[go: up one dir, main page]
More Web Proxy on the site http://driver.im/
Next Issue
Volume 14, November
Previous Issue
Volume 14, September
You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 

Biomolecules, Volume 14, Issue 10 (October 2024) – 142 articles

Cover Story (view full-size image): This paper delves into the mechanistic role of glycosphingolipids (GSLs) in cardiovascular disease, emphasizing their influence on inflammation, energy metabolism, and key signaling pathways that are crucial for heart function. GSLs interact with membrane microdomains, modulate mitochondrial dynamics, and impact lipid homeostasis, driving processes that contribute to heart failure. Through human and animal model studies, this work uncovers how disruptions in GSL metabolism are linked to oxidative stress, cardiac remodeling, and hypertrophy. These findings position GSLs as promising biomarkers and therapeutic targets for advancing cardiovascular treatment strategies. View this paper
  • Issues are regarded as officially published after their release is announced to the table of contents alert mailing list.
  • You may sign up for e-mail alerts to receive table of contents of newly released issues.
  • PDF is the official format for papers published in both, html and pdf forms. To view the papers in pdf format, click on the "PDF Full-text" link, and use the free Adobe Reader to open them.
Order results
Result details
Section
Select all
Export citation of selected articles as:
26 pages, 1923 KiB  
Review
tRNA and tsRNA: From Heterogeneity to Multifaceted Regulators
by Yun Li, Zongyu Yu, Wenlin Jiang, Xinyi Lyu, Ailian Guo, Xiaorui Sun, Yiting Yang and Yunfang Zhang
Biomolecules 2024, 14(10), 1340; https://doi.org/10.3390/biom14101340 - 21 Oct 2024
Viewed by 2283
Abstract
As the most ancient RNA, transfer RNAs (tRNAs) play a more complex role than their constitutive function as amino acid transporters in the protein synthesis process. The transcription and maturation of tRNA in cells are subject to stringent regulation, resulting in the formation [...] Read more.
As the most ancient RNA, transfer RNAs (tRNAs) play a more complex role than their constitutive function as amino acid transporters in the protein synthesis process. The transcription and maturation of tRNA in cells are subject to stringent regulation, resulting in the formation of tissue- and cell-specific tRNA pools with variations in tRNA overall abundance, composition, modification, and charging levels. The heterogeneity of tRNA pools contributes to facilitating the formation of histocyte-specific protein expression patterns and is involved in diverse biological processes. Moreover, tRNAs can be recognized by various RNase under physiological and pathological conditions to generate tRNA-derived small RNAs (tsRNAs) and serve as small regulatory RNAs in various biological processes. Here, we summarize these recent insights into the heterogeneity of tRNA and highlight the advances in the regulation of tRNA function and tsRNA biogenesis by tRNA modifications. We synthesize diverse mechanisms of tRNA and tsRNA in embryonic development, cell fate determination, and epigenetic inheritance regulation. We also discuss the potential clinical applications based on the new knowledge of tRNA and tsRNA as diagnostic and prognostic biomarkers and new therapeutic strategies for multiple diseases. Full article
(This article belongs to the Special Issue Advances in tRNA Biology)
Show Figures

Figure 1

Figure 1
<p>Transfer RNA (tRNA) heterogeneity is manifested in multiple aspects. tRNA heterogeneity is reflected in overall tRNA abundance; the composition of tRNA isotypes, isoacceptors, and isodecoders; the abundance of various RNA modifications; and the aminoacylation levels in cellular tRNA pools, which may be regulated by multiple effectors, such as chromatin accessibility, transcription factor binding, and RNA polymerase III (Pol III) activity, as well as the abundance and activity of tRNA-modifying enzymes and aminoacyl-tRNA synthetases (AARS). The tRNA isotypes are distinguished by different color schemes, and the varying shades within the same color range in-dicate different tRNA isoacceptors. The different colored dots on the tRNA sequence represent different types of RNA modifications. tRNAs are not always fully aminoacylated and that the level varies depending on cell type and growth conditions. Created in BioRender. Yang, Y. (2024) <a href="http://BioRen-der.com/a28u622" target="_blank">BioRen-der.com/a28u622</a> (accessed on 15 August 2024).</p>
Full article ">Figure 2
<p>RNA modification on tRNAs and biogenesis of tsRNAs. (<b>a</b>) Possible tRNA modification types, sites, and corresponding enzymes. (<b>b</b>) 5′tsRNAs generate from D loop nuclease or anticodon loop nuclease; 3′tsRNAs generate from TΨC loop or anticodon loop nuclease; inner’tsRNAs generate from D loop and TΨC loop nuclease.</p>
Full article ">Figure 3
