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

Journals

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 (495)

Search Parameters:
Keywords = trimethylamine

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
19 pages, 687 KiB  
Review
Exploring Trimethylaminuria: Genetics and Molecular Mechanisms, Epidemiology, and Emerging Therapeutic Strategies
by Antonina Sidoti, Rosalia D’Angelo, Andrea Castagnetti, Elisa Viciani, Concetta Scimone, Simona Alibrandi and Giuseppe Giannini
Biology 2024, 13(12), 961; https://doi.org/10.3390/biology13120961 - 22 Nov 2024
Viewed by 381
Abstract
Trimethylaminuria (TMAU) is a rare metabolic syndrome caused by the accumulation of trimethylamine in the body, causing odor emissions similar to rotten fish in affected patients. This condition is determined by both genetic and environmental factors, especially gut dysbiosis. The multifactorial nature of [...] Read more.
Trimethylaminuria (TMAU) is a rare metabolic syndrome caused by the accumulation of trimethylamine in the body, causing odor emissions similar to rotten fish in affected patients. This condition is determined by both genetic and environmental factors, especially gut dysbiosis. The multifactorial nature of this syndrome makes for a complex and multi-level diagnosis. To date, many aspects of this disease are still unclear. Recent research revealed the FMO3 haplotypes’ role on the enzyme’s catalytic activity. This could explain why patients showing only combined polymorphisms or heterozygous causative variants also manifest the TMAU phenotype. In addition, another research hypothesized that the behavioral disturbances showed by patients may be linked to gut microbiota alterations. Our review considers current knowledge about TMAU, clarifying its molecular aspects, the therapeutic approaches used to limit this condition, and the new therapies that are under study. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Catalytic cycle of FMO enzymes. This figure represents TMA oxidation by FMO enzymes, supported by NAD and FAD coenzymes.</p>
Full article ">
17 pages, 925 KiB  
Review
Microbiome-Derived Trimethylamine N-Oxide (TMAO) as a Multifaceted Biomarker in Cardiovascular Disease: Challenges and Opportunities
by Kinga Jaworska, Wojciech Kopacz, Mateusz Koper and Marcin Ufnal
Int. J. Mol. Sci. 2024, 25(23), 12511; https://doi.org/10.3390/ijms252312511 - 21 Nov 2024
Viewed by 440
Abstract
Biomarkers play a crucial role in various stages of disease management, including screening, diagnosis, prediction, prognosis, treatment, and safety monitoring. Although they are powerful tools in disease diagnosis, management, and drug development, identifying and validating reliable biomarkers remains a significant challenge. Among potential [...] Read more.
Biomarkers play a crucial role in various stages of disease management, including screening, diagnosis, prediction, prognosis, treatment, and safety monitoring. Although they are powerful tools in disease diagnosis, management, and drug development, identifying and validating reliable biomarkers remains a significant challenge. Among potential microbiome-derived biomarkers, trimethylamine N-oxide (TMAO) has gained notable attention for its link to atherosclerosis and cardiovascular risk. However, despite the growing body of research on TMAO, its practical application in clinical settings for disease management and patient outcome enhancement is still not a reality. This paper presents recent data on the utility of TMAO as a cardiovascular biomarker, categorized by its various roles: diagnostic, prognostic, susceptibility/risk, monitoring, pharmacodynamic/response, predictive, and safety. It also briefly discusses research on TMAO’s potential role in cardiovascular disease development. While TMAO shows promise, particularly in prognostic applications, its reliability as a biomarker has been inconsistent across studies. These variances may result from several confounding factors that affect TMAO plasma levels, including diet, kidney function, and demographic variables. The review aims to elucidate the specific contexts in which TMAO can be valuable, potentially leading to more personalized and effective management of cardiovascular disease. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>TMA/TMAO metaorganismal pathway. TMA—trimethylamine; TMAO—trimethylamine N-oxide; FMO3—flavin-containing monooxygenase 3.</p>
Full article ">Figure 2
<p>Hypothetical TMAO effects on cardiovascular pathology. ER stress—endoplasmic reticulum stress; FOXO1—Forkhead box protein O1; IL—interleukin; mt ROS—mitochondrial reactive oxygen species; NF-κB—nuclear factor kappa-light-chain-enhancer of activated B cells; NLRP3—NOD-, LRR-, and pyrin domain-containing protein 3; PERK—protein kinase R-like endoplasmic reticulum kinase; TMAO—trimethylamine N-oxide; TNFα—tumor necrosis factor alpha; VCAM-1—vascular cell adhesion molecule 1.</p>
Full article ">
25 pages, 6147 KiB  
Review
The Gut–Heart Axis: Molecular Perspectives and Implications for Myocardial Infarction
by Katherine Rivera, Leticia Gonzalez, Liena Bravo, Laura Manjarres and Marcelo E. Andia
Int. J. Mol. Sci. 2024, 25(22), 12465; https://doi.org/10.3390/ijms252212465 - 20 Nov 2024
Viewed by 712
Abstract
Myocardial infarction (MI) remains the leading cause of death globally, imposing a significant burden on healthcare systems and patients. The gut–heart axis, a bidirectional network connecting gut health to cardiovascular outcomes, has recently emerged as a critical factor in MI pathophysiology. Disruptions in [...] Read more.
Myocardial infarction (MI) remains the leading cause of death globally, imposing a significant burden on healthcare systems and patients. The gut–heart axis, a bidirectional network connecting gut health to cardiovascular outcomes, has recently emerged as a critical factor in MI pathophysiology. Disruptions in this axis, including gut dysbiosis and compromised intestinal barrier integrity, lead to systemic inflammation driven by gut-derived metabolites like lipopolysaccharides (LPSs) and trimethylamine N-oxide (TMAO), both of which exacerbate MI progression. In contrast, metabolites such as short-chain fatty acids (SCFAs) from a balanced microbiota exhibit protective effects against cardiac damage. This review examines the molecular mediators of the gut–heart axis, considering the role of factors like sex-specific hormones, aging, diet, physical activity, and alcohol consumption on gut health and MI outcomes. Additionally, we highlight therapeutic approaches, including dietary interventions, personalized probiotics, and exercise regimens. Addressing the gut–heart axis holds promise for reducing MI risk and improving recovery, positioning it as a novel target in cardiovascular therapy. Full article
Show Figures

