[go: up one dir, main page]

 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (4,464)

Search Parameters:
Keywords = atherosclerosis

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
87 pages, 3719 KiB  
Review
Beyond Anticoagulation: A Comprehensive Review of Non-Vitamin K Oral Anticoagulants (NOACs) in Inflammation and Protease-Activated Receptor Signaling
by Shirin Jannati, Rajashree Patnaik and Yajnavalka Banerjee
Int. J. Mol. Sci. 2024, 25(16), 8727; https://doi.org/10.3390/ijms25168727 (registering DOI) - 10 Aug 2024
Viewed by 239
Abstract
Non-vitamin K oral anticoagulants (NOACs) have revolutionized anticoagulant therapy, offering improved safety and efficacy over traditional agents like warfarin. This review comprehensively examines the dual roles of NOACs—apixaban, rivaroxaban, edoxaban, and dabigatran—not only as anticoagulants, but also as modulators of inflammation via protease-activated [...] Read more.
Non-vitamin K oral anticoagulants (NOACs) have revolutionized anticoagulant therapy, offering improved safety and efficacy over traditional agents like warfarin. This review comprehensively examines the dual roles of NOACs—apixaban, rivaroxaban, edoxaban, and dabigatran—not only as anticoagulants, but also as modulators of inflammation via protease-activated receptor (PAR) signaling. We highlight the unique pharmacotherapeutic properties of each NOAC, supported by key clinical trials demonstrating their effectiveness in preventing thromboembolic events. Beyond their established anticoagulant roles, emerging research suggests that NOACs influence inflammation through PAR signaling pathways, implicating factors such as factor Xa (FXa) and thrombin in the modulation of inflammatory responses. This review synthesizes current evidence on the anti-inflammatory potential of NOACs, exploring their impact on inflammatory markers and conditions like atherosclerosis and diabetes. By delineating the mechanisms by which NOACs mediate anti-inflammatory effects, this work aims to expand their therapeutic utility, offering new perspectives for managing inflammatory diseases. Our findings underscore the broader clinical implications of NOACs, advocating for their consideration in therapeutic strategies aimed at addressing inflammation-related pathologies. This comprehensive synthesis not only enhances understanding of NOACs’ multifaceted roles, but also paves the way for future research and clinical applications in inflammation and cardiovascular health. Full article
(This article belongs to the Special Issue New Trends in Diabetes, Hypertension and Cardiovascular Diseases 2.0)
Show Figures

Figure 1

Figure 1
<p>The current model of the blood coagulation cascade, depicting NOACs’ mechanism of action. There are two pathways, the intrinsic pathway and the extrinsic pathway. These multicomponent processes are illustrated as enzymes, inhibitors, zymogens, or complexes. On injury to the vessel wall, tissue factor, the cofactor for the extrinsic tenase complex, is exposed to circulating FVIIa and forms the extrinsic tenase. FIX and FX are converted to their serine proteases FIXa and FXa, which then form the intrinsic tenase and the prothrombinase complexes, respectively. The combined actions of the intrinsic and extrinsic tenase and the prothrombinase complexes lead to an explosive burst of the enzyme thrombin (IIa). In addition to its multiple procoagulant roles, thrombin also acts in an anticoagulant capacity when combined with the cofactor thrombomodulin in the protein Case complex. The product of the protein Case reaction, activated protein C (APC), inactivates the cofactors FVa and FVIIIa. The cleaved species, FVai and FVIIIai, no longer support the respective procoagulant activities. Once thrombin is generated through procoagulant mechanisms, thrombin cleaves fibrinogen (releasing fibrinopeptide A and B [FPA and FPB]), as well as activating FXIII to form a cross-linked fibrin clot. Thrombin–thrombomodulin also activates thrombin activate-able fibrinolysis inhibitor, which slows fibrin degradation by plasmin. The procoagulant response is downregulated by the stoichiometric inhibitor tissue factor pathway inhibitor (TFPI) and antithrombin III (AT-III). TFPI serves to attenuate the activity of the extrinsic tenase trigger of coagulation. AT-III directly inhibits thrombin, FIXa, and FXa. The accessory pathway provides an alternate route for the generation of FIXa. Thrombin has also been shown to activate FXI. The fibrin clot is eventually degraded by plasmin, yielding soluble fibrin peptides. Factor Xa inhibitors (apixaban, edoxaban, and rivaroxaban) act by binding to the active site of factor Xa, inhibiting the conversion of prothrombin to thrombin, the final enzyme in the coagulation cascade. Dabigatran, conversely, functions as a direct thrombin inhibitor. It binds with high affinity to the active site of thrombin, inhibiting its ability to convert fibrinogen to fibrin, thereby preventing clot formation.</p>
Full article ">Figure 2
<p>Schematic representation of structural domains of FXa and binding sites of FXa inhibitors. A schematic representation of FXa protein structural domains and the location of the binding sites of FXa inhibitors (apixaban, edoxaban, and rivaroxaban) are depicted. (<b>A</b>) illustrates the domain organization of FXa, highlighting the serine protease domain in the heavy chain (indicated by green) where the binding site is located. (<b>B</b>–<b>D</b>) depict the structures of apixaban, edoxaban, and rivaroxaban, respectively, as obtained from the Protein Data Bank, indicating their binding sites with FXa, which exhibit enzyme kinetics similar to competitive inhibitors.</p>
Full article ">Figure 3
<p>Schematic representation of PAR1- and PAR2-mediated signal transduction. PAR1 and PAR2 are G protein-coupled receptors that can be activated by thrombin and FXa, initiating a cascade of cellular responses. Upon cleavage, PAR1/2 interact with different G proteins like Gα<sub>i</sub>, Gα<sub>12/13</sub>, Gα<sub>s</sub>, and Gα<sub>q</sub>. Gα<sub>12/13</sub> leads to Ras homolog family member A (RhoA) activation, via Rho guanine nucleotide exchange factors (RhoGEFs) influencing cell hypertrophy. Gα<sub>q</sub> activates phospholipase C-β, generating second messengers that trigger calcium release and Protein Kinase C (PKC) activation. PKC can further activate the nuclear factor kappa B (NF-κB) signaling pathway to upregulate production of SRY-box transcription factor 4 (SOX4) and A disintegrin and metalloproteinase with thrombospondin motifs 5 (ADAMTS5). Gα<sub>i</sub> can inhibit adenylate cyclase (AC) to regulate downstream cAMP, whereas Gα<sub>s</sub> can increase cAMP. β-arrestin can activate the ERK1/2 signaling pathway but exhibits inhibitory effects on PKC and calcium release. Anticoagulants like dabigatran (thrombin inhibitor) and apixaban, edoxaban, and rivaroxaban (FXa inhibitors) can potentially disrupt this signaling by preventing PAR activation. Ultimately, these signal transduction pathways can trigger physiological changes like inflammatory and immune responses, cell hypertrophy, and cell migration.</p>
Full article ">Figure 4
<p>Schematic representation of PAR4-mediated signal transduction. PAR4 can activate signaling pathways involving Gα<sub>12/13</sub> and Gα<sub>q</sub>. Gα<sub>12/13</sub> prompts RhoGEFs to activate RhoA, while Gα<sub>q</sub> -Phospholipase C- β (PLC- β) leads to downstream effects such as upregulation of inositol triphosphate (IP3) and diacylglycerol (DAG), resulting in calcium alterations and PKC upregulation, which ultimately leads to activation of the NF-κB signaling pathway. Additionally, β-arrestin can facilitate ERK1/2 phosphorylation but has inhibitory effects on PKC. Thrombin, known for its ability to cleave PAR4, can influence these pathways, therefore dabigatran (thrombin inhibitor) can modulate signal transduction by attenuating thrombin’s effects. As seen, PAR4 activation can cause physiological alterations such as inflammatory and immune response, endothelial barrier dysfunction, and platelet activation.</p>
Full article ">Figure 5
<p>Proposed mechanism of apixaban’s modulatory effects on factor Xa and associated inflammatory signaling pathways in an osteoarthritic chondrocyte model. This illustration delineates the pathways through which apixaban may exert anti-inflammation in the context of osteoarthritis. Apixaban targets FXa, inhibiting its ability to bind and activate PAR2, which is represented by the red inhibitory line. This intervention most likely attenuates the downstream signaling cascades involved in OA pathophysiology: 1. PAR2 inhibition: The blockage of PAR2 activation by apixaban may ameliorate the downstream signaling events mediated by ERK1/2 that lead to the production of pro-inflammatory cytokines, such as TNF-α and IL-1β, potentially alleviating chronic pain associated with OA. 2. Cytokine modulation: The expected reduction in TNF-α and IL-1β due to apixaban’s action on FXa mitigates the upregulation of molecules like MCP-1, which are involved in monocyte recruitment and the NF-κB signaling pathway, both key contributors to inflammation and osteoclastogenesis. 3. Protein expression: The illustration also indicates the potential effects of apixaban on the expression of regulatory proteins, including SOX4 and ADAMTS5, and their impact on critical components like aggrecan, which is essential for cartilage integrity. 4. Chondrocyte integrity and bone health: By modulating these inflammatory and catabolic pathways, apixaban may help preserve chondrocyte integrity, mitigate the generation of reactive oxygen species (ROS), and contribute to maintaining joint health by potentially impacting the RANK/RANKL pathway, which is crucial for osteoclast activity and bone resorption.</p>
Full article ">
17 pages, 4705 KiB  
Article
TNF Induces Laminin-332-Encoding Genes in Endothelial Cells and Laminin-332 Promotes an Atherogenic Endothelial Phenotype
by Assim Hayderi, Mulugeta Melkie Zegeye, Sare Meydan, Allan Sirsjö, Ashok Kumar Kumawat and Liza U. Ljungberg
Int. J. Mol. Sci. 2024, 25(16), 8699; https://doi.org/10.3390/ijms25168699 - 9 Aug 2024
Viewed by 186
Abstract
Laminins are essential components of the basement membranes, expressed in a tissue- and cell-specific manner under physiological conditions. During inflammatory circumstances, such as atherosclerosis, alterations in laminin composition within vessels have been observed. Our study aimed to assess the influence of tumor necrosis [...] Read more.
Laminins are essential components of the basement membranes, expressed in a tissue- and cell-specific manner under physiological conditions. During inflammatory circumstances, such as atherosclerosis, alterations in laminin composition within vessels have been observed. Our study aimed to assess the influence of tumor necrosis factor-alpha (TNF), a proinflammatory cytokine abundantly found in atherosclerotic lesions, on endothelial laminin gene expression and the effects of laminin-332 (LN332) on endothelial cells’ behavior. We also evaluated the expression of LN332-encoding genes in human carotid atherosclerotic plaques. Our findings demonstrate that TNF induces upregulation of LAMB3 and LAMC2, which, along with LAMA3, encode the LN332 isoform. Endothelial cells cultured on recombinant LN332 exhibit decreased claudin-5 expression and display a loosely connected phenotype, with an elevated expression of chemokines and leukocyte adhesion molecules, enhancing their attractiveness and adhesion to leukocytes in vitro. Furthermore, LAMB3 and LAMC2 are upregulated in human carotid plaques and show a positive correlation with TNF expression. In summary, TNF stimulates the expression of LN332-encoding genes in human endothelial cells and LN332 promotes an endothelial phenotype characterized by compromised junctional integrity and increased leukocyte interaction. These findings highlight the importance of basement membrane proteins for endothelial integrity and the potential role of LN332 in atherosclerosis. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