<p>Neural network model for transmitting parental traits to offspring through the sperm RNA code. The paternally acquired phenotypes are encoded as the sperm RNA code, with various features. It is possible that these features may interact with each other, either directly or indirectly, to influence the biological process in the early embryo and subsequently affect the phenotypes of the offspring. Created in BioRender. Yang, Y. (2024) <a href="http://BioRender.com/x33h223" target="_blank">BioRender.com/x33h223</a> (accessed on 15 August 2024).</p>
Full article ">
23 pages, 1998 KiB  
Review
Astragalus membranaceus: A Traditional Chinese Medicine with Multifaceted Impacts on Breast Cancer Treatment
by Zhong Tang and Xuefei Tian
Biomolecules 2024, 14(10), 1339; https://doi.org/10.3390/biom14101339 - 21 Oct 2024
Viewed by 1944
Abstract
Breast cancer, the most prevalent malignant tumor among women globally, remains a critical area of focus for researchers striving to refine therapeutic approaches. As an important component of traditional Chinese medicine, Astragalus membranaceus (AM) has demonstrated potential for multifaceted impacts on breast cancer treatment [...] Read more.
Breast cancer, the most prevalent malignant tumor among women globally, remains a critical area of focus for researchers striving to refine therapeutic approaches. As an important component of traditional Chinese medicine, Astragalus membranaceus (AM) has demonstrated potential for multifaceted impacts on breast cancer treatment through various mechanisms. To guide clinical practice and further explore the under-researched field of AM in breast cancer treatment, this paper mainly reviews the regulatory roles of AM-derived compounds and extracts on breast cancer cell proliferation, migration, invasion, and chemoresistance. Furthermore, this study delves into the synergistic effects observed when AM is co-administered with chemotherapeutic agents, including the enhancement of chemosensitivity, mitigation of toxic side effects, and reversal of drug resistance. This review indicates that AM holds promise not only as a therapy in breast cancer treatment but also paves the way for innovative integrated treatment approaches that combine the benefits of traditional medicine with modern pharmaceuticals. Nevertheless, future research endeavors are also urged to elucidate the in vivo pharmacological effects and underlying mechanisms of AM to inform more effective clinical treatment strategies. Full article
(This article belongs to the Special Issue Antitumor Agents from Natural Sources 2024–2025)
Show Figures

Figure 1

Figure 1
<p>Signal pathways of calycosin in breast cancer. Calycosin can suppress EMT by inhibiting the BATF/TGF-β1 pathway. Calycosin targets the ER and downstream pathways to inhibit the proliferation of cancer cells. In addition, calycosin can promote apoptosis by inducing the Bax/BCL-2 pathway. Orange arrows indicate inhibitory effects, while green arrows represent inducing effects.</p>
Full article ">Figure 2
<p>Signal pathways of formononetin in breast cancer. Formononetin can inhibit the proliferation, autophagy, invasion, and migration of breast cancer cells by suppressing the NF-κB p65, p38, Akt, and p53 signaling pathways. Formononetin can enhance autophagy through the MiR-199a-3p/mTOR pathway. Formononetin can modulate drug resistance by affecting the lncRNA AFAP1-AS1. Orange arrows indicate inhibitory effects, while green arrows represent inducing effects.</p>
Full article ">Figure 3
<p>Signal pathways of astragaloside IV in breast cancer. Astragaloside IV can suppress cell invasion by inhibiting the MAPK signaling pathway and induce TRHDE-ASI to inhibit cell proliferation. Additionally, it can influence FOXO1, promoting the polarization of M2 macrophages. Orange arrows indicate inhibitory effects, while green arrows represent inducing effects.</p>
Full article ">Figure 4
<p>Signal pathways of biochanin A in breast cancer. Biochanin A can suppress the viability, invasion, and growth of breast cancer cells by inhibiting the Erk1/2, mTOR, Akt, NF-κB, and p53 signaling pathway, while it can enhance cell proliferation through the MiR-199a-3p/Era pathway. Orange arrows indicate inhibitory effects, while green arrows represent inducing effects.</p>
Full article ">
16 pages, 2676 KiB  
Article
Cooperative Substructure and Energetics of Allosteric Regulation of the Catalytic Core of the E3 Ubiquitin Ligase Parkin by Phosphorylated Ubiquitin
by Xiang Ye, Sravya Kotaru, Rosana Lopes, Shannen Cravens, Mauricio Lasagna and A. Joshua Wand
Biomolecules 2024, 14(10), 1338; https://doi.org/10.3390/biom14101338 - 21 Oct 2024
Viewed by 1266
Abstract
Mutations in the parkin gene product Parkin give rise to autosomal recessive juvenile parkinsonism. Parkin is an E3 ubiquitin ligase that is a critical participant in the process of mitophagy. Parkin has a complex structure that integrates several allosteric signals to maintain precise [...] Read more.