Figure 1

Figure 1
<p>Host–microorganism interface. (<b>A</b>) Schematic representation of the main components of the intestinal barrier. (<b>B</b>) Junctional complexes linking adjacent epithelial cells in normal and impaired intestinal barrier.</p>
Full article ">Figure 2
<p>Complexity of the gut microbiota and its adaptation to different microenvironments in the lower GI tract. Four major bacterial phyla (Bacteroidetes, Firmicutes, Proteobacteria, and Actinobacteria) are found in different sections of the GI tract. Oxygen levels decrease progressively from the stomach to the colon, reflecting a shift from an aerobic to an anaerobic environment. Population density and mucus thickness both increase from the stomach to the colon, corresponding with higher microbial diversity and density in the large intestine, while pH decreases along the tract, providing favorable conditions for specific bacterial communities in different regions.</p>
Full article ">Figure 3
<p>Risk factors in the gut–heart axis in health and disease. In a healthy state (eubiosis), factors like exercise and a fiber- and antioxidant-rich diets support beneficial gut bacteria, boosting SCFA production and limiting harmful compounds like TMA and LPS. Conversely, risk factors such as a Western diet, aging, antibiotics, and pollution lead to gut dysbiosis, where pathogenic bacteria increase inflammatory mediators, impair gut integrity, and raise systemic inflammation and MI risk.</p>
Full article ">
26 pages, 1702 KiB  
Review
Gut Microbe-Generated Metabolite Trimethylamine-N-Oxide and Ischemic Stroke
by Zhen Li, Xinyi He, Qi Fang and Xulong Yin
Biomolecules 2024, 14(11), 1463; https://doi.org/10.3390/biom14111463 - 18 Nov 2024
Viewed by 522
Abstract
Trimethylamine-N-oxide (TMAO) is a gut microbiota-derived metabolite, the production of which in vivo is mainly regulated by dietary choices, gut microbiota, and the hepatic enzyme flavin monooxygenase (FMO), while its elimination occurs via the kidneys. The TMAO level is positively correlated with the [...] Read more.
Trimethylamine-N-oxide (TMAO) is a gut microbiota-derived metabolite, the production of which in vivo is mainly regulated by dietary choices, gut microbiota, and the hepatic enzyme flavin monooxygenase (FMO), while its elimination occurs via the kidneys. The TMAO level is positively correlated with the risk of developing cardiovascular diseases. Recent studies have found that TMAO plays an important role in the development of ischemic stroke. In this review, we describe the relationship between TMAO and ischemic stroke risk factors (hypertension, diabetes, atrial fibrillation, atherosclerosis, thrombosis, etc.), disease risk, severity, prognostic outcomes, and recurrence and discuss the possible mechanisms by which they interact. Importantly, TMAO induces atherosclerosis and thrombosis through lipid metabolism, foam cell formation, endothelial dysfunction (via inflammation, oxidative stress, and pyroptosis), enhanced platelet hyper-reactivity, and the upregulation and activation of vascular endothelial tissue factors. Although the pathogenic mechanisms underlying TMAO’s aggravation of disease severity and its effects on post-stroke neurological recovery and recurrence risk remain unclear, they may involve inflammation, astrocyte function, and pro-inflammatory monocytes. In addition, this paper provides a summary and evaluation of relevant preclinical and clinical studies on interventions regarding the gut-microbiota-dependent TMAO level to provide evidence for the prevention and treatment of ischemic stroke through the gut microbe–TMAO pathway. Full article
(This article belongs to the Section Molecular Medicine)
Show Figures

Figure 1

Figure 1
<p>The relationship between TMAO and ischemic stroke. TMAO is associated with ischemic stroke risk factors (hypertension, diabetes, dyslipidemia, obesity, atrial fibrillation, atherosclerosis, and thrombosis), disease risk, severity, prognostic outcomes, and recurrence. The pathogenic mechanisms underlying the aggravation of the disease severity by TMAO and its effects on prognostic outcomes and recurrence risk remain unclear but may involve inflammation, astrocyte function, and pro-inflammatory monocytes. TMAO: trimethylamine-N-oxide.</p>
Full article ">Figure 2
<p>TMAO plays an important role in ischemic stroke risk. It may be involved in the pathogenesis of diseases such as hypertension, diabetes, dyslipidemia, obesity, atrial fibrillation, atherosclerosis, and thrombosis, ultimately increasing the ischemic stroke risk. Importantly, TMAO induces atherosclerosis and thrombosis through several mechanisms, including lipid metabolism, foam cell formation, endothelial dysfunction (via inflammation, oxidative stress, pyroptosis), enhanced platelet hyper-reactivity, and the upregulation and activation of vascular endothelial tissue factor. AGEs: advanced glycation end products, RCT: the reverse cholesterol transport, TMAO: trimethylamine-N-oxide.</p>
Full article ">Figure 3
<p>TMAO and interventions. The therapeutic strategies targeting gut dysbiosis and TMAO include diet, movement, probiotics and prebiotics, antibiotics, FMT, TMA- and TMAO-related targeted therapies, phytochemicals, and traditional Chinese medicine. FMT: fecal microbiota transplantation, TMA: trimethylamine, TMAO: trimethylamine-N-oxide.</p>
Full article ">
18 pages, 369 KiB  
Review
The Oral Microbiota, Microbial Metabolites, and Immuno-Inflammatory Mechanisms in Cardiovascular Disease
by Zheng Wang, Robert C. Kaplan, Robert D. Burk and Qibin Qi
Int. J. Mol. Sci. 2024, 25(22), 12337; https://doi.org/10.3390/ijms252212337 - 17 Nov 2024
Viewed by 443
Abstract
Cardiovascular diseases (CVDs) remain a leading cause of global morbidity and mortality. Recent advancements in high-throughput omics techniques have enhanced our understanding of the human microbiome’s role in the development of CVDs. Although the relationship between the gut microbiome and CVDs has attracted [...] Read more.
Cardiovascular diseases (CVDs) remain a leading cause of global morbidity and mortality. Recent advancements in high-throughput omics techniques have enhanced our understanding of the human microbiome’s role in the development of CVDs. Although the relationship between the gut microbiome and CVDs has attracted considerable research attention and has been rapidly evolving in recent years, the role of the oral microbiome remains less understood, with most prior studies focusing on periodontitis-related pathogens. In this review, we summarized previously reported associations between the oral microbiome and CVD, highlighting known CVD-associated taxa such as Porphyromonas gingivalis, Fusobacterium nucleatum, and Aggregatibacter actinomycetemcomitans. We also discussed the interactions between the oral and gut microbes. The potential mechanisms by which the oral microbiota can influence CVD development include oral and systemic inflammation, immune responses, cytokine release, translocation of oral bacteria into the bloodstream, and the impact of microbial-related products such as microbial metabolites (e.g., short-chain fatty acids [SCFAs], trimethylamine oxide [TMAO], hydrogen sulfide [H2S], nitric oxide [NO]) and specific toxins (e.g., lipopolysaccharide [LPS], leukotoxin [LtxA]). The processes driven by these mechanisms may contribute to atherosclerosis, endothelial dysfunction, and other cardiovascular pathologies. Integrated multi-omics methodologies, along with large-scale longitudinal population studies and intervention studies, will facilitate a deeper understanding of the metabolic and functional roles of the oral microbiome in cardiovascular health. This fundamental knowledge will support the development of targeted interventions and effective therapies to prevent or reduce the progression from cardiovascular risk to clinical CVD events. Full article
(This article belongs to the Special Issue Microbial Omics)
15 pages, 2606 KiB  
Review
Intestinal Insights: The Gut Microbiome’s Role in Atherosclerotic Disease: A Narrative Review
by Luana Alexandrescu, Adrian Paul Suceveanu, Alina Mihaela Stanigut, Doina Ecaterina Tofolean, Ani Docu Axelerad, Ionut Eduard Iordache, Alexandra Herlo, Andreea Nelson Twakor, Alina Doina Nicoara, Cristina Tocia, Andrei Dumitru, Eugen Dumitru, Laura Maria Condur, Cristian Florentin Aftenie and Ioan Tiberiu Tofolean
Microorganisms 2024, 12(11), 2341; https://doi.org/10.3390/microorganisms12112341 - 16 Nov 2024
Viewed by 518
Abstract
Recent advances have highlighted the gut microbiota as a significant contributor to the development and progression of atherosclerosis, which is an inflammatory cardiovascular disease (CVD) characterized by plaque buildup within arterial walls. The gut microbiota, consisting of a diverse collection of microorganisms, impacts [...] Read more.
Recent advances have highlighted the gut microbiota as a significant contributor to the development and progression of atherosclerosis, which is an inflammatory cardiovascular disease (CVD) characterized by plaque buildup within arterial walls. The gut microbiota, consisting of a diverse collection of microorganisms, impacts the host’s metabolism, immune responses, and lipid processing, all of which contribute to atherosclerosis. This review explores the complex mechanisms through which gut dysbiosis promotes atherogenesis. We emphasize the potential of integrating microbiota modulation with traditional cardiovascular care, offering a holistic approach to managing atherosclerosis. Important pathways involve the translocation of inflammatory microbial components, modulation of lipid metabolism through metabolites such as trimethylamine-N-oxide (TMAO), and the production of short-chain fatty acids (SCFAs) that influence vascular health. Studies reveal distinct microbial profiles in atherosclerosis patients, with increased pathogenic bacteria (Megamonas, Veillonella, Streptococcus) and reduced anti-inflammatory genera (Bifidobacterium, Roseburia), highlighting the potential of these profiles as biomarkers and therapeutic targets. Probiotics are live microorganisms that have health benefits on the host. Prebiotics are non-digestible dietary fibers that stimulate the growth and activity of beneficial gut bacteria. Interventions targeting microbiota, such as probiotics, prebiotics, dietary modifications, and faecal microbiota transplantation (FMT), present effective approaches for restoring microbial equilibrium and justifying cardiovascular risk. Future research should focus on longitudinal, multi-omics studies to clarify causal links and refine therapeutic applications. Full article
(This article belongs to the Section Gut Microbiota)
Show Figures