Figure 1
<p>Laminin–receptor interactions and their consequences. Schematic figure illustrating the interaction between laminins and their receptors, such as integrins, dystroglycans, Lutheran/basal cell adhesion molecule (Lu/BCAM), and melanoma cell adhesion molecule (MCAM/CD146), and the consequences of these interactions on the behavior and phenotype of the cells. Created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>.</p>
Full article ">Figure 2
<p>Major cells and cytokines of atherosclerotic plaques. A schematic figure illustrating the key cellular components and cytokines present within atherosclerotic plaques, including macrophages, vascular smooth muscle cells (VSMCs), and T cells. These cells contribute to the progression of atherosclerosis through various mechanisms. Upon the uptake of modified low-density lipoproteins (LDL), these cells secrete proinflammatory cytokines that act on different cell types, thereby accelerating the disease process. Among these cytokines, TNF is a potent activator of endothelial cells and plays a crucial role in modulating laminin gene expression. The figure also highlights potential inhibitors of these cytokines, offering insight into therapeutic strategies aimed at mitigating atherosclerotic progression. Created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>.</p>
Full article ">Figure 3
<p>TNF alters the mRNA expression of laminin-encoding genes in human endothelial cells. The mRNA expression of <span class="html-italic">LAMA1–5</span> (<b>a</b>–<b>e</b>), <span class="html-italic">LAMB1–4</span> (<b>f</b>–<b>i</b>), and <span class="html-italic">LAMC1</span> and <span class="html-italic">LAMC2</span> (<b>j</b>,<b>k</b>) in human endothelial cells exposed to 50 ng/mL of TNF-α for 4–48 h. The data are presented as mean ± SD of 3 independent experiments. One-way ANOVA followed by Dunnett’s multiple comparison were performed to calculate statistical significance. All the timepoints are compared to CTL. * <span class="html-italic">p</span>-value &lt; 0.05, ** <span class="html-italic">p</span>-value &lt; 0.01, *** <span class="html-italic">p</span>-value &lt; 0.001.</p>
Full article ">Figure 4
<p>TNF alters the protein expression of LN332-encoding genes in human endothelial cells. Protein expression of LN332-encoding genes in HUVECs following treatment with TNF for 48 h (<b>a</b>–<b>d</b>). Figure (<b>a</b>) shows representative cropped Western blot image of LN332 chains, while Figures (<b>b</b>–<b>d</b>) show relative levels of LN332 chains from three independent experiments (CTL = 1 a.u.). Full-length Western blot images are shown in <a href="#app1-ijms-25-08699" class="html-app">Supplementary Figure S1</a>. The data are presented as mean ± SD. Student’s <span class="html-italic">t</span>-test was conducted to calculate statistical significance. * <span class="html-italic">p</span>-value &lt; 0.05.</p>
Full article ">Figure 5
<p>Human endothelial cells cultured on LN332 display irregular shape, appears loosely connected and express less tight junction protein claudin-5. A representative PECAM-1 and VE-cadherin staining of human endothelial cells cultured on uncoated or LN332-coated surfaces for 48 h (<b>a</b>). mRNA and protein expression of claudin-5 in human HUVECs cultured on plastic, LN332, or LN511 for 48 h (<b>b</b>). The data are presented as mean ± SD of three independent experiments. One-way ANOVA and Bonferoni test were performed to assess statistical significance. Cells cultured on laminins are compared to cells cultured on plastic (uncoated). * <span class="html-italic">p</span>-value &lt; 0.05, ** <span class="html-italic">p</span>-value &lt; 0.01.</p>
Full article ">Figure 6
<p>Endothelial cells cultured on LN332 have higher expression and secretion of leukocyte adhesion molecules. mRNA expression of <span class="html-italic">E-selectin</span> (<b>a</b>), <span class="html-italic">ICAM-1</span> (<b>b</b>), <span class="html-italic">VCAM-1</span> (<b>c</b>), and <span class="html-italic">PECAM-1</span> (<b>d</b>) in human endothelial cells cultured on uncoated plastic, LN332, or the normal vascular laminin isoform, LN511, for 48 h. Protein levels of E-selectin (<b>e</b>), ICAM-1 (<b>f</b>), and VCAM-1 (<b>g</b>) in cell lysate and protein levels of ICAM-1 (<b>h</b>) and VCAM-1 (<b>i</b>) in supernatant of endothelial cells cultured on plastic, LN332, or LN511 for 48 h. The data are presented as mean ± SD of three independent experiments. One-way ANOVA and Bonferroni test were performed to calculate statistical significance. * <span class="html-italic">p</span>-value &lt; 0.05, ** <span class="html-italic">p</span>-value &lt; 0.01 and *** <span class="html-italic">p</span>-value &lt; 0.001 comparing cells cultured on laminins to cells cultured on uncoated plastic.</p>
Full article ">Figure 7
<p>Human endothelial cells cultured on LN332 have higher expression and secretion of chemokines. Volcano plot showing significantly up- and downregulated proteins in supernatant of endothelial cells cultured on LN332 in relation to cells cultured on uncoated surface for 48 h as measured by OLINK’s proximity extension assay (<b>a</b>). Red-colored dots indicate proteins that are significantly upregulated, and green-colored dots indicate proteins that are significantly downregulated (Log2FC &lt; 0.58 and false discovery rate 5%). Heatmap showing comparison of proteins detected in supernatant from cells cultured on LN332 with cells cultured on uncoated or LN511-coated surfaces (<b>b</b>). Gene expression of upregulated chemokines in human endothelial cells cultured on plastic, LN332, or LN511 for 48 h determined by qRT-PCR (<b>c</b>–<b>h</b>). OLINK data are presented as mean log2 fold change of four independent experiments. qRT-PCR data are presented as mean ± SD of three independent experiments. One-way ANOVA and Bonferroni test were performed to calculate statistical significance for PCR data, whereas <span class="html-italic">t</span>-test and Benjamini–Hochberg tests were used to evaluate statistical significance for OLINK data. Cells cultured on laminins are compared to cells cultured on plastic (uncoated). ** <span class="html-italic">p</span>-value &lt; 0.01, *** <span class="html-italic">p</span>-value &lt; 0.001.</p>
Full article ">Figure 8
<p>Enrichment analysis predicts that human endothelial cells cultured on LN332 release proteins that regulate leucocyte migration/chemotaxis. Ingenuity Pathway Analysis (IPA) was used to perform enrichment analysis of differentially regulated proteins (Log2FC ± 0.58 and false discovery rate 20%) released from human endothelial cells cultured on LN332 compared to cells cultured on plastic. A bar graph showing the top ten functions (Z-score &gt; 1.5) enriched by LN332 regulated proteins with their respective <span class="html-italic">p</span>-values (−Log10) (<b>a</b>). Proteins that enriched the functions “chemotaxis of leucocytes” (<b>b</b>) “and recruitment of phagocytes” or “migration of monocytes” (<b>c</b>) are presented with their respective predicted impacts on the activation state of the functions. The red color indicates upregulation in release of proteins while green indicates downregulation. Orange lines indicate that a protein leads to predicted activation of function, while yellow lines show disagreement between the state of differentially regulated protein expression and the predicted sate of function. The gray lines indicate that no prediction could be made.</p>
Full article ">Figure 9
<p>Monocytes tend to migrate more towards supernatant from endothelial cells cultured on LN332 and leukocytes adhere more to endothelial cells cultured on LN332. In vitro migration of CD14<sup>+</sup> monocytes towards the supernatant from endothelial cells cultured on plastic, LN332, or LN511 performed in Boyden’s transwell system (<b>a</b>) (n = 5). Adhesion of leukocytes to endothelial cells cultured on plastic, LN332, or LN511 (<b>b</b>) (n = 3). The data are presented as mean ± SD. One-way ANOVA followed by Bonferroni test were performed to calculate statistical significance. * <span class="html-italic">p</span>-value &lt; 0.05 comparing cells cultured on laminins to cells cultured on uncoated plastic.</p>
Full article ">Figure 10
<p>LN332-encoding genes’ transcripts are elevated and correlate with TNF in human carotid atherosclerotic lesions. Gene expression of <span class="html-italic">LAMA3</span> (<b>a</b>), <span class="html-italic">LAMB3</span> (<b>b</b>), and <span class="html-italic">LAMC2</span> (<b>c</b>) in human carotid atherosclerotic tissues and adjacent macroscopically intact tissues (n = 32). Pearson’s correlation of <span class="html-italic">LAMA3</span> (<b>d</b>), <span class="html-italic">LAMB3</span> (<b>e</b>), and <span class="html-italic">LAMC2</span> (<b>f</b>) with <span class="html-italic">TNF</span> in human carotid atherosclerotic lesions (n = 32). Solid line indicates Pearson’s correlation coefficient (r), and dashed line indicates 95% confidence band of the best-fit line. Data are acquired from human carotid atheroma gene expression (accession number, GSE43292). <span class="html-italic">p</span>-value smaller than 0.05 is considered statistically significant.</p>
Full article ">
31 pages, 2324 KiB  
Review
Cardiovascular Protective Properties of GLP-1 Receptor Agonists: More than Just Diabetic and Weight Loss Drugs
by Richard Le, Mau T. Nguyen, Momina A. Allahwala, James P. Psaltis, Chinmay S. Marathe, Jessica A. Marathe and Peter J. Psaltis
J. Clin. Med. 2024, 13(16), 4674; https://doi.org/10.3390/jcm13164674 - 9 Aug 2024
Viewed by 325
Abstract
Owing to their potent glucose-lowering efficacy and substantial weight loss effects, glucagon-like peptide-1 receptor agonists (GLP-1 RAs) are now considered part of the frontline therapeutic options to treat both type 2 diabetes mellitus and nondiabetic overweight/obesity. Stemming from successful demonstration of their cardiometabolic [...] Read more.
Owing to their potent glucose-lowering efficacy and substantial weight loss effects, glucagon-like peptide-1 receptor agonists (GLP-1 RAs) are now considered part of the frontline therapeutic options to treat both type 2 diabetes mellitus and nondiabetic overweight/obesity. Stemming from successful demonstration of their cardiometabolic modulation and reduction of major adverse cardiovascular events in clinical outcome trials, GLP-1 RAs have since been validated as agents with compelling cardiovascular protective properties. Studies spanning from the bench to preclinical and large-scale randomised controlled trials have consistently corroborated the cardiovascular benefits of this pharmacological class. Most notably, there is converging evidence that they exert favourable effects on atherosclerotic ischaemic endpoints, with preclinical data indicating that they may do so by directly modifying the burden and composition of atherosclerotic plaques. This narrative review examines the underlying pharmacology and clinical evidence behind the cardiovascular benefits of GLP-1 RAs, with particular focus on atherosclerotic cardiovascular disease. It also delves into the mechanisms that underpin their putative plaque-modifying actions, addresses existing knowledge gaps and therapeutic challenges and looks to future developments in the field, including the use of combination incretin agents for diabetes and weight loss management. Full article
(This article belongs to the Section Pharmacology)
Show Figures