Mutations in the parkin gene product Parkin give rise to autosomal recessive juvenile parkinsonism. Parkin is an E3 ubiquitin ligase that is a critical participant in the process of mitophagy. Parkin has a complex structure that integrates several allosteric signals to maintain precise control of its catalytic activity. Though its allosterically controlled structural reorganization has been extensively characterized by crystallography, the energetics and mechanisms of allosteric regulation of Parkin are much less well understood. Allostery is fundamentally linked to the energetics of the cooperative (sub)structure of the protein. Herein, we examine the mechanism of allosteric activation by phosphorylated ubiquitin binding to the enzymatic core of Parkin, which lacks the antagonistic Ubl domain. In this way, the allosteric effects of the agonist phosphorylated ubiquitin can be isolated. Using native-state hydrogen exchange monitored by mass spectrometry, we find that the five structural domains of the core of Parkin are energetically distinct. Nevertheless, association of phosphorylated ubiquitin destabilizes structural elements that bind the ubiquitin-like domain antagonist while promoting the dissociation of the catalytic domain and energetically poises the protein for transition to the fully activated structure. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Cartoon representation of the structure of the Parkin(∆Ubl)·pUb complex. RING0 (residues 142–225, red), RING1 (residues 226–329, yellow), IBR (residues 330–378, magenta), REP (residues 379–410, cyan), RING2 (residues 411–465, blue), zinc atoms (wheat) and pUb (orange). Drawn using PyMol from the structure determined by Wauer et al. (PDB code: 5caw) [<a href="#B10-biomolecules-14-01338" class="html-bibr">10</a>].</p>
Full article ">Figure 2
<p>Parkin(∆Ubl) denaturation by urea followed by circular dichroism at 222 nm. The solid line results from fitting the solid circles to Equation (1). For reasons outlined in the main text, the fitted ∆<span class="html-italic">G<sub>unf</sub></span> and m values indicate a relatively uncooperative multistate unfolding of the protein, which is consistent with the hydrogen exchange studies described below. The hollow circles correspond to a separate transition likely involving unfolding of structure stabilized by high-affinity zinc fingers. See main text.</p>
Full article ">Figure 3
<p>Structural fluctuations in Parkin(∆Ubl). Native-state hydrogen exchange time profiles representative of the various domains of Parkin(∆Ubl) measured by peptide fragmentation mass spectrometry (<b>A</b>–<b>I</b>). The domain and primary sequence residue numbers are shown in each panel. Fitting of these data (solid lines) gave average R<sup>2</sup> and Sy.x values of 0.99 ± 0.02 and 0.16 ± 0.09 amu, respectively.</p>
Full article ">Figure 4
<p>Structural fluctuations in the Parkin(∆Ubl)·pUb complex. Native-state hydrogen exchange time profiles representative of the various domains of Parkin(∆Ubl) in the Parkin(∆Ubl)·pUb complex measured by peptide fragmentation mass spectrometry (<b>A</b>–<b>I</b>). The domain and primary sequence residue numbers are shown in each panel. Fitting of these data (solid lines) gave average R<sup>2</sup> and Sy.x values of 0.98 ± 0.02 and 0.16 ± 0.10 amu, respectively.</p>
Full article ">Figure 5
<p>Comparison of the energetics of the protein ensemble revealed by native-state hydrogen exchange <math display="inline"><semantics> <mrow> <msubsup> <mrow> <mo>∆</mo> <mi>G</mi> </mrow> <mrow> <mi>H</mi> <mi>X</mi> </mrow> <mrow> <mi>d</mi> <mi>e</mi> <mi>p</mi> </mrow> </msubsup> </mrow> </semantics></math> in Parkin(∆Ubl) and the Parkin(∆Ubl)·pUb complex. Denaturant dependence of the apparent free energy of hydrogen exchange (∆G<sub>HX</sub>) of representative peptides derived from the domains of Parkin(∆Ubl) (<b>A</b>–<b>I</b>). The domain and primary sequence residue numbers are shown in each panel. fitted with one or two exponentials as statistically warranted (see <a href="#biomolecules-14-01338-t001" class="html-table">Table 1</a>). Fitting of these data (solid lines) gave average R<sup>2</sup> and Sy.x values of 0.96 ± 0.05 and 0.32 ± 0.17 kJ mol<sup>−1</sup>, respectively.</p>