Figure 1

Figure 1
<p>In vivo metabolic pathways of TMAO and its precursors. Choline is metabolized by the choline trimethylamine-lyase system. Betaine can be catalyzed by L-carnitine dehydrogenase and subsequently reduced to TMA. L-carnitine can be immediately turned into TMA, and TMA can be oxidized to TMAO in the liver and intestines (jejunum and cecum) before entering the bloodstream. Created with <a href="http://BioRender.com" target="_blank">BioRender.com</a> [<a href="#B31-microorganisms-12-02341" class="html-bibr">31</a>] (accessed on 29 October 2024).</p>
Full article ">Figure 2
<p>How the oral microbiome can contribute to an increased frequency of strokes. The presentation focuses on the significant adverse consequences that dysbiosis has on the central nervous system, the vascular system, the gut, the liver, the metabolism of lipids and carbohydrates, and fat tissue. Created with <a href="http://BioRender.com" target="_blank">BioRender.com</a> [<a href="#B31-microorganisms-12-02341" class="html-bibr">31</a>] (accessed on 29 October 2024).</p>
Full article ">Figure 3
<p>Microbiota locations that impact atherosclerosis. Bacterial DNA identified in atherosclerotic plaques may originate from bacteria located in the superior or inferior gastrointestinal tract. Created with <a href="http://BioRender.com" target="_blank">BioRender.com</a> (accessed on 29 October 2024).</p>
Full article ">Figure 4
<p>Pathway from bacterial invasion to blood clot: Bacterial cells release various components, including LPS, pathogen-associated molecular patterns (PAMPs), and outer membrane vesicles (OMVs), which can trigger an immune response. The invasion activates immune receptors such as CXCR2, CCR2, TLR-4/2, and NLRP3. This leads to an increase in inflammatory mediators, including IL-1β, IL-10, IL-17, Th17, IFN-γ, GM-CSF, G-CSF, IL-8, TNF-α, MCP1, and CRP. The immune response causes a reduction in nitric oxide (NO) levels and increases expression of endothelial adhesion molecules, such as E-selectin, ZO-1, vWF, PECAM-1, VCAM-1, and ICAM-1, disrupting endothelial function. Plaque formation involves oxidative stress, inflammatory cell infiltration, an imbalance in M1/M2 macrophage ratio, formation of foam cells, and lipid accumulation within the arterial wall. As plaque forms, factors like Cdc42 activation and increased collagen-binding proteins contribute to clotting. This leads to altered clotting and partial thromboplastin times, ultimately promoting thrombus (blood clot) formation [<a href="#B17-microorganisms-12-02341" class="html-bibr">17</a>]. Created with <a href="http://BioRender.com" target="_blank">BioRender.com</a> [<a href="#B31-microorganisms-12-02341" class="html-bibr">31</a>] (accessed on 29 October 2024).</p>
Full article ">Figure 5
<p>Potential health impacts of gut microbiota imbalance and role of FMT: Gut dysbiosis has been linked to neurological disorders like multiple sclerosis, cardiovascular conditions such as atherosclerosis, metabolic issues including obesity and type 2 diabetes, liver diseases like non-alcoholic fatty liver disease, and gastrointestinal disorders such as C. difficile infection, IBS, and IBD. Created with <a href="http://BioRender.com" target="_blank">BioRender.com</a> [<a href="#B31-microorganisms-12-02341" class="html-bibr">31</a>] (accessed on 29 October 2024).</p>
Full article ">Figure 6
<p>The potential and challenges of microbiota-based therapies in atherosclerosis. Created with <a href="http://BioRender.com" target="_blank">BioRender.com</a> [<a href="#B31-microorganisms-12-02341" class="html-bibr">31</a>] (accessed on 13 November 2024).</p>
Full article ">
20 pages, 1232 KiB  
Review
Primary Prevention Strategy for Non-Communicable Diseases (NCDs) and Their Risk Factors: The Role of Intestinal Microbiota
by Itzel Ivonn López-Tenorio, Óscar Rodrigo Aguilar-Villegas, Yoshua Espinoza-Palacios, Lorena Segura-Real, Berenice Peña-Aparicio, Amedeo Amedei and María Magdalena Aguirre-García
Biomedicines 2024, 12(11), 2529; https://doi.org/10.3390/biomedicines12112529 - 5 Nov 2024
Viewed by 752
Abstract
Non-communicable diseases (NCDs) are the leading cause of morbidity and mortality worldwide. These conditions have numerous health consequences and significantly impact patients’ lifestyles. Effective long-term treatment is essential since NCDs are irreversible. Therefore, primary healthcare must be both exclusive and of the highest [...] Read more.
Non-communicable diseases (NCDs) are the leading cause of morbidity and mortality worldwide. These conditions have numerous health consequences and significantly impact patients’ lifestyles. Effective long-term treatment is essential since NCDs are irreversible. Therefore, primary healthcare must be both exclusive and of the highest quality, ensuring comprehensive care. The primary goal should be to improve quality of life with a focus on patients, families, and communities, as most of these diseases can be prevented and controlled, although not cured. Several factors have been linked to individual health, including social, cultural, and economic aspects, lifestyle, and certain environmental factors, including work, that can have positive or negative effects. More of these variables may contribute to the onset of NCDs, which are defined by their chronic nature, propensity for prolongation, and generally slow rate of progression. Examples of NCDs include hypertension, type 2 diabetes (T2D), dyslipidemia, and fatty liver disease linked to metabolic dysfunction. The onset of these diseases has been associated with an imbalance in certain microbial niches, such as the gut, which hosts billions of microorganisms performing multiple metabolic functions, such as the production of metabolites like bile acids (BAs), short-chain fatty acids (SCFAs), and trimethylamine N-oxide (TMAO). Therefore, lifestyle changes and personal habits can significantly impact the gut microbiota (GM), potentially preventing chronic diseases associated with metabolism. NCDs are highly prevalent worldwide, prompting increased attention to strategies for modifying the intestinal microbiota (IM). Approaches such as probiotics, prebiotics, synbiotics, and fecal transplantation (FMT) have demonstrated improvements in the quality of life for individuals with these conditions. Additionally, lifestyle changes and the adoption of healthy habits can significantly impact IM and may help prevent chronic diseases related to metabolism. Therefore, the main aim of this review is to analyze and understand the importance of microbiota intervention in the prevention of non-communicable diseases. R3:A1 Full article
(This article belongs to the Section Microbiology in Human Health and Disease)
Show Figures

