anna Jegier, Katarzyna Szmigielska and Magdalena Kwaśniewska
Nutrients 2024, 16(20), 3438; https://doi.org/10.3390/nu16203438 - 10 Oct 2024
Viewed by 1773
Abstract
Background/Objectives: Dietary habits are among the most significant determinants of health. The aim of this study was to assess the nutritional quality and cardiovascular profiles of individuals following plant-based diet. Methods: The study population comprised 199 individuals (136 women, 63 men; mean age [...] Read more.
Background/Objectives: Dietary habits are among the most significant determinants of health. The aim of this study was to assess the nutritional quality and cardiovascular profiles of individuals following plant-based diet. Methods: The study population comprised 199 individuals (136 women, 63 men; mean age 33.9  ±  8.9 years) including vegans (VG; n = 50), vegetarians (VN; n = 101) and omnivores (OV; n = 48). In this analysis the following procedures were assessed: a questionnaire interview, anthropometric and blood pressure measurements, and a blood sample collection. Dietary patterns were evaluated using the Food Frequency Questionnaire and a 24-h dietary recall. Results: Vegans exhibited the lowest protein intake relative to the other groups (p < 0.05) and a significantly higher intake of polyunsaturated fatty acids and lower intake of cholesterol compared to VN and OV (p < 0.05). Vegans had significantly lower levels of serum cholesterol, LDL cholesterol, and triglycerides, fasting glucose and high-sensitivity C-reactive protein (p < 0.05). No cases of overweight or obesity were observed among VN and VG participants. No instances of impaired fasting glucose or elevated blood pressure were noted among vegans. Hypercholesterolemia was identified in 56.2% of OV, 26.7% in VN and 16.0% in VG (p < 0.05), elevated blood pressure was recorded in one vegetarian and in 6.2% of OV participants. Conclusions: Our research indicates that plant-based diets are associated with a better cardiovascular profile compared to traditional diets. Moreover, suboptimal intake of essential nutrients, underscores the need for more effective public health interventions and improved nutrition education regardless of dietary patterns. Full article
Show Figures

Figure 1

Figure 1
<p>Flow chart of participant recruitment.</p>
Full article ">
Back to TopTop