Figure 1

Figure 1
<p>Intracellular signalling pathways induced by GLP-1 RA binding to GLP-1R in pancreatic β cells. These result in attenuation of endoplasmic reticulum (ER) stress (pink), inhibition of apoptosis (purple), increased insulin exocytosis (green), β cell proliferation (yellow) and improved glucose handling and homeostasis (white). AC: adenylyl cyclase, AKT/PKB: protein kinase B, AMPK: adenosine monophosphate-activated protein kinase, ATF4: activating transcription factor 4, ATP: adenosine triphosphate, B-raf: serine/threonine–protein kinase B-raf, Ca<sup>2+</sup>: calcium ions, cAMP: cyclic adenosine monophosphate, CREB: cAMP-response element binding protein, c-Src: tyrosine–protein kinase Src, DAG: diacylglycerol, dephosph-eIF2α: dephosphorylated eukaryotic initiation factor 2α, EGFR: epidermal growth factor receptor, Epac2: exchange protein activated by cAMP 2, ERK1/2: extracellular signal-regulated kinase 1/2, FOXO1: forkhead box protein O1, GLP-1R: glucagon-like peptide 1 receptor, GLP-1 RA: glucagon-like peptide 1 receptor agonist, HIF-α: hypoxia-inducible factor 1-α, IGF1R: insulin-like growth factor 1 receptor, IGF2: insulin-like growth factor 2, IP3: inositol 1,4,5-trisphosphate, IP3R: inositol 1,4,5-trisphosphate receptor, Irs2: insulin receptor substrate 2, mTOR: mammalian target of rapamycin, PDX-1: pancreatic and duodenal homeobox 1, PI3K: phosphoinositide 3-kinase, PIP2: phosphatidylinositol (4,5)-diphosphate, PLC: phospholipase C, PKA: protein kinase A, Rap1: Ras-proximate-1, RyR: ryanodine receptor, SUR1: sulfonylurea receptor 1, TCF7L2: transcription factor 7 like 2, VGCC: voltage-gated calcium channel, Wnt: wingless-related integration site. Created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>.</p>
Full article ">Figure 2
<p>Summary of the cardiometabolic benefits of GLP-1 RA therapy. Green upward arrow: increases; Red downward arrow, decreases. Created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>.</p>
Full article ">Figure 3
<p>Putative Molecular Mechanisms for Anti-Atherogenic Effects of GLP-1 RAs. ABCA1: ATP-binding cassette transporter A1, ANG-II: angiotensin-II, AMPK: AMP-activated protein kinase, ANP: atrial natriuretic peptide, Arg-1: arginase-1, ATF6: cyclic AMP-dependent transcription factor-6, CCL2: C–C motif chemokine ligand 2, CCR7: C-C chemokine receptor type 7, CD62p: P-selectin, CD163+ Mac: cluster of differentiation 163+ macrophage, cGMP: cyclic guanine monophosphate, CRP: C-reactive protein, eNOS: endothelial nitric oxide synthase, FOXO3a: forkhead transcription factor O subfamily member 3a, HTN: hypertension, iNOS: inducible nitric oxide synthase, IFN-γ: interferon-γ, IL-1β: interleukin-1β, IL-6: interleukin-6, IL-10: interleukin-10, IRE1α: inositol-requiring enzyme 1, JNK: c-Jun N-terminal kinase, KLF2: Krüppel-like factor 2, LDL-C: low-density lipoprotein cholesterol, MAPK: mitogen-activated protein kinase, Mφ: macrophage, M1: M1 macrophage, M2: M2 macrophage, MMP: matrix metalloproteinase, MCP-1: monocyte chemoattractant protein-1, NLRP3: nucleotide-binding domain (NOD)-like receptor protein 3, NO: nitric oxide, PERK: protein kinase R-like ER kinase, PKG: protein kinase G, RAGE: receptor for advanced glycation end-products, ROS: reactive oxygen species, SIRT1: sirtuin 1, STAT1: signal transducer and activator of transcription 1, STAT3: signal transducer and activator of transcription 3, TAG: triacylglyceride, TIMP: tissue inhibitor of metalloproteinases, TNF-α: tumour necrosis factor-α, VASP: vasodilator-stimulated phosphoprotein, VCAM1: vascular cell adhesion protein 1, VSMC: vascular smooth muscle cell. Green upward arrow, increases. Red downward arrow, reduces. Red cross, inhibits. Created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>.</p>
Full article ">
14 pages, 679 KiB  
Article
VEGF Polymorphisms (VEGF-936 C/T, VEGF-634 G/C and VEGF-2578 C/A) and Cardiovascular Implications in Long COVID Patients
by Angela Cozma, Adela Viviana Sitar-Tăuț, Olga Hilda Orășan, Violeta Briciu, Daniel Leucuța, Nicolae-Dan Sporiș, Andrada-Luciana Lazăr, Toma-Vlad Mălinescu, Andreea-Maria Ganea, Bianca Mihaela Sporiș, Călin Vasile Vlad, Mihaela Lupșe, Mădălina-Gabriela Țâru and Lucia Maria Procopciuc
Int. J. Mol. Sci. 2024, 25(16), 8667; https://doi.org/10.3390/ijms25168667 - 8 Aug 2024
Viewed by 305
Abstract
The COVID-19 pandemic has raised awareness of the virus’s long-term non-pulmonary consequences. This study examined the relationship between genetic polymorphisms of VEGF and cardiac dysfunction and subclinical atherosclerosis in patients recovering from COVID-19. This study included 67 patients previously diagnosed with COVID-19. VEGF-936C/T, [...] Read more.
The COVID-19 pandemic has raised awareness of the virus’s long-term non-pulmonary consequences. This study examined the relationship between genetic polymorphisms of VEGF and cardiac dysfunction and subclinical atherosclerosis in patients recovering from COVID-19. This study included 67 patients previously diagnosed with COVID-19. VEGF-936C/T, VEGF-634G/C, and VEGF-2578C/A statuses were determined. Conventional echocardiography and arterial parameters assessments were performed at inclusion and at six months after the first assessment. For VEGF-936C/T, dominant and over-dominant models showed a significant increase in ejection fraction at six months after COVID (p = 0.044 and 0.048) and was also a predictive independent factor for the augmentation index (β = 3.07; p = 0.024). The dominant model showed a rise in RV-RA gradient (3.702 mmHg) (p = 0.028 95% CI: 0.040–7.363), with the over-dominant model indicating a greater difference (4.254 mmHg) (p = 0.025 95% CI: 0.624–7.884). The findings for VEGF-634G/C were not statistically significant, except for a difference in TAPSE during initial evaluation, using the codominant model. For VEGF-2578C/A, a difference in ventricular filling pressure (E/E’ratio) was best described under the recessive model. Our research suggests that the VEG-936C/T genotype may impact the baseline level and subsequent changes in cardiac function and subclinical atherosclerosis. These findings offer valuable insights into the complex correlation between genetic polymorphisms and cardiovascular disfunction in long COVID patients. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