Figure 1

Figure 1
<p>Gut microbiota (GM) in eubiosis and dysbiosis and associated metabolites. (<b>A</b>) In a state of eubiosis; blue box, GM is characterized by a diverse and rich bacterial community that the increase of beneficial metabolites, like short-chain fatty acids (SCFAs). These metabolites have anti-inflammatory properties, regulate cholesterol and glucose metabolism and provide energy and maintain the integrity of the gut barrier, reducing the risk of degenerative diseases and preserving the gut barrier. (<b>B</b>) The figure also illustrates the consequences of intestinal disbiosis; red box, the loss of gut permeability, caused by an imbalance in microbial composition and bacterial translocation. This state is marked by an increase in levels of harmful metabolites, such as trimethylamine N-oxide (TMAO) and lipopolysaccharides (LPS), which contribute to increased intestinal permeability, endotoxemia, and chronic inflammation. Dysbiosis also activates toll-like receptor 4 (TLR4), triggering an inflammatory cytokine cascade. Elevated TMAO and LPS levels have been linked to insulin resistance, inflammation, and a higher risk of metabolic disorders, including obesity, diabetes type 2 (T2DM), systemic arterial hypertension, and cardiovascular diseases (CVD).</p>
Full article ">
22 pages, 5535 KiB  
Article
Monitoring Meat Freshness with Intelligent Colorimetric Labels Containing Red Cabbage Anthocyanins Copigmented with Gelatin and Gallic Acid
by Minyoung Kwak and Sea C. Min
Foods 2024, 13(21), 3464; https://doi.org/10.3390/foods13213464 - 29 Oct 2024
Viewed by 557
Abstract
Polyvinyl alcohol (PVA)-based pH-responsive color indicators were developed using red cabbage anthocyanin (Anth) copigmented with gelatin and gallic acid (GA). The indicator prepared with gelatin and GA (GA/gelatin/Anth/PVA) was highly resistant to light exposure. GA/gelatin/Anth/PVA exhibited distinct color changes in pH 2–11 buffer [...] Read more.
Polyvinyl alcohol (PVA)-based pH-responsive color indicators were developed using red cabbage anthocyanin (Anth) copigmented with gelatin and gallic acid (GA). The indicator prepared with gelatin and GA (GA/gelatin/Anth/PVA) was highly resistant to light exposure. GA/gelatin/Anth/PVA exhibited distinct color changes in pH 2–11 buffer solutions and stable color indication in acidic and neutral solid systems (pH 2 and 7) at 97% relative humidity. GA/gelatin/Anth/PVA exhibited the highest sensitivity to dimethylamine, followed by ammonia and trimethylamine. The addition of gelatin and GA facilitated hydrogen bonding, which enhanced thermal stability and water solubility without compromising tensile properties. A color change from purple to blue signaled spoilage when total volatile basic nitrogen values for beef and squid reached 21.0 and 37.8 mg/100 g, respectively. The GA/gelatin/Anth/PVA indicator shows potential for indicating the freshness of raw beef. Full article
(This article belongs to the Section Food Packaging and Preservation)
Show Figures

Figure 1

Figure 1
<p>Changes in ∆<span class="html-italic">E</span> values of polyvinyl alcohol (PVA)-based pH-responsive color indicators using anthocyanin (Anth; Anth/PVA), Anth copigmented with gallic acid (GA; GA/Anth/PVA), Anth copigmented with gelatin (Gelatin/Anth/PVA), Anth copigmented with gelatin and GA (GA/gelatin/Anth/PVA), Anth copigmented with gelatin and aluminum ions (AL; AL/gelatin/Anth/PVA), and Anth copigmented with gelatin, GA, and AL (AL/GA/gelatin/Anth/PVA) determined during storage for 12 days at 4 °C (<b>A</b>) under fluorescent light and (<b>B</b>) dark conditions (<span class="html-italic">n</span> = 6).</p>
Full article ">Figure 2
<p>(<b>A</b>) Images and (<b>B</b>) UV–Vis spectra of the Anth solution in various pH buffer solutions.</p>
Full article ">Figure 3
<p>Changes in ∆<span class="html-italic">E</span> values of the polyvinyl alcohol (PVA)-based pH-responsive color indicator using anthocyanin (Anth) copigmented with gelatin and gallic acid (GA; GA/gelatin/Anth/PVA) at pH 2, 7, and 11 and 74, 84, and 97% RH with respect to storage time during storage at (<b>A</b>) 4 °C, (<b>B</b>) 10 °C, and (<b>C</b>) 25 °C. Means followed by different small letters (a–d) above the symbols indicate significant differences over storage time (<span class="html-italic">p</span> &lt; 0.05), reflecting that the color changes of the indicators at the same pH level vary over time at the same RH. Means (<span class="html-italic">n</span> = 6) followed by different capital letters (A–C) above the symbols indicate significant differences over relative humidity (<span class="html-italic">p</span> &lt; 0.05), illustrating the impact of varying RH levels on the color changes of the indicators at the same storage time point for each pH level.</p>
Full article ">Figure 4
<p>Changes in ∆<span class="html-italic">E</span> values of the polyvinyl alcohol (PVA)-based pH-responsive color indicator using anthocyanin (Anth) copigmented with gelatin and gallic acid (GA; GA/gelatin/Anth/PVA) at pH 2 and 7 at 4, 10, and 25 °C with respect to storage temperature during storage at (<b>A</b>) 74%, (<b>B</b>) 84%, and (<b>C</b>) 97% RH. Means (<span class="html-italic">n</span> = 6) followed by different small letters (a–c) above the symbols indicate significant differences over storage temperature (<span class="html-italic">p</span> &lt; 0.05), reflecting the impact of varying temperatures on the color changes of the indicators at the same storage time point for each pH level.</p>
Full article ">Figure 5
<p>Scanning electron microscopy images of the surfaces and cross-sections of the (<b>A</b>) polyvinyl alcohol (PVA) film, (<b>B</b>) PVA-based pH-responsive color indicator using anthocyanin (Anth; Anth/PVA), and (<b>C</b>) PVA-based pH-responsive color indicator using Anth copigmented with gelatin and gallic acid(GA; GA/gelatin/Anth/PVA).</p>
Full article ">Figure 6
<p>(<b>A</b>) Fourier-transform infrared spectroscopy spectra and (<b>B</b>) thermal degradation curves of the polyvinyl alcohol (PVA) film, PVA-based pH-responsive color indicator using anthocyanin (Anth; Anth/PVA), and polyvinyl alcohol-based pH-responsive color indicator using Anth copigmented with gelatin and gallic acid (GA; GA/gelatin/Anth/PVA).</p>
Full article ">Figure 7
<p>Changes in the pH and TVBN values (<span class="html-italic">n</span> = 4) of raw beef and the color of the polyvinyl alcohol (PVA)-based pH-responsive color indicator using anthocyanin (Anth) copigmented with gelatin and gallic acid (GA; GA/gelatin/Anth/PVA) during storage at (<b>A</b>) 4 and (<b>B</b>) 25 °C.</p>
Full article ">
15 pages, 6263 KiB  
Article
Palmitoleic Acid Inhibits Hepatotoxic Effects by Reducing Trimethylamine-N-Oxide (TMAO) Formation in High L-Carnitine-Treated Mice
by Qingzheng Han, Yu Liu, Xinyu Liu, Yue Geng, Qiu Wu and Hang Xiao
Nutrients 2024, 16(21), 3599; https://doi.org/10.3390/nu16213599 - 23 Oct 2024
Viewed by 955
Abstract
Background/Objectives: This study investigated the effects of palmitoleic acid (POA) consumption on liver function, intestinal microbiota, and trimethylamine-N-oxide (TMAO) levels in the serum of mice treated with 3% L-carnitine drinking water. The purpose was to highlight the impact of POA on [...] Read more.
Background/Objectives: This study investigated the effects of palmitoleic acid (POA) consumption on liver function, intestinal microbiota, and trimethylamine-N-oxide (TMAO) levels in the serum of mice treated with 3% L-carnitine drinking water. The purpose was to highlight the impact of POA on liver injury associated with high L-carnitine intake. Methods: A correlation analysis was carried out. The physiological and biochemical results showed that the administration of POA could alleviate liver injury induced by high L-carnitine ingestion, as reflected by a reduction in liver function indices (ALT, AST, AKP, and TBA activities) and modulation of antioxidant enzyme activities (SOD, GSH-Px, MDA, and RAHFR). The study also monitored the levels of total cholesterol (TC), triglycerides (TG), low-density lipoprotein cholesterol (LDL-C), and high-density lipoprotein cholesterol (HDL-C). Additionally, to assess the impact of POA on intestinal microbiota, we conducted a 16S rRNA high-throughput sequencing analysis. Results: The findings indicated that POA administration resulted in lower levels of TMAO in treated mice. Furthermore, POA could regulate the composition of intestinal microbiota in L-carnitine mice, particularly affecting Bacteroides vulgatus, Parabacteroides distasonis, Alistipes shahii, Lachnospiraceae NK4A136 group, and Parasutterella secunda, which were closely related to liver injury. Conclusions: In summary, POA could repair liver damage caused by high intake of L-carnitine by regulating the distribution of intestinal flora and subsequently decreasing serum TMAO levels. Full article
(This article belongs to the Special Issue Health Effects of Edible Oils and Their Functional Components)
Show Figures