Full article ">Figure 6
<p>Allosteric transition in Parkin(∆Ubl) upon binding of pUb. Cartoon representations of the structure of the RING1 domain (shown in yellow) of (<b>A</b>) <span class="html-italic">hs</span>Parkin(∆Ubl) (PDB code: 4bm9) and (<b>B</b>) the <span class="html-italic">Ph</span>Parkin(∆Ubl)·pUb complex (PDB code: 5caw). pUb is colored orange. Shown in green are glutamate and arginine residues participating in an ion pair in <span class="html-italic">hs</span>Parkin(∆Ubl), which is disrupted in the <span class="html-italic">Ph</span>Parkin(∆Ubl)·pUb complex. Note that the involved E and R side chains are ill defined (disordered) in the <span class="html-italic">Ph</span>Parkin(∆Ubl))·pUb complex. The urea dependence of the apparent free energy of the fluctuations leading to hydrogen exchange in the helix spanning residues 268–280 in <span class="html-italic">hs</span>Parkin(∆Ubl) (<b>C</b>) and in the homologous region in the <span class="html-italic">Ph</span>Parkin(∆Ubl)·pUb complex (<b>D</b>). The HX time courses of this region in both the free and complexed Parkin(∆Ubl) are best fitted with two exponentials. Fitting of these data gave average R<sup>2</sup> and Sy.x values of 0.94 ± 0.06 and 0.36 ± 0.11 kJ mol<sup>−1</sup>, respectively. Deconvolution using overlapping peptides indicates that the fast phase is associated with the C-terminus of this helix while the slower phase is associated with the N-terminal region. The C-terminus is destabilized by the binding of pUb. In contrast, the N-terminal region is stabilized by the binding of pUb. Cartoons drawn with PyMol.</p>
Full article ">Figure 7
<p>Urea dependence of the activation free energy of dissociation of pUb from Parkin(∆Ubl). Rates of dissociation of pUb from Parkin(∆Ubl) were extracted from two population binomial fits of the <span class="html-italic">m</span>/<span class="html-italic">z</span> distribution HX time courses of the Parkin(∆Ubl)·pUb complex as a function of urea (see <a href="#app1-biomolecules-14-01338" class="html-app">Figure S1</a>). The fitted m-value was 0.83 ± 0.11 kJ mol<sup>−1</sup> M<sup>−1</sup> and R<sup>2</sup> 0.89.</p>
Full article ">
1 pages, 143 KiB  
Correction
Correction: Choi et al. β-Ionone Attenuates Dexamethasone-Induced Suppression of Collagen and Hyaluronic Acid Synthesis in Human Dermal Fibroblasts. Biomolecules 2021, 11, 619
by Dabin Choi, Wesuk Kang, Soyoon Park, Bomin Son and Taesun Park
Biomolecules 2024, 14(10), 1337; https://doi.org/10.3390/biom14101337 - 21 Oct 2024
Viewed by 479
Abstract
The authors would like to modify the Conflicts of Interest section of the published paper [...] Full article
20 pages, 1322 KiB  
Article
Chemical Profiling of Polar Lipids and the Polyphenolic Fraction of Commercial Italian Phaseolus Seeds by UHPLC-HRMS and Biological Evaluation
by Vadym Samukha, Francesca Fantasma, Gilda D’Urso, Ester Colarusso, Anna Schettino, Noemi Marigliano, Maria Giovanna Chini, Gabriella Saviano, Vincenzo De Felice, Gianluigi Lauro, Francesco Maione, Giuseppe Bifulco, Agostino Casapullo and Maria Iorizzi
Biomolecules 2024, 14(10), 1336; https://doi.org/10.3390/biom14101336 - 20 Oct 2024
Viewed by 1218
Abstract
The common bean (Phaseolus vulgaris L.) is one of the oldest food crops in the world. In this study, the ultra-high-performance liquid chromatography high-resolution mass spectrometry (UHPLC-MS/MS) technique was used to characterize the polar lipid composition and polyphenolic fraction of five bean [...] Read more.