Figure 1
<p><span class="html-italic">VEGF-936C/T</span> polymorphism identification. Lane 1—pBRHaeIIIDigest DNA molecular marker; lanes 2, 3,6,9,10,12,20—CC genotype, fragment of 208 bp; lanes 4,5,7,13,14,15,16,17,18,19—CT genotype, fragments of 288 bp, 122 bp, 86 bp; lane 8,11—TT genotype, fragments of 122 bp and 86 bp.</p>
Full article ">Figure 2
<p><span class="html-italic">VEGF-634G/C</span> polymorphism identification. Lane 1—pBRHaeIIIDigest DNA molecular marker; lanes 2—PCR product, fragment of 304 bp; lanes 3,4,6,8—GC genotype, fragments of 304 bp, 193 bp, 111 bp; lanes 5,7—GG genotype, fragments of 193 bp and 111 bp.</p>
Full article ">Figure 3
<p><span class="html-italic">VEGF-2578C/A</span> polymorphism identification. Lane 1—pBRHaeIIIDigest DNA molecular marker; lanes 2,6,7,8—CC genotype, fragment of 324 bp; lanes 3—CA genotype, fragments of 324 bp, 202 bp, 122 bp; lanes 4,5—AA genotype, fragments of 202 bp and 122 bp.</p>
Full article ">
12 pages, 498 KiB  
Article
The Evaluation Value of Non-Invasive Indices of Arterial Stiffness in the Early Stage of Coronary Artery Disease: Preliminary Results from an Exploratory Study
by Fei Wang, Hui Zhang, Kotaro Uchida, Takuya Sugawara, Shintaro Minegishi, Hiroshi Doi, Rie Nakashima-Sasaki, Lin Chen and Tomoaki Ishigami
J. Vasc. Dis. 2024, 3(3), 278-289; https://doi.org/10.3390/jvd3030022 - 8 Aug 2024
Viewed by 148
Abstract
Background: Recently, the arterial velocity pulse index (AVI) and arterial pressure volume index (API) have been used to evaluate arterial stiffness and endothelial function. As arterial stiffness and endothelial injury are risk factors for coronary artery disease (CAD), these two indexes are therefore [...] Read more.
Background: Recently, the arterial velocity pulse index (AVI) and arterial pressure volume index (API) have been used to evaluate arterial stiffness and endothelial function. As arterial stiffness and endothelial injury are risk factors for coronary artery disease (CAD), these two indexes are therefore expected to predict and evaluate the future risk of CAD and cardiovascular events before clinical manifestations. Methods: A total of 90 consecutive patients with coronary angiography (CAG) were enrolled. After excluding normal patients and acute coronary syndrome patients, forty-seven patients with CAD and thirty-two patients with coronary atherosclerosis, and baseline characteristics data were collected. A multifunctional blood pressure monitoring device, AVE-1500 (Shisei Datum, Tokyo, Japan), was used to measure the AVI and API before CAG, and immediately and 2 h, 24 h, and 48 h after CAG and (or) PCI in all the selected participants. Results: After adjusting for various variables using stepwise multiple linear regression analyses, we found that the AVI in the CAD subjects was significantly higher than that in the coronary atherosclerosis subjects before CAG (p = 0.02), immediately after CAG/PCI (p = 0.01), and 48 h after CAG/PCI (p = 0.01), whereas the AVI decreased 24–48 h rather than immediately after CAG/PCI in the CAD group. Moreover, we also found that the API clearly changed in both groups during the periprocedural period of CAG (p = 0.01). Conclusions: In accordance with the results, we propose that the API and AVI may be useful for predicting the early stage of CAD and may be promising as indicators to assess the effect of early revascularization. Full article
(This article belongs to the Section Peripheral Vascular Diseases)
Show Figures

Figure 1

Figure 1
<p>The distribution and comparison of AVI and API measurement at different times between two groups. (<b>A</b>): After adjusting for confounders and variables with statistically significant differences in the univariate analysis by multivariate linear regression, there also were statistically significant differences between the preprocedural AVI, immediately postprocedural AVI, and 48 h postprocedural AVI in the two groups; (<b>B</b>): The univariate analysis showed that the API in the CAD group was significantly higher than in the non-CAD group in the 2 h postprocedure (<span class="html-italic">p</span> = 0.019), however, after adjusting for confounders and variables with statistically significant differences in the univariate analysis by multivariate linear regression, the difference in the 2 h postprocedural API between the two groups could not be considered statistically significant. * <span class="html-italic">p</span> &lt; 0.05. CAD, coronary artery disease; AVI, arterial velocity pulse index; API, arterial pressure volume index.</p>
Full article ">
16 pages, 1050 KiB  
Review
Neural Regulation of Vascular Development: Molecular Mechanisms and Interactions
by Yu Zhang, Xinyu Shen, Shunze Deng, Qiurong Chen and Bing Xu
Biomolecules 2024, 14(8), 966; https://doi.org/10.3390/biom14080966 - 8 Aug 2024
Viewed by 246
Abstract
As a critical part of the circulatory system, blood vessels transport oxygen and nutrients to every corner of the body, nourishing each cell, and also remove waste and toxins. Defects in vascular development and function are closely associated with many diseases, such as [...] Read more.
As a critical part of the circulatory system, blood vessels transport oxygen and nutrients to every corner of the body, nourishing each cell, and also remove waste and toxins. Defects in vascular development and function are closely associated with many diseases, such as heart disease, stroke, and atherosclerosis. In the nervous system, the nervous and vascular systems are intricately connected in both development and function. First, peripheral blood vessels and nerves exhibit parallel distribution patterns. In the central nervous system (CNS), nerves and blood vessels form a complex interface known as the neurovascular unit. Second, the vascular system employs similar cellular and molecular mechanisms as the nervous system for its development. Third, the development and function of CNS vasculature are tightly regulated by CNS-specific signaling pathways and neural activity. Additionally, vascular endothelial cells within the CNS are tightly connected and interact with pericytes, astrocytes, neurons, and microglia to form the blood–brain barrier (BBB). The BBB strictly controls material exchanges between the blood and brain, maintaining the brain’s microenvironmental homeostasis, which is crucial for the normal development and function of the CNS. Here, we comprehensively summarize research on neural regulation of vascular and BBB development and propose directions for future research. Full article
Show Figures