Figure 1

Figure 1
<p>Effects of POA on the transaminase levels and the four indicators of liver function in livers of high L-carnitine-treated mice—ALT (<b>A</b>), AST (<b>B</b>), TBA (<b>C</b>), ALB (<b>D</b>), TP (<b>E</b>), and AKP (<b>F</b>). (Asterisks (*) indicate levels of statistical significance where * represents <span class="html-italic">p</span> &lt; 0.05 and ** represents <span class="html-italic">p</span> &lt; 0.01 compared to the control group).</p>
Full article ">Figure 2
<p>Effects of POA on the antioxidant capacity and lipid homeostasis of liver in high L-carnitine-treated mice—TC (<b>A</b>), TG (<b>B</b>), HDL-C (<b>C</b>), LDL-C (<b>D</b>), SOD (<b>E</b>), RAHFR (<b>F</b>), MDA (<b>G</b>) and GSH-PX (<b>H</b>). (Asterisks (*) indicate levels of statistical significance where * represents <span class="html-italic">p</span> &lt; 0.05 and ** represents <span class="html-italic">p</span> &lt; 0.01 compared to the control group).</p>
Full article ">Figure 3
<p>Effects of POA on histopathological changes in the livers of mice treated with high doses of L-carnitine—liver hepatocytes stained with H&amp;E (Normal, Model, POA 50, POA 100, and POA 200) (<b>A</b>), and the TMAO content in the serum (<b>B</b>).</p>
Full article ">Figure 4
<p>Venn diagram (<b>A</b>) of the gut microbiota of mice in different groups, rarefaction curve analysis (<b>B</b>), PCA plot (<b>C</b>), PCoA plot (<b>D</b>), NMDS clustering plot (<b>E</b>), and bar chart of relative species abundance at the phylum level (<b>F</b>).</p>
Full article ">Figure 5
<p>Relative abundance of gut microbiota at the genus level in mice of different groups (<b>A</b>) and an abundance heatmap at the genus level (<b>B</b>).</p>
Full article ">Figure 6
<p>LEfSe analysis of gut microbiota in the Model group and in POA group mice—Significant difference chart (<b>A</b>) and representative cladogram (<b>B</b>).</p>
Full article ">Figure 7
<p>Spearman correlation heatmap of gut microbiota composition and liver injury markers. (Asterisks (*) indicate statistically significant correlations (<span class="html-italic">p</span> &lt; 0.05)).</p>
Full article ">
24 pages, 4250 KiB  
Article
Gut Microbiota and Metabolic Alterations Associated with Heart Failure and Coronary Artery Disease
by Adel A. Yafarova, Elena V. Dementeva, Olga A. Zlobovskaya, Anna F. Sheptulina, Elena V. Lopatukhina, Yuriy S. Timofeev, Evgeniya V. Glazunova, Aleksey V. Lyundup, Yuriy V. Doludin, Anton R. Kiselev, German A. Shipulin, Valentin V. Makarov, Oxana M. Drapkina and Sergey M. Yudin
Int. J. Mol. Sci. 2024, 25(20), 11295; https://doi.org/10.3390/ijms252011295 - 20 Oct 2024
Viewed by 1565
Abstract
This study investigates the role of gut microbiota in cardiovascular diseases, with an additional focus on pro-atherogenic metabolites. We use advanced network analysis and machine learning techniques to identify key microbial features linked to coronary artery disease (CAD) and heart failure with reduced [...] Read more.
This study investigates the role of gut microbiota in cardiovascular diseases, with an additional focus on pro-atherogenic metabolites. We use advanced network analysis and machine learning techniques to identify key microbial features linked to coronary artery disease (CAD) and heart failure with reduced ejection fraction (HFrEF). This cross-sectional study included 189 participants divided into three groups: coronary artery disease (n = 93), heart failure with reduced ejection fraction (n = 43), and controls (n = 53). Assessments included physical exams, echocardiography, dietary surveys, blood analysis, and fecal analysis. Gut microbiota composition was analyzed using next-generation sequencing (NGS) and quantitative polymerase chain reaction (qPCR). Statistical analysis methods for testing hypotheses and correlations, alpha and beta-diversity analyses, co-occurrence networks, and machine learning were conducted using Python libraries or R packages with multiple comparisons corrected using the Benjamini–Hochberg procedure. Significant gut microbiota alterations were observed, with higher Bacillota/Bacteroidota ratios in CAD and HFrEF groups compared to controls (p < 0.001). Significant differences were observed in α-diversity indices (Pielou, Chao1, Faith) between disease groups and controls (p < 0.001). β-diversity analyses also revealed distinct microbial profiles (p = 0.0015). Interestingly, trimethylamine N-oxide (TMAO) levels were lower in CAD and HFrEF groups compared to controls (p < 0.05), while indoxyl sulfate (IS) levels were comparable between the study groups. Co-occurrence network analysis and machine learning identified key microbial features linked to these conditions, highlighting complex interactions within the gut microbiota associated with cardiovascular disease. Full article
Show Figures