The common bean (Phaseolus vulgaris L.) is one of the oldest food crops in the world. In this study, the ultra-high-performance liquid chromatography high-resolution mass spectrometry (UHPLC-MS/MS) technique was used to characterize the polar lipid composition and polyphenolic fraction of five bean varieties commonly consumed in Italy: Cannellino (PVCA), Controne (PVCO), Borlotti (PVBO), Stregoni (PVST), and Vellutina (PVVE). Lipid content represents a minor fraction of the whole metabolome in dry beans, and little is known about their polar lipids, which could be potentially bioactive components. Thirty-three compounds were detected through UHPLC-MS/MS, including oxylipins, phospholipids, N-acyl glycerolipids, and several fatty acids. The dichloromethane extracts were subjected to principal component analysis (PCA), with the results showing greater differentiation for the Borlotti variety. Moreover, 27 components belonging to different polyphenol classes, such as phenolic acids, flavonoids, catechins, anthocyanins and their glycosides, and some saponins, were identified in the hydroalcoholic seed extracts. In addition, the mineral content of the beans was determined. Considering the high number of compounds in the five apolar seed extracts, all samples were examined to determine their in vitro inhibitory activity against the enzyme cyclooxygenase-2 (COX-2), which is inducible in inflammatory cells and mediates inflammatory responses. Only PVCO showed the best inhibition of the COX-2 enzyme with an IC50 = 31.15 ± 2.16 µg/mL. In light of these results, the potential anti-inflammatory properties of PVCO were evaluated in the LPS-stimulated murine macrophage cell line J774A.1. Herein, we demonstrate, for the first time, that PVCO at 30 µg/mL can significantly reduce the release of TNF-α, with a less significant anti-inflammatory effect being observed in terms of IL-6 release. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Selected Italian commercial bean varieties: Vellutina (PVVE) (<b>a</b>); Borlotti (PVBO) (<b>b</b>); Stregoni (PVST) (<b>c</b>); Controne (PVCO) (<b>d</b>); Cannellino (PVCA) (<b>e</b>).</p>
Full article ">Figure 2
<p>Score scatter plot of the partial least squares–discriminant analysis (PLS-DA) performed as a targeted approach on the detected compounds of the lipophilic extracts of five Italian commercial <span class="html-italic">P. vulgaris</span> varieties: Borlotti (PVBO) (green), Cannellino (PVCA) (blue), Controne (PVCO) (red), Stregoni (PVST) (yellow), and Vellutina (PVVE) (light blue).</p>
Full article ">Figure 3
<p>Loading scatter plot of the targeted partial least squares–discriminant analysis (PLS-DA) performed on the lipophilic extracts of five Italian commercial <span class="html-italic">P. vulgaris</span> varieties: Borlotti (PVBO (B)), Cannellino (PVCA (C)), Controne (PVCO (O)), Stregoni (PVST (S)), and Vellutina (PVVE (V)).</p>
Full article ">Figure 4
<p>(<b>A</b>,<b>B</b>) An in vitro cytotoxic examination, evaluated using an MTT assay, was performed on the murine macrophage J774A.1 cell line, following 4 (<b>A</b>) and 24 h (<b>B</b>) of treatment with selected concentrations of PVCO (3, 10, 30, and 100 μg/mL). The dotted lines indicate the 75% cell viability limit. Data are expressed as cell viability (% of control) and presented as the means ± S.D. of three independent experiments. Our statistical analysis was conducted using one-way ANOVA followed by Bonferroni correction for multiple comparisons. (<b>C</b>,<b>F</b>) Murine macrophages J774A.1 were stimulated with LPS (10 µg/mL) and treated with PVCO at the concentration of 30 µg/mL for 4 and 24 h (<b>C</b>). Thereafter, cell supernatants were assayed using ELISA to determine TNF-α and IL-6 levels (expressed as pg/mL) at both 4 (<b>D</b>,<b>F</b>, respectively) and 24 h (<b>E</b>,<b>G</b>, respectively). Data are presented as the means ± S.D. of three independent experiments. <sup>####</sup> <span class="html-italic">p</span> ≤ 0.0001 vs. the Ctrl group; * <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> ≤ 0.01 vs. the LPS group.</p>
Full article ">
17 pages, 764 KiB  
Article
Increased Cardiometabolic Risk in Men with Hypoprolactinemia: A Pilot Study
by Robert Krysiak, Karolina Kowalcze, Witold Szkróbka and Bogusław Okopień
Biomolecules 2024, 14(10), 1335; https://doi.org/10.3390/biom14101335 - 20 Oct 2024
Viewed by 1213
Abstract
Low prolactin levels in men predispose them to mood disturbances, sexual dysfunction, and diabetes. The purpose of the current study was to assess cardiometabolic risk in males with hypoprolactinemia. This prospective study included three age-matched groups of young and middle-aged men: individuals with [...] Read more.