Figure 1

Figure 1
<p>Similarity between the nervous and vascular systems. (<b>A</b>). Two models of neurovascular congruency. In the one-patterns-the-other model, the nerves and blood vessels secrete different factors that mutually guide their growth. In the independent patterning model, the nerves and blood vessels respond to the same signals, but grow independently, finally forming the neurovascular congruency according to the gradient of these factors. (<b>B</b>). Similar cellular structures of axons and endothelial cells during directional growth. Both the growth cone (specialized structure of axonal terminal) and the endothelial tip cell (specialized endothelial cell) contain actin-enriched filopodia to explore the surrounding microenvironment. They can grow or retract according to attractive or repulsive cues, finally guiding the correct growth of axons or blood vessels. (<b>C</b>). The functions of four classical axon guidance cues in endothelial cells. Through different receptors, axon guidance cues exert different functions on the vascular development.</p>
Full article ">Figure 2
<p>Neural regulation of CNS vascular and BBB development. Different cell types in the CNS regulate CNS vascular development and BBB permeability by secreting different factors. Neural activity can also affect CNS vascular development and BBB permeability according to the intensity of neural activity and developmental period. NPCs regulate brain vascular development via secreting different angiogenic factors, for example Wnt, VEGF, etc. The integrin αVβ8 in NPCs is also critical for brain vascular development. Astrocytes mainly regulate BBB development and maintenance via secreting a variety of factors, including VEGF, Shh, angiopoietin, angiotensin, RA, etc. Microglia also can regulate the brain vascular development and repairment via VEGF-C, and Notch signaling pathways. Neurons can regulate brain vascular development and function through secreting different factors, including VEGF, Nogo-A, Semaphorin-3A, glutamate, Norrin, etc. Furthermore, neural activity also plays important roles in brain vascular and BBB development and function.</p>
Full article ">
10 pages, 232 KiB  
Review
Artificial Intelligence and Health Inequities in Dietary Interventions on Atherosclerosis: A Narrative Review
by Dominique J. Monlezun and Keir MacKay
Nutrients 2024, 16(16), 2601; https://doi.org/10.3390/nu16162601 - 7 Aug 2024
Viewed by 682
Abstract
Poor diet is the top modifiable mortality risk factor globally, accounting for 11 million deaths annually with half being from diet-linked atherosclerotic cardiovascular disease (ASCVD). Yet, most of the world cannot afford a healthy diet—as the hidden costs of the inadequate global food [...] Read more.
Poor diet is the top modifiable mortality risk factor globally, accounting for 11 million deaths annually with half being from diet-linked atherosclerotic cardiovascular disease (ASCVD). Yet, most of the world cannot afford a healthy diet—as the hidden costs of the inadequate global food system total over USD 13 trillion annually—let alone the much more clinically, financially, and ecologically costly and resource-intensive medical interventions required to address the disease progression and acute complications of ASCVD. Yet, AI is increasingly understood as a force multiplying revolutionary technology which may catalyze multi-sector efforts in medicine and public health to better address these significant health challenges. This novel narrative review seeks to provide the first known overview of the state-of-the-art in clinical interventions and public health policies in healthy diets for ASCVD, accelerated by health equity-focused AI. It is written from the first-hand practitioner perspective to provide greater relevance and applicability for health professionals and data scientists. The review summarizes the emerging trends and leading use cases in population health risk stratification and precision public health, AI democratizing clinical diagnosis, digital twins in precision nutrition, and AI-enabled culinary medicine as medical education and treatment. This review may, therefore, help inform and advance the evidence-based foundation for more clinically effective, financially efficient, and societally equitable dietary and nutrition interventions for ASCVD. Full article
(This article belongs to the Special Issue Impact of Diet Behavior and Nutrition Intake on Atherosclerosis)
20 pages, 5105 KiB  
Review
Beyond the Obstructive Paradigm: Unveiling the Complex Landscape of Nonobstructive Coronary Artery Disease
by Andreea Tudurachi, Larisa Anghel, Bogdan-Sorin Tudurachi, Alexandra Zăvoi, Alexandr Ceasovschih, Radu Andy Sascău and Cristian Stătescu
J. Clin. Med. 2024, 13(16), 4613; https://doi.org/10.3390/jcm13164613 - 7 Aug 2024
Viewed by 349
Abstract
Traditionally focused on obstructive atherosclerosis, contemporary research indicates that up to 70% of patients undergoing coronary angiography for angina and ischemic symptoms do not exhibit significant stenoses. Nonobstructive coronary artery disease (CAD) has emerged as a prevalent phenotype among these patients. This review [...] Read more.
Traditionally focused on obstructive atherosclerosis, contemporary research indicates that up to 70% of patients undergoing coronary angiography for angina and ischemic symptoms do not exhibit significant stenoses. Nonobstructive coronary artery disease (CAD) has emerged as a prevalent phenotype among these patients. This review emphasizes the emerging understanding that nonobstructive coronary artery disease, encompassing conditions such as ANOCA (Angina with No Obstructive Coronary Artery Disease), INOCA (Ischemia with No Obstructive Coronary Artery Disease), and MINOCA (Myocardial Infarction with No Obstructive Coronary Arteries), represents the most prevalent phenotype in cardiac patients. It delves into the complex pathophysiology underlying these conditions, focusing on microvascular dysfunction and coronary vasoreactivity, which contribute to myocardial ischemia despite the absence of significant coronary obstructions. Additionally, the review critically examines the limitations of current treatments which primarily target obstructive lesions and underscores the necessity for tailored therapies that address the specific microvascular and immunoinflammatory pathways involved in nonobstructive CAD. The main focus of this review is to advocate for a shift in diagnostic and therapeutic strategies to better identify and manage this widely prevalent yet under-recognized subset of CAD. Full article
(This article belongs to the Section Cardiology)
Show Figures

Figure 1

Figure 1
<p>Physiopathology of non-obstructive coronary artery disease. ANOCA, angina with no obstructive coronary artery disease; INOCA, ischemia with no obstructive coronary artery disease; MINOCA, myocardial infarction with no obstructive coronary arteries.</p>
Full article ">Figure 2
<p>Diagnostic algorithm for invasive functional evaluation in patients with non-obstructive coronary artery disease. Ach—acetylcholine; CAD—coronary artery disease; CFR—coronary flow reserve; FFR—fractional flow reserve; IMR—index of microvascular resistance. Adapted from [<a href="#B1-jcm-13-04613" class="html-bibr">1</a>].</p>
Full article ">Figure 3
<p>Physiopathology of coronary artery spasm. Ach—acetylcholine; CaM—calmodulin; ET-1—endothelin-1; NO—nitric oxide; MLC—myosin light chain; MLCK—myosin light chain kinase; PLC—phospholipase C; PKC—protein kinase C; ROS—reactive oxygen species; RhoA—Ras homolog gene member A; VSMCs—vascular smooth muscle cells.</p>
Full article ">Figure 4
<p>Physiopathology of coronary microvascular dysfunction. ET-1—endothelin-1; EDHF—endothelium-dependent hyperpolarization factors; NO—nitric oxide; eNOS—endothelial NO synthase; H2O2—hydrogen peroxide; ROS—reactive oxygen species; RhoA—Ras homolog gene member A; VSMCs—vascular smooth muscle cells; WSS—wall shear stress.</p>
Full article ">
16 pages, 5036 KiB  
Review
Exploring the Spectrum of Long Non-Coding RNA CARMN in Physiological and Pathological Contexts
by Hui Li, Chuannan Sun, Bin Luo, Chuzhi Zhan, Weitao Li, Lu Deng, Kang Kang and Deming Gou
Biomolecules 2024, 14(8), 954; https://doi.org/10.3390/biom14080954 - 6 Aug 2024
Viewed by 330
Abstract
Cardiac mesoderm enhancer-associated non-coding RNA (CARMN), an evolutionarily conserved long non-coding RNA (lncRNA), serves as the host gene for the miR143/145 cluster. It plays a crucial role in cardiovascular cell differentiation and the maintenance of vascular smooth muscle cell (VSMC) homeostasis, which are [...] Read more.
Cardiac mesoderm enhancer-associated non-coding RNA (CARMN), an evolutionarily conserved long non-coding RNA (lncRNA), serves as the host gene for the miR143/145 cluster. It plays a crucial role in cardiovascular cell differentiation and the maintenance of vascular smooth muscle cell (VSMC) homeostasis, which are vital for normal physiological processes. Specifically, CARMN is associated with the pathological progression of cardiovascular diseases such as atherosclerosis, abdominal aortic aneurysm, and chronic heart failure. Moreover, it acts as a tumor suppressor in various cancers, including hepatocellular carcinoma, bladder cancer, and breast cancer, highlighting its potential as a beneficial biomarker and therapeutic target. This review provides a detailed examination of the roles of CARMN, its evolutionary conservation, expression patterns, and regulatory mechanisms. It also outlines its significant implications in the diagnosis, prognosis, and treatment of these diseases, underscoring the need for further translational research to exploit its clinical potential. Full article
Show Figures

Figure 1

Figure 1
<p>A schematic representation of the CARMN genomic loci in humans, mice, and rats. (<b>A</b>,<b>B</b>) The human CARMN locus encodes 25 isoforms (<b>A</b>), while the mouse counterpart produces 7 isoforms, according to the Ensembl database (<b>B</b>). (<b>C</b>) LncRNA LOC120098113, co-located with miR-143/145, is suggested to be the unannotated CARMN gene in rats, as indicated in the Ensembl database, yielding 5 isoforms.</p>
Full article ">Figure 1 Cont.
<p>A schematic representation of the CARMN genomic loci in humans, mice, and rats. (<b>A</b>,<b>B</b>) The human CARMN locus encodes 25 isoforms (<b>A</b>), while the mouse counterpart produces 7 isoforms, according to the Ensembl database (<b>B</b>). (<b>C</b>) LncRNA LOC120098113, co-located with miR-143/145, is suggested to be the unannotated CARMN gene in rats, as indicated in the Ensembl database, yielding 5 isoforms.</p>
Full article ">Figure 2
<p>Physiological roles of CARMN. CARMN plays a pivotal role in the regulation of CPC specification, maintenance of homeostasis in BMSC and MaSC, and modulation of SMC and DPC differentiation. CM: cardiomyocyte; VSMC: vascular smooth muscle cell; MaSC: mammary gland stem cell; DPC: dental pulp cell.</p>
Full article ">Figure 3
<p>Role of CARMN in modulating molecular pathways across cardiovascular, gastrointestinal, and respiratory diseases. It elucidates the diverse roles of CARMN in influencing pathophysiological processes in atherosclerosis, abdominal aortic aneurysm, chronic heart failure, Hirschsprung disease, and airway stenosis. The specific molecular pathways and interactions implicated in these conditions are detailed. The red arrow signifies upregulation, while the blue arrow denotes downregulation.</p>
Full article ">Figure 4
<p>Impact of CARMN on tumor pathophysiology and molecular interactions. The role of CARMN as a tumor suppressor is depicted through its interactions with miRNAs and critical signaling pathways. The figure illustrates how CARMN modulates cancer cell proliferation, migration, and invasion across various tumor types. The red arrow signifies upregulation, while the blue arrow denotes downregulation.</p>
Full article ">
16 pages, 732 KiB  
Review
Age-Associated Calcification: Insights from Murine Models
by Sonia Nasi, Mario Romani and Nathalie Busso
Gout Urate Cryst. Depos. Dis. 2024, 2(3), 236-251; https://doi.org/10.3390/gucdd2030018 - 6 Aug 2024
Viewed by 400
Abstract
Calcification refers to the deposition of calcium-containing crystals either intracellularly or within the extracellular matrix. Physiologic calcification is a normal process occurring during bone and tooth development and growth. In contrast, pathologic calcification occurs in soft tissues that typically do not undergo mineralization, [...] Read more.
Calcification refers to the deposition of calcium-containing crystals either intracellularly or within the extracellular matrix. Physiologic calcification is a normal process occurring during bone and tooth development and growth. In contrast, pathologic calcification occurs in soft tissues that typically do not undergo mineralization, such as blood vessels, cartilage, tendons, and skin. Pathological calcification is significantly associated with tissue impairment and the development of secondary diseases, such as atherosclerosis, osteoarthritis, tendinopathy, and skin ulcers. Aging, a natural process linked to numerous pathologic conditions, is one of the most recognized risk factors for pathological calcification. In this manuscript, we review the current state of knowledge regarding the role of aging in calcification across different tissues. We focus on the mechanisms activated during normal aging, including cellular senescence, decreased pyrophosphate levels, increased secretion of extracellular vesicles, elevated oxidative stress, and higher levels of pro-mineralizing cytokines, all of which can contribute to pathological calcification. Finally, we discuss the available animal models used to study the impact of aging on calcification. Full article
Show Figures