Figure 1

Figure 1
<p>Serum IS and TMAO concentrations in patients with HFrEF, CAD, and the control group.</p>
Full article ">Figure 2
<p>Alpha-diversity indices in the studied groups.</p>
Full article ">Figure 3
<p>Principal Coordinates Analysis (PCoA) analysis of microbial communities in HFrEF, CAD, and control groups. Each point on the graph represents a single microbial community sample from the HFrEF, CAD, or control groups.</p>
Full article ">Figure 4
<p>Histogram of the community composition of gut microbiota at the phylum level in patients with HFrEF, CAD, and the control group according to Next-Generation Sequencing data (RDP database).</p>
Full article ">Figure 5
<p>Volcano plots for the MaAsLin2 results. (<b>A</b>) CAD-control; (<b>B</b>) HFrEF-control.</p>
Full article ">Figure 5 Cont.
<p>Volcano plots for the MaAsLin2 results. (<b>A</b>) CAD-control; (<b>B</b>) HFrEF-control.</p>
Full article ">Figure 6
<p>Microbial network for bacteria important for differentiating diseases and those with at least five nodes.</p>
Full article ">Figure 7
<p>Study design: (<b>A</b>) samples and data collection; (<b>B</b>) data analysis. HFrEF—Heart Failure with reduced Ejection Fraction; CAD—Coronary Artery Disease; IS—Indoxyl Sulfate; TMAO—trimethylamine N-oxide; QIIME2—Quantitative Insights Into Microbial Ecology; DADA2—Divisive Amplicon Denoising Algorithm; RDP—Ribosomal Database Project; OTU—Operational Taxonomic Unit; N/A—Not Available; RF—Random Forest.</p>
Full article ">Figure 7 Cont.
<p>Study design: (<b>A</b>) samples and data collection; (<b>B</b>) data analysis. HFrEF—Heart Failure with reduced Ejection Fraction; CAD—Coronary Artery Disease; IS—Indoxyl Sulfate; TMAO—trimethylamine N-oxide; QIIME2—Quantitative Insights Into Microbial Ecology; DADA2—Divisive Amplicon Denoising Algorithm; RDP—Ribosomal Database Project; OTU—Operational Taxonomic Unit; N/A—Not Available; RF—Random Forest.</p>
Full article ">
17 pages, 2192 KiB  
Article
Metabolite, Biochemical, and Dietary Intake Alterations Associated with Lifestyle Interventions in Obese and Overweight Malaysian Women
by Fatin Saparuddin, Mohd Naeem Mohd Nawi, Liyana Ahmad Zamri, Fazliana Mansor, Mohd Fairulnizal Md Noh, Mohd Azahadi Omar, Nur Shahida Abdul Aziz, Norasyikin A. Wahab, Ahmed Mediani, Nor Fadilah Rajab and Razinah Sharif
Nutrients 2024, 16(20), 3501; https://doi.org/10.3390/nu16203501 - 16 Oct 2024
Viewed by 680
Abstract
Differences in metabolic regulation among obesity phenotypes, specifically metabolically healthy obese (MHO) and metabolically unhealthy obese (MUO) women, may lead to varied responses to interventions, which could be elucidated through metabolomics. Therefore, this study aims to investigate the differences in metabolite profiles between [...] Read more.
Differences in metabolic regulation among obesity phenotypes, specifically metabolically healthy obese (MHO) and metabolically unhealthy obese (MUO) women, may lead to varied responses to interventions, which could be elucidated through metabolomics. Therefore, this study aims to investigate the differences in metabolite profiles between MHO and MUO women and the changes following a lifestyle intervention. Serum samples from 36 MHO and 34 MUO women who participated in a lifestyle intervention for weight loss were analysed using untargeted proton nuclear magnetic resonance spectroscopy (1H NMR) at baseline and 6 months post-intervention. Anthropometric, clinical, and dietary intake parameters were assessed at both time points. Both groups showed differential metabolite profiles at baseline and after six months. Seven metabolites, including trimethylamine-N-oxide (TMAO), arginine, ribose, aspartate, carnitine, choline, and tyrosine, significantly changed between groups post-intervention, which all showed a decreasing pattern in MHO. Significant reductions in body weight and body mass index (BMI) in the MUO correlated with changes in the carnitine and tyrosine levels. In conclusion, metabolite profiles differed significantly between MHO and MUO women before and after a lifestyle intervention. The changes in carnitine and tyrosine levels in MUO were correlated with weight loss, suggesting potential targets for therapeutic intervention. Full article
(This article belongs to the Section Nutrition in Women)
Show Figures

Figure 1

Figure 1
<p>Summary of the lifestyle intervention programme.</p>
Full article ">Figure 2
<p>Metabolites that significantly changed between MHO and MUO following intervention.</p>
Full article ">Figure 3
<p>Metabolic pathways affected at sixth month.</p>
Full article ">Figure 4
<p>Heatmap of correlation between the significantly changed metabolites with body weight, BMI, WC, and systolic blood pressure in MUO. Correlation determined using Pearson correlation. Green indicates a positive correlation, while red indicates a negative correlation.</p>
Full article ">Figure 5
<p>Heatmap of correlation between the significantly changed metabolites with changes in anthropometry, clinical, and biochemical variables. Correlation determined using Pearson correlation analysis in both groups combined. Blue indicates a positive correlation, while red indicates a negative correlation.</p>
Full article ">
14 pages, 710 KiB  
Article
Preservative Effect of Alga Flour Extract on Frozen Horse Mackerel (Trachurus trachurus) Lipids
by Miriam Martínez, Marcos Trigo, Santiago P. Aubourg and Alicia Rodríguez
Foods 2024, 13(20), 3265; https://doi.org/10.3390/foods13203265 - 14 Oct 2024
Viewed by 852
Abstract
The aim of this study was to investigate the preservative properties of alga Gelidium sp. flour when included in the glazing medium employed for the frozen storage (−18 °C) of horse mackerel (Trachurus trachurus). Different concentrations (low, medium, and high) of [...] Read more.
The aim of this study was to investigate the preservative properties of alga Gelidium sp. flour when included in the glazing medium employed for the frozen storage (−18 °C) of horse mackerel (Trachurus trachurus). Different concentrations (low, medium, and high) of an aqueous extract were tested and compared to a control water-glazing condition. Quality changes (lipid oxidation and hydrolysis, fatty acid (FA) profile, and trimethylamine (TMA) formation) were determined after 3- and 6-month storage periods. A general quality loss (lipid oxidation with hydrolysis development and TMA formation) with the frozen storage period was detected in all samples. The presence of an alga flour (AF) extract in the glazing medium led to a lower (p < 0.05) TBARS and fluorescent compound formation and to higher (p < 0.05) polyene values in frozen fish. Furthermore, a preserving effect on free fatty acids was detected in AF-treated fish. On the contrary, the AF-glazing treatment did not affect (p > 0.05) the TMA formation and the total n3/total n6 FA ratio. In general, preservative effects were found to be higher in frozen fish corresponding to the medium concentration tested. Current results show the potential of Gelidium sp. flour as a natural source of preservative hydrophilic compounds for the quality enhancement of frozen horse mackerel. Full article
(This article belongs to the Special Issue New Technological Advances in Meat Packaging: Shelf-Life and Safety)
Show Figures