Figure 1

Figure 1
<p>Calcium-containing crystal formation in calcification-competent cells. The extracellular concentration of PP<sub>i</sub> and P<sub>i</sub> is tightly regulated by different enzymes and transporters. This leads to either CPP or BCP crystal formation. The triggers of pathologic calcification include alteration of PPi, Pi, and Ca<sup>2+</sup> homeostasis, impaired mitochondrial homeostasis, blockage of DNA reparation, a jammed cell cycle, compromised autophagy and proteostasis, deregulation of the circadian rhythm, and an increase in pro-mineralization cytokines.</p>
Full article ">
26 pages, 1510 KiB  
Review
The Role of Antioxidants in the Therapy of Cardiovascular Diseases—A Literature Review
by Ewelina Młynarska, Joanna Hajdys, Witold Czarnik, Piotr Fularski, Klaudia Leszto, Gabriela Majchrowicz, Wiktoria Lisińska, Jacek Rysz and Beata Franczyk
Nutrients 2024, 16(16), 2587; https://doi.org/10.3390/nu16162587 - 6 Aug 2024
Viewed by 968
Abstract
Antioxidants are endogenous and exogenous substances with the ability to inhibit oxidation processes by interacting with reactive oxygen species (ROS). ROS, in turn, are small, highly reactive substances capable of oxidizing a wide range of molecules in the human body, including nucleic acids, [...] Read more.
Antioxidants are endogenous and exogenous substances with the ability to inhibit oxidation processes by interacting with reactive oxygen species (ROS). ROS, in turn, are small, highly reactive substances capable of oxidizing a wide range of molecules in the human body, including nucleic acids, proteins, lipids, carbohydrates, and even small inorganic compounds. The overproduction of ROS leads to oxidative stress, which constitutes a significant factor contributing to the development of disease, not only markedly diminishing the quality of life but also representing the most common cause of death in developed countries, namely, cardiovascular disease (CVD). The aim of this review is to demonstrate the effect of selected antioxidants, such as coenzyme Q10 (CoQ10), flavonoids, carotenoids, and resveratrol, as well as to introduce new antioxidant therapies utilizing miRNA and nanoparticles, in reducing the incidence and progression of CVD. In addition, new antioxidant therapies in the context of the aforementioned diseases will be considered. This review emphasizes the pleiotropic effects and benefits stemming from the presence of the mentioned substances in the organism, leading to an overall reduction in cardiovascular risk, including coronary heart disease, dyslipidaemia, hypertension, atherosclerosis, and myocardial hypertrophy. Full article
(This article belongs to the Special Issue Diet, Nutrition and Cardiovascular Health)
Show Figures

Figure 1

Figure 1
<p>Selected sources of reactive oxygen species [<a href="#B11-nutrients-16-02587" class="html-bibr">11</a>]. Abbreviations: ROS—reactive oxygen species; NADPH—nicotinamide adenine dinucleotide phosphate; NO—nitric oxide; ER—endoplasmic reticulum.</p>
Full article ">Figure 2
<p>Existing forms of CoQ10.</p>
Full article ">Figure 3
<p>Chemical structures of the different classes of polyphenols. Extracted and modified from [<a href="#B36-nutrients-16-02587" class="html-bibr">36</a>].</p>
Full article ">Figure 4
<p>Flavonoids’ properties.</p>
Full article ">
20 pages, 3448 KiB  
Review
Inflammation: Is It a Healer, Confounder, or a Promoter of Cardiometabolic Risks?
by Amit R. Tate and Gundu H. R. Rao
Biomolecules 2024, 14(8), 948; https://doi.org/10.3390/biom14080948 - 6 Aug 2024
Viewed by 380
Abstract
Inflammation is the body’s non-specific response to injury or infection. It is a natural defense mechanism that helps to maintain homeostasis and promotes tissue repair. However, excessive inflammation can lead to cellular, tissue, or organ dysfunction, as well as contribute to the development [...] Read more.
Inflammation is the body’s non-specific response to injury or infection. It is a natural defense mechanism that helps to maintain homeostasis and promotes tissue repair. However, excessive inflammation can lead to cellular, tissue, or organ dysfunction, as well as contribute to the development of acute vascular events and diseases like Crohn’s disease, psoriasis, obesity, diabetes, and cancer. The initial response to injury involves the activation of platelets and coagulation mechanisms to stop bleeding. This is followed by the recruitment of immune cells and the release of cytokines to promote tissue repair. Over time, the injured tissue undergoes remodeling and returns to its pre-injury state. Inflammation is characterized by the activation of inflammatory signaling pathways involving cytokines, chemokines, and growth factors. Mast cells play a role in initiating inflammatory responses. Pattern recognition receptors (PRRs) such as Toll-like receptors (TLRs) and nucleotide-binding domain (NOD)-like receptors (NLRs) are involved in the activation of these inflammatory pathways. Inflammasomes, which are cytoplasmic complexes, also contribute to inflammation by activating cytokines. Inflammation can also be triggered by factors like dietary components and the composition of the gut microbiota. Dysregulation of the gut microbiome can lead to excessive inflammation and contribute to diseases like atherosclerosis and irritable bowel syndrome (IBS). The immune system and gut-associated lymphoid tissue (GALT) play crucial roles in the inflammatory response and the development of conditions like colorectal cancer. Anti-inflammatory therapy can play a significant role in reducing or inducing the remission of inflammatory diseases such as Crohn’s disease and ulcerative colitis. The fetal origin of adult diseases theory suggests that conditions during fetal development, such as low birth weight and maternal obesity, can influence the risk of cardiometabolic diseases later in life. All of the known risk factors associated with cardiometabolic diseases such as hypertension, excess weight, obesity, type-2 diabetes, and vascular diseases are accompanied by chronic low-grade inflammation. Inflammation seems to have a role in precipitating even acute vascular events such as heart attacks and stroke. Common markers of inflammation associated with cardiometabolic disease include interleukin (IL)-1β, IL-6, tumor necrosis factor (TNF-α), C-reactive protein (CRP), and soluble TNF receptors such as sTNFR1 and sTNFR2. These markers serve as indicators of systemic inflammation. However, these markers are not disease-specific but provide an insight into the overall chronic inflammatory status. In fact, inflammation has been identified as a potential target for future treatments to reduce or reverse the risk of atherosclerosis-related complications. The regulation of inflammation is complex, and further research is needed to better understand its mechanisms and develop strategies for managing inflammatory disorders. In summary, inflammation is a natural response to injury or infection, but excessive or prolonged inflammation can lead to the progression of various diseases. Understanding the underlying mechanisms of inflammation is important for developing treatments and preventive measures for inflammatory disorders. Full article
(This article belongs to the Special Issue New Insights into Cardiometabolic Diseases)
Show Figures

Figure 1

Figure 1
<p>Schematic representation of wound-healing mechanisms. (University of Minnesota Artists, Personal Collection).</p>
Full article ">Figure 2
<p>Scanning electron micrographs of healthy endothelium and endothelial cells. [Courtesy: (Late) Professor James G White, University of Minnesota.] (<b>A</b>) Healthy endothelium. (<b>B</b>) Two endothelial cells held together.</p>
Full article ">Figure 3
<p>Scanning electron micrographs of platelet interaction with endothelium. [Courtesy of (Late) Professor James G. White, University of Minnesota.] (<b>A</b>) Dysfunctional endothelium. (<b>B</b>) Damaged endothelium.</p>
Full article ">Figure 4
<p>Schematic presentation of altered production of vasoactive molecules. (University of Minnesota, Artists. Personal Collection).</p>
Full article ">Figure 5
<p>Three-dimensional ultrasound images of carotid arteries. (Courtesy: Dr Aaron Fenster, Research Director, Robarts Research Institute, Canada.) (<b>A</b>) Carotid artery with minimal stenosis. (<b>B</b>) Carotid artery with plaque.</p>
Full article ">Figure 6
<p>Thrombus formation on denuded endothelium. (Personal Collection).</p>
Full article ">Figure 7
<p>Altered prostaglandin metabolism in drug-induced diabetic rats. (Courtesy: Dr Jonathan Gerrard, Professor University of Winnipeg, Canada).</p>
Full article ">
13 pages, 2489 KiB  
Article
Conjugated Linoleic Acid Production in Pine Nut Oil: A Lactiplantibacillus plantarum Lp-01 Fermentation Approach
by Gang Wei, Ge Wu, Jiajia Sun, Yi Qi, Qi Zhao, Fengde Xu, Zhi Zhang and Lanzhi Peng
Foods 2024, 13(16), 2472; https://doi.org/10.3390/foods13162472 - 6 Aug 2024
Viewed by 270
Abstract
Conjugated linoleic acid (CLA) is a class of bioactive fatty acids that exhibit various physiological activities such as anti-cancer, anti-atherosclerosis, and lipid-lowering. It is an essential fatty acid that cannot be synthesized by the human body and must be derived from dietary sources. [...] Read more.
Conjugated linoleic acid (CLA) is a class of bioactive fatty acids that exhibit various physiological activities such as anti-cancer, anti-atherosclerosis, and lipid-lowering. It is an essential fatty acid that cannot be synthesized by the human body and must be derived from dietary sources. The natural sources of CLA are limited, predominantly relying on chemical and enzymatic syntheses methods. Microbial biosynthesis represents an environmentally benign approach for CLA production. Pine nut oil, containing 40–60% linoleic acid, serves as a promising substrate for CLA enrichment. In the present study, we developed a novel method for the production of CLA from pine nut oil using Lactiplantibacillus plantarum (L. plantarum) Lp-01, which harbors a linoleic acid isomerase. The optimal fermentation parameters for CLA production were determined using a combination of single-factor and response surface methodologies: an inoculum size of 2%, a fermentation temperature of 36 °C, a fermentation time of 20 h, and a pine nut oil concentration of 11%. Under these optimized conditions, the resultant CLA yield was 33.47 μg/mL. Gas chromatography analysis revealed that the fermentation process yielded a mixture of c9, t11CLA and t10, c12 CLA isomers, representing 4.91% and 4.86% of the total fatty acid content, respectively. Full article
(This article belongs to the Section Food Biotechnology)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Bioinformatic analysis of LAI in <span class="html-italic">L. plantarum Lp-01</span>. (<b>a</b>) Phylogenetic tree analysis of LAI in <span class="html-italic">L. plantarum Lp-01</span>. (<b>b</b>) Tertiary structure prediction of <span class="html-italic">L. plantarum Lp-01</span> LAI by the SWISS-MODEL based on homology modeling.</p>
Full article ">Figure 2
<p>Effects of different fermentation parameters on CLA yield. (<b>a</b>) Effect of inoculum size on CLA yield. (<b>b</b>) Effect of fermentation temperature on CLA yield. (<b>c</b>) Effect of fermentation time on CLA yield. (<b>d</b>) Effect of pine nut oil concentration on CLA yield.</p>
Full article ">Figure 3
<p>Response surface and contour plots utilizing the Box–Behnken design for optimizing CLA yield: (<b>AB</b>) inoculum size vs. fermentation temperature; (<b>AC</b>) inoculum size vs. fermentation time; (<b>AD</b>) inoculum size vs. pine nut oil concentration; (<b>BC</b>) fermentation temperature vs. fermentation time; (<b>BD</b>) fermentation temperature vs. pine nut oil concentration; (<b>CD</b>) fermentation time vs. pine nut oil concentration. A: Inoculum size, B: fermentation temperature, C: fermentation time, D: pine nut oil concentration.</p>
Full article ">Figure 4
<p>Spectral analysis of fermentation extract.</p>
Full article ">Figure 5
<p>Analysis of pine nut oil fermentation products. (<b>a</b>) Gas chromatography chromatogram of conjugated linoleic acid standard samples. (<b>b</b>) Gas chromatography chromatogram of pine nut oil fermentation products.</p>
Full article ">
14 pages, 4196 KiB  
Article
Photodynamic Therapy of Atherosclerotic Plaque Monitored by T1 and T2 Relaxation Times of Magnetic Resonance Imaging
by Piotr Wańczura, David Aebisher, Dawid Leksa, Wiktoria Mytych, Klaudia Dynarowicz, Angelika Myśliwiec, Natalia Leksa, Adrian Truszkiewicz and Dorota Bartusik-Aebisher
Int. J. Transl. Med. 2024, 4(3), 505-518; https://doi.org/10.3390/ijtm4030034 - 5 Aug 2024
Viewed by 193
Abstract
Atherosclerosis, marked by plaque accumulation within arteries, results from lipid dysregulation, inflammation, and vascular remodeling. Plaque composition, including lipid-rich cores and fibrous caps, determines stability and vulnerability. Photodynamic therapy (PDT) has emerged as a promising treatment, leveraging photosensitizers to induce localized cytotoxicity upon [...] Read more.
Atherosclerosis, marked by plaque accumulation within arteries, results from lipid dysregulation, inflammation, and vascular remodeling. Plaque composition, including lipid-rich cores and fibrous caps, determines stability and vulnerability. Photodynamic therapy (PDT) has emerged as a promising treatment, leveraging photosensitizers to induce localized cytotoxicity upon light activation. PDT targets plaque components selectively, reducing burden and inflammation. Challenges remain in optimizing PDT parameters and translating preclinical success to clinical efficacy. Nonetheless, PDT offers a minimally invasive strategy for atherosclerosis management, promising personalized interventions for cardiovascular health. The objective of the current study was to present the findings from quantitative non-contrast MRI of atherosclerosis post-PDT by assessing relaxation times. The study aimed to utilize and optimize a 1.5T MRI system. Clinical scanners were used for MRI examinations. The research involved analyzing T1 and T2 relaxation times. Following treatment of the samples with Rose Bengal and exposure to pure oxygen, PDT irradiation was administered. The results indicated that the therapy impacted the crus, evidenced by a significant decrease in relaxation times in the MRI data. Full article
Show Figures

Figure 1

Figure 1
<p>View of the retrieved vessel after defrosting.</p>
Full article ">Figure 2
<p>Samples with Rose Bengal disodium salt at concentrations of 0.01 mM (No. 1), 0.02 mM (No. 2), 0.03 mM (No. 3), 0.04 mM (No. 4), and 0.05 mM (No. 5).</p>
Full article ">Figure 3
<p>Pre and post samples irradiated at 532 nm for 15 min. Samples with Rose Bengal disodium salt at concentrations of 0.01 mM (No. 1), 0.02 mM (No. 2), 0.03 mM (No. 3), 0.04 mM (No. 4), and 0.05 mM (No. 5).</p>
Full article ">Figure 4
<p>Tesla Optima MR360 MRI used to determine T1 and T2 relaxation times.</p>
Full article ">Figure 5
<p>Determined T1 and T2 relaxation times of atherosclerotic samples before PDT. The yellow box is the area of the Voxel.</p>
Full article ">Figure 6
<p>Determined T1 and T2 relaxation times of atherosclerotic samples after PDT. The yellow box is the area of the Voxel.</p>
Full article ">Figure 7
<p>(<b>a</b>,<b>d</b>) contain the results of mapping the longitudinal relaxation times. They present maps of the distribution of T1 times. The sharp demarcation between the fluid and the examined structures is clearly visible. (<b>b</b>,<b>e</b>) present the distribution of the R<sup>2</sup> coefficient—it is a measure of the fit of the approximating curve describing the measurement data. It is clearly seen that this coefficient is close to “1”. This proves a very good fit. These figures also show a decrease in the R<sup>2</sup> value for regions more distant from the coil plane. This is a characteristic phenomenon because the coil used has the characteristics of a flat-loop coil which produces very good figures in its plane, but when moving away from it, the signal quality decreases and noise increases. This type of coil was chosen due to the geometric characteristics of the tested objects. The aim of the study was to image the structures lying in the plane of the urethra as well as possible. The figures presented in (<b>c</b>,<b>f</b>) are histograms, allowing determination of the quantitative distribution of pixels in the examined figures. The histogram plot is the number of pixels in the image (vertical axis) with a particular brightness value (horizontal axis). The histogram plot is the distribution of the number of pixels according to their intensities, corresponding to the time value that is calculated.</p>
Full article ">Figure 8
<p>(<b>a</b>,<b>d</b>) contain the results of mapping the transverse relaxation times. It should be said that in this case the noise is significantly increased. Very short relaxation times resulted in poorer image quality—the system used for research has limitations regarding the parameters that can be set for TE and TR times. This is most noticeable in the parts of the image showing fluids. The regions imaging the structures being examined are mapped with greater accuracy—their times are significantly longer than the T2 of fluid areas. In the context of the decrease in the quality of fitting the curves to the measurement data, the figures for R<sup>2</sup> (<b>b</b>,<b>e</b>) are very telling, where the fluid areas are dark blue and therefore R<sup>2</sup> is close to the value “0”. (<b>c</b>,<b>f</b>) are histograms showing the distribution of the number of pixels in the examined region. The histogram plot is the number of pixels in the image (vertical axis) with a particular brightness value (horizontal axis). The histogram plot is the distribution of the number of pixels according to their intensities, corresponding to the time value that is calculated.</p>
Full article ">
17 pages, 5065 KiB  
Article
A Mixture of Lactobacillus HY7601 and KY1032 Regulates Energy Metabolism in Adipose Tissue and Improves Cholesterol Disposal in High-Fat-Diet-Fed Mice
by Kippeum Lee, Hyeon-Ji Kim, Joo-Yun Kim, Jae-Jung Shim and Jae-Hwan Lee
Nutrients 2024, 16(15), 2570; https://doi.org/10.3390/nu16152570 - 5 Aug 2024
Viewed by 527
Abstract
We aimed to characterize the anti-obesity and anti-atherosclerosis effects of Lactobacillus curvatus HY7601 and Lactobacillus plantarum KY1032 using high-fat diet (HFD)-fed obese C57BL/6 mice. We divided the mice into control (CON), HFD, HFD with 108 CFU/kg/day probiotics (HFD + KL, HY7301:KY1032 = [...] Read more.
We aimed to characterize the anti-obesity and anti-atherosclerosis effects of Lactobacillus curvatus HY7601 and Lactobacillus plantarum KY1032 using high-fat diet (HFD)-fed obese C57BL/6 mice. We divided the mice into control (CON), HFD, HFD with 108 CFU/kg/day probiotics (HFD + KL, HY7301:KY1032 = 1:1), and HFD with 109 CFU/kg/day probiotics (HFD + KH, HY7301:KY1032 = 1:1) groups and fed/treated them during 7 weeks. The body mass, brown adipose tissue (BAT), inguinal white adipose tissue (iWAT), and epididymal white adipose tissue (eWAT) masses and the total cholesterol and triglyceride concentrations were remarkably lower in probiotic-treated groups than in the HFD group in a dose-dependent manner. In addition, the expression of uncoupling protein 1 in the BAT, iWAT, and eWAT was significantly higher in probiotic-treated HFD mice than in the HFD mice, as demonstrated by immunofluorescence staining and Western blotting. We also measured the expression of cholesterol transport genes in the liver and jejunum and found that the expression of those encoding liver-X-receptor α, ATP-binding cassette transporters G5 and G8, and cholesterol 7α-hydroxylase were significantly higher in the HFD + KH mice than in the HFD mice. Thus, a Lactobacillus HY7601 and KY1032 mixture with 109 CFU/kg/day concentration can assist with body weight regulation through the management of lipid metabolism and thermogenesis. Full article
(This article belongs to the Special Issue Nutritional and Metabolic Changes Affecting Adipose Tissue Biology)
Show Figures