Figure 1

Figure 1
<p>Assessment of the fluorescence ratio in frozen horse mackerel subjected to glazing including different alga flour (AF) contents. Average values of three independent determinations (<span class="html-italic">n</span> = 3); standard deviations are indicated by bars. Different lowercase letters denote significant differences (<span class="html-italic">p</span> &lt; 0.05) with the frozen storage time; at each frozen storage time, different capital letters denote significant differences (<span class="html-italic">p</span> &lt; 0.05) with the glazing condition. Glazing conditions as expressed in <a href="#foods-13-03265-t001" class="html-table">Table 1</a>.</p>
Full article ">
12 pages, 747 KiB  
Article
Elevated Circulating Levels of Gut Microbe-Derived Trimethylamine N-Oxide Are Associated with Systemic Sclerosis
by Karen J. Ho, Lutfiyya N. Muhammad, Linh Ngo Khanh, Xinmin S. Li, Mary Carns, Kathleen Aren, Seok-Jo Kim, Priyanka Verma, Stanley L. Hazen and John Varga
J. Clin. Med. 2024, 13(19), 5984; https://doi.org/10.3390/jcm13195984 - 8 Oct 2024
Viewed by 925
Abstract
Background/Objectives: Alterations in fecal microbial communities in patients with systemic sclerosis (SSc) are common, but the clinical significance of this observation is poorly understood. Gut microbial production of trimethylamine (TMA), and its conversion by the host to trimethylamine N-oxide (TMAO), has clinical [...] Read more.
Background/Objectives: Alterations in fecal microbial communities in patients with systemic sclerosis (SSc) are common, but the clinical significance of this observation is poorly understood. Gut microbial production of trimethylamine (TMA), and its conversion by the host to trimethylamine N-oxide (TMAO), has clinical and mechanistic links to cardiovascular and renal diseases. Direct provision of TMAO has been shown to promote fibrosis and vascular injury, hallmarks of SSc. We sought to determine levels of TMAO and related metabolites in SSc patients and investigate associations between the metabolite levels with disease features. Methods: This is an observational case:control study. Adults with SSc (n = 200) and non-SSc controls (n = 400) were matched for age, sex, indices of renal function, diabetes mellitus, and cardiovascular disease. Serum TMAO, choline, betaine, carnitine, γ-butyrobetaine, and crotonobetaine were measured using stable isotope dilution liquid chromatography tandem mass spectrometry. Results: Median TMAO concentration was higher (p = 0.020) in SSc patients (3.31 [interquartile range 2.18, 5.23] µM) relative to controls (2.85 [IQR 1.88, 4.54] µM). TMAO was highest among obese and male SSc participants compared to all other groups. Following adjustment for sex, BMI, age, race, and eGFR in a quantile regression model, elevated TMAO levels remained associated with SSc at each quantile of TMAO. Conclusions: Patients with SSc have increased circulating levels of TMAO independent of comorbidities including age, sex, renal function, diabetes mellitus, and cardiovascular disease. As a potentially modifiable factor, further studies examining the link between TMAO and SSc disease severity and course are warranted. Full article
(This article belongs to the Special Issue Advances in Clinical Rheumatology)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Comparative analysis of serum TMAO concentration. Each comparison is shown with and without outlier observations. For the comparison of control and SSc groups, box plots of TMAO in the control and SSc groups is shown with (<b>A</b>) and without (<b>D</b>) the outlier observations in both groups. For the comparison based on SSc group and obesity, box plots of TMAO in each group are shown with (<b>B</b>) and without (<b>E</b>) the outlier observations in the non-obese groups (2 in control group and 1 in SSc group). For the comparison based on SSc group and sex, box plots of TMAO in each group are shown with (<b>C</b>) and without (<b>F</b>) the outlier observations in the female groups (two in control group and one in SSc group). Data shown are median with interquartile range. (<b>G</b>) Quantile regression analysis of the differences in TMAO at each quantile between the SSc and control groups. After adjusting for sex, BMI, age, race, and eGFR, SSc participants had an elevated TMAO level in comparison to control participants at all quantile of TMAO. Quantile regression estimated coefficients at each TMAO quantile are shown in <a href="#jcm-13-05984-t001" class="html-table">Table 1</a>.</p>
Full article ">Figure 2
<p>Heat map of serum metabolite concentrations in the SSc cohort. Metabolites are represented in the columns and individual participants are represented in the rows. As described in the text, 7 hierarchical clusters were initially identified and then regrouped into 2 clusters based on sample sizes. The final 2 groups are indicated on the left side of the figure by dark brown (<span class="html-italic">n</span> = 92) or tan (<span class="html-italic">n</span> = 108).</p>
Full article ">
15 pages, 4200 KiB  
Review
Cardiovascular Disease May Be Triggered by Gut Microbiota, Microbial Metabolites, Gut Wall Reactions, and Inflammation
by Leon M. T. Dicks
Int. J. Mol. Sci. 2024, 25(19), 10634; https://doi.org/10.3390/ijms251910634 - 2 Oct 2024
Viewed by 1614
Abstract
Cardiovascular disease (CVD) may be inherited, as recently shown with the identification of single nucleotide polymorphisms (SNPs or “snips”) on a 250 kb DNA fragment that encodes 92 proteins associated with CVD. CVD is also triggered by microbial dysbiosis, microbial metabolites, metabolic disorders, [...] Read more.
Cardiovascular disease (CVD) may be inherited, as recently shown with the identification of single nucleotide polymorphisms (SNPs or “snips”) on a 250 kb DNA fragment that encodes 92 proteins associated with CVD. CVD is also triggered by microbial dysbiosis, microbial metabolites, metabolic disorders, and inflammatory intestinal epithelial cells (IECs). The epithelial cellular adhesion molecule (Ep-CAM) and trefoil factor 3 (TFF3) peptide keeps the gut wall intact and healthy. Variations in Ep-CAM levels are directly linked to changes in the gut microbiome. Leptin, plasminogen activator inhibitor 1 (PAI1), and alpha-1 acid glycoprotein 1 (AGP1) are associated with obesity and may be used as biomarkers. Although contactin 1 (CNTN1) is also associated with obesity and adiposity, it regulates the bacterial metabolism of tryptophan (Trp) and thus appetite. A decrease in CNTN1 may serve as an early warning of CVD. Short-chain fatty acids (SCFAs) produced by gut microbiota inhibit pro-inflammatory cytokines and damage vascular integrity. Trimethylamine N-oxide (TMAO), produced by gut microbiota, activates inflammatory Nod-like receptors (NLRs) such as Nod-like receptor protein 3 (NLRP3), which increase platelet formation. Mutations in the elastin gene (ELN) cause supra valvular aortic stenosis (SVAS), defined as the thickening of the arterial wall. Many of the genes expressed by human cells are regulated by gut microbiota. The identification of new molecular markers is crucial for the prevention of CVD and the development of new therapeutic strategies. This review summarizes the causes of CVD and identifies possible CVD markers. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
Show Figures