Figure 1

Figure 1
<p>Effects of <span class="html-italic">Lactobacillus</span> HY7601 and KY1032 on the body and tissue masses of the mice. (<b>A</b>) Body masses of the mice after 7 weeks. (<b>B</b>) Images of brown adipose tissue (BAT), inguinal white adipose tissue (inguinal WAT), and epididymal adipose tissue (epididymal WAT) samples. Masses of the (<b>C</b>) epididymal WAT, (<b>D</b>) inguinal WAT, (<b>E</b>) BAT, (<b>F</b>) livers, and (<b>G</b>) spleens of the mice. (<b>H</b>) Food intake and (<b>I</b>) water intake per unit of body weight. Data are expressed as mean ± SD (n = 12). Groups accompanied by different letters were significantly different: <span class="html-italic">p</span> &lt; 0.05 (a &gt; ab &gt; b &gt; bc &gt; c &gt; d). CON, control; HFD, HFD-fed obese mice; HFD-KL, 10<sup>8</sup> CFU/kg/day <span class="html-italic">Lactobacillus</span> HY7601 and KY103 plus HFD; HFD-KH, 10<sup>9</sup> CFU/kg/day <span class="html-italic">Lactobacillus</span> HY7601 and KY103 plus HFD.</p>
Full article ">Figure 2
<p>Histology of adipose tissue depots in each of the mouse groups. (<b>A</b>) Brown adipose tissue (BAT, top), inguinal white adipose tissue (WAT, middle), and epididymal white adipose tissue (bottom), stained with hematoxylin and eosin (scale bar = 50 μm). (<b>B</b>) Number of adipocytes in epididymal WAT and inguinal WAT, determined using ImageJ (version 1.53t). N = 6–8 mice/group. Data are expressed as mean ± SD (n = 12). Groups accompanied by different letters were significantly different: <span class="html-italic">p</span> &lt; 0.05 (a &gt; b &gt; c).</p>
Full article ">Figure 3
<p>Effects of <span class="html-italic">Lactobacillus</span> HY7601 and KY1032 on serum lipid and cholesterol-related parameters in HFD-fed mice. Serum concentrations of (<b>A</b>) adiponectin, (<b>B</b>) triglyceride (TG), (<b>C</b>) total cholesterol (T-Chol), (<b>D</b>) low-density lipoprotein cholesterol (LDL-Chol), (<b>E</b>) high-density lipoprotein cholesterol (HDL-Chol), and (<b>F</b>) blood urea nitrogen (BUN). Data are mean ± SD (n = 12). Groups accompanied by different letters were significantly different: <span class="html-italic">p</span> &lt; 0.05 (a &gt; ab &gt; b &gt; c).</p>
Full article ">Figure 4
<p>Effects of <span class="html-italic">Lactobacillus</span> HY7601 and KY1032 on the rectal temperature and serum parameters related to glucose metabolism in HFD-fed mice. (<b>A</b>) Core temperature, measured using a thermometer. Serum concentrations of (<b>B</b>) glucose, (<b>C</b>) glycated or glycosylated hemoglobin A1c (HbA1C), (<b>D</b>) creatine kinase (CK), and (<b>E</b>) lactate. Data are expressed as mean ± SD (n = 12). Groups accompanied by different letters were significantly different: <span class="html-italic">p</span> &lt; 0.05 (a &gt; ab &gt; b &gt; c).</p>
Full article ">Figure 5
<p>Adipose sections immunostained for uncoupling protein 1 (UCP1) in the various mouse groups. Representative images of (<b>A</b>) BAT and (<b>B</b>) inguinal WAT (bright field, UCP1 (red), DAPI (blue), and merged UCP1 and DAPI).</p>
Full article ">Figure 6
<p>Effects of <span class="html-italic">Lactobacillus</span> HY7601 and KY1032 on the expression of key thermogenic proteins in (<b>A</b>) BAT, (<b>B</b>) inguinal WAT, and (<b>C</b>) epididymal WAT. Western blot data for sirtuin 1 (SirT1), UCP1, phosphorylated-AMP-activated protein kinase (<span class="html-italic">p</span>-AMPK), AMPK, peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α), and glyceraldehyde 3-phosphate dehydrogenase (GAPDH). Groups accompanied by different letters were significantly different: <span class="html-italic">p</span> &lt; 0.05 (a &gt; ab &gt; b &gt; c).</p>
Full article ">Figure 7
<p>Effects of <span class="html-italic">Lactobacillus</span> HY7601 and KY1032 on the circulating concentrations of liver-related enzymes and the liver mRNA expression of the mouse groups. Serum activities of (<b>A</b>) alanine transferase (ALT), (<b>B</b>) aspartate transaminase (AST), and (<b>C</b>) lactate dehydrogenase (LDH). Expression of the genes encoding (<b>D</b>) 3-hydroxy-3-methylglutaryl-coenzyme A reductase (<span class="html-italic">Hmgcr</span>), (<b>E</b>) sterol regulatory element-binding protein 2 (<span class="html-italic">Srdbp2</span>), (<b>F</b>) peroxisome proliferator-activated receptor alpha (<span class="html-italic">PPARa</span>), (<b>G</b>) ATP-binding cassette (ABC) transporter G5 (<span class="html-italic">Abcg5</span>), (<b>H</b>) <span class="html-italic">Agcg8</span>, (<b>I</b>) liver X receptor alpha (<span class="html-italic">LXRb</span>), (<b>J</b>) <span class="html-italic">LXRβ</span>, and (<b>K</b>) cholesterol 7 alpha-hydroxylase (<span class="html-italic">Cyp7a1</span>), normalized to that of <span class="html-italic">Gapdh</span>. Groups accompanied by different letters were significantly different: <span class="html-italic">p</span> &lt; 0.05 (a &gt; ab &gt; b &gt; bc &gt; c).</p>
Full article ">Figure 8
<p>Effects of the <span class="html-italic">Lactobacillus</span> HY7601 and KY1032 mixture on cholesterol-metabolism-related parameters in the jejuna of the mouse groups. Expression of genes encoding (<b>A</b>) ATP-binding cassette (ABC) transporter G5 (<span class="html-italic">Abcg5</span>), (<b>B</b>) <span class="html-italic">Agcg8</span>, (<b>C</b>) liver X receptor alpha (<span class="html-italic">LXRα</span>), and (<b>D</b>) NPC1-like intracellular cholesterol transporter 1 (<span class="html-italic">Npcl1</span>), normalized to that of <span class="html-italic">Gapdh</span>. Fecal concentrations of (<b>E</b>) bile acids and (<b>F</b>) total cholesterol. Groups accompanied by different letters were significantly different: <span class="html-italic">p</span> &lt; 0.05 (a &gt; ab &gt; b &gt; c).</p>
Full article ">
Back to TopTop