Figure 1

Figure 1
<p>A basic explanation for the role of plasma kallikrein in inflammation and blot clotting. Prekallikrein is encoded by the trans-pQTL gene <span class="html-italic">KLKB1</span> in the liver. Factor XII converts prekallikrein to plasma kallikrein and leads to the production of more factor XI. The early stages of blood clotting and the formation of bradykinin are initiated by a combined reaction of plasma kallikrein and factor XII. Bradykinin increases the permeability of blood vessel walls, leading to leakage and inflammation. Trans-pQTL = trans-protein quantitative trait loci. Created using <a href="http://Biorender.com" target="_blank">Biorender.com</a> (accessed on 22 August 2024).</p>
Full article ">Figure 2
<p>A schematic representation explaining the role paraoxonases (PON1, PON2, and PON3) play in cardiovascular disease (CVD). PON1 and PON3 are attached to high-density lipoprotein (HDL) and oxidize lipid peroxides to prevent their accumulation on low-density lipoprotein (LDL). Oxidized LDL (OX-LDL) triggers macrophages to an anti-inflammatory state and forms foam cells. Foam cells and the release of pro-inflammatory cytokines from adipose tissue lead to plaque formation. Plague formation may also be triggered by hyperglycemia. The HDL-PON complex prevents the aggregation of platelets. PON2 represses the formation of reactive oxygen species (ROS) and has an antiatherogenic effect. ATP-binding cassette transporter A1 (ABCA1) mediates the cellular efflux of phospholipids and cholesterol to lipid-poor apolipoprotein A1 (apoA1)-HDL and plays a significant role in the metabolism of HDL. Blue dots in adipose tissue represent cytokines, lighter circles in the foam cell represent the accumulation of triglycerides, the yellow area in the artery represents hyperglycemia and atherosclerosis, LRP1 = low-density lipoprotein receptor-related protein 1, PAI1 = plasminogen activator inhibitor 1, ABCA1 = ATP-binding cassette transporter A1, SR = scavenger receptor. Created using <a href="http://Biorender.com" target="_blank">Biorender.com</a> (accessed on 20 August 2024).</p>
Full article ">Figure 3
<p>The link between gut microbiota and atherosclerosis. Gut microbiota produces trimethylamine (TMA) from choline, derived from dietary phosphatidylcholine. Choline is microbially converted to TMA and then oxidized to the pro-atherogenic metabolite trimethylamine N-oxide (TMAO). TMAO may contribute to atherosclerosis by interference with cholesterol transportation, foam cell formation, and platelet aggregation. Platelet aggregation leads to atherosclerosis. Disruption of intestinal permeability by damaged intestinal epithelial cells (IECs) results in the leakage of bacterial toxins such as lipopolysaccharides (LPS) formed by microbiota, and cell wall peptidoglycan into the bloodstream. These toxins react with Toll-like receptors (TLRs), leading to systemic inflammation and the aggravation of atherosclerosis. A diet with less dietary fiber decreases the microbial production of short-chain fatty acids (SCFAs) such as butyrate. Butyrate is the main energy source for colonocytes and has an immunomodulatory effect on gut mucosa. Dotted lines denote the effect of diets on IECs and inflammation, lighter circles in the foam cell represent the accumulation of triglycerides, BAs = bile acids, ROS = reactive oxygen species, C/NF-κB = canonical nuclear factor kappa-B, FXRs = Farnesoid receptors, NLRP3 = Nod-like receptor protein 3, FMO3 = Flavin-containing monooxygenase 3. Created using <a href="http://Biorender.com" target="_blank">Biorender.com</a> (accessed on 20 August 2024).</p>
Full article ">
16 pages, 1544 KiB  
Article
Preservative Effect of a Gelatin-Based Film Including a Gelidium sp. Flour Extract on Refrigerated Atlantic Mackerel
by Lucía López, Antonio Gómez, Marcos Trigo, José M. Miranda, Jorge Barros-Velázquez and Santiago P. Aubourg
Appl. Sci. 2024, 14(19), 8817; https://doi.org/10.3390/app14198817 - 30 Sep 2024
Viewed by 657
Abstract
This research evaluated the preservative properties of flour from the alga Gelidium sp., which is a waste substrate resulting from commercial phycocolloid extraction. Gelatin-based biofilms, which included two different concentrations of red alga flour, were developed and used as packaging systems during refrigerated [...] Read more.
This research evaluated the preservative properties of flour from the alga Gelidium sp., which is a waste substrate resulting from commercial phycocolloid extraction. Gelatin-based biofilms, which included two different concentrations of red alga flour, were developed and used as packaging systems during refrigerated storage (up to 9 days at 4 °C) of Atlantic mackerel (Scomber scombrus) muscle. In all batches tested, a progressive decrease in quality could be observed in the muscle of the fish as the storage time increased. Compared with the control fish, the Gelidium alga flour extract had an inhibitory effect (p < 0.05) on microbial activity (total aerobes, psychrotrophs, and proteolytic bacteria), lipid oxidation (peroxide, thiobarbituric acid, fluorescence and polyene indices), lipid hydrolysis (formation of free fatty acids) and pH increase in refrigerated mackerel muscle. In contrast, no significant effect (p > 0.05) was observed on trimethylamine formation, Enterobacteriaceae, or lipolytic bacteria counts. A preservative effect resulting from the incorporation of Gelidium alga flour into the gelatin-based biofilm was observed, indicating both quality and safety enhancement. In accordance with current global interest in the search for natural and waste sources, a novel and beneficial use of Gelidium flour for enhancing the quality of refrigerated fish has been proposed. Full article
Show Figures

Figure 1

Figure 1
<p>Determination of aerobic counts (log CFU·g<sup>−1</sup> muscle) in refrigerated mackerel subjected to different packaging conditions. Average values ± standard deviations (<span class="html-italic">n</span> = 3). The packaging conditions are expressed in <a href="#applsci-14-08817-t001" class="html-table">Table 1</a>. Different lowercase letters denote significant differences (<span class="html-italic">p</span> &lt; 0.05) with chilling time; different capital letters denote significant differences (<span class="html-italic">p</span> &lt; 0.05) as a result of the packaging condition. Time 0 corresponds to initial fish.</p>
Full article ">Figure 2
<p>Determination of proteolytic counts (log CFU·g<sup>−1</sup> muscle) in refrigerated mackerel subjected to different packaging conditions. Average values ± standard deviations (<span class="html-italic">n</span> = 3). The packaging conditions are expressed in <a href="#applsci-14-08817-t001" class="html-table">Table 1</a>. Different lowercase letters denote significant differences (<span class="html-italic">p</span> &lt; 0.05) with chilling time; different capital letters denote significant differences (<span class="html-italic">p</span> &lt; 0.05) as a result of the packaging condition. Time 0 corresponds to initial fish.</p>
Full article ">Figure 3
<p>Determination of the free fatty acid (FFA; g·kg<sup>−1</sup> lipids) content in refrigerated mackerel subjected to different packaging conditions. Average values ± standard deviations (<span class="html-italic">n</span> = 3). The packaging conditions are expressed in <a href="#applsci-14-08817-t001" class="html-table">Table 1</a>. Different lowercase letters denote significant differences (<span class="html-italic">p</span> &lt; 0.05) with chilling time; different capital letters denote significant differences (<span class="html-italic">p</span> &lt; 0.05) as a result of the packaging condition. Time 0 corresponds to initial fish.</p>
Full article ">
Back to TopTop