Low prolactin levels in men predispose them to mood disturbances, sexual dysfunction, and diabetes. The purpose of the current study was to assess cardiometabolic risk in males with hypoprolactinemia. This prospective study included three age-matched groups of young and middle-aged men: individuals with cabergoline-induced hypoprolactinemia (n = 15), cabergoline-treated subjects with prolactin levels within the reference range (n = 20), and untreated men with normal prolactin levels (n = 31). In men with hypoprolactinemia, the cabergoline dose was reduced in order to normalize prolactin concentration. Anthropometric parameters, blood pressure, QRISK3 score; plasma concentrations of prolactin, glucose, insulin, lipids, uric acid, high-sensitivity C-reactive protein (hsCRP), fibrinogen, homocysteine, and testosterone; whole-blood levels of glycated hemoglobin (HbA1C); urinary albumin-to-creatinine ratio (UACR); and carotid intima–media thickness were assessed at baseline and six months later. Men with hypoprolactinemia were characterized by higher body mass index, fat content, waist circumference, systolic blood pressure, fasting and 2 h post-load glucose, HbA1C, HOMA1-IR, uric acid, hsCRP, fibrinogen, homocysteine, and UACR; by lower HDL cholesterol and testosterone; by greater intima–media thickness; and by a higher QRISK3 score than their peers with normal prolactin levels. There were no statistically significant differences in the measured parameters between both groups of men with normal prolactin levels. Normalization of prolactin concentration was accompanied by normalization of biochemical variables, systolic blood pressure, and QRISK3 score. Although cabergoline dose reduction did not cause statistically significant changes in the remaining anthropometric parameters and intima–media thickness, six months later, they did not differ from those observed in the remaining study groups. Our findings suggest that iatrogenic hypoprolactinemia is associated with increased cardiometabolic risk, which is reversible and resolves after the normalization of prolactin levels. Full article
(This article belongs to the Special Issue Advances in Cardiometabolic Health)
Show Figures

Figure 1

Figure 1
<p>A diagram presenting the flow of patients in this study.</p>
Full article ">Figure 2
<p>Correlations between cabergoline-dose-reduction-induced changes in prolactin levels and the changes in levels of cardiometabolic risk factors and QRISK3 score in in patients with iatrogenic hypoprolactinemia. Abbreviations: hsCRP—high-sensitivity C-reactive protein; UACR—urinary albumin-to-creatinine ratio.</p>
Full article ">
19 pages, 1379 KiB  
Review
Potential Roles of IP3 Receptors and Calcium in Programmed Cell Death and Implications in Cardiovascular Diseases
by Chanon Piamsiri, Nadezhda Fefelova, Sri Harika Pamarthi, Judith K. Gwathmey, Siriporn C. Chattipakorn, Nipon Chattipakorn and Lai-Hua Xie
Biomolecules 2024, 14(10), 1334; https://doi.org/10.3390/biom14101334 - 20 Oct 2024
Cited by 1 | Viewed by 2048
Abstract
Inositol 1,4,5-trisphosphate receptors (IP3Rs) play a crucial role in maintaining intracellular/cytosolic calcium ion (Ca2+i) homeostasis. The release of Ca2+ from IP3Rs serves as a second messenger and a modulatory factor influencing various intracellular and interorganelle [...] Read more.
Inositol 1,4,5-trisphosphate receptors (IP3Rs) play a crucial role in maintaining intracellular/cytosolic calcium ion (Ca2+i) homeostasis. The release of Ca2+ from IP3Rs serves as a second messenger and a modulatory factor influencing various intracellular and interorganelle communications during both physiological and pathological processes. Accumulating evidence from in vitro, in vivo, and clinical studies supports the notion that the overactivation of IP3Rs is linked to the pathogenesis of various cardiac conditions. The overactivation of IP3Rs results in the dysregulation of Ca2+ concentration ([Ca2+]) within cytosolic, mitochondrial, and nucleoplasmic cellular compartments. In cardiovascular pathologies, two isoforms of IP3Rs, i.e., IP3R1 and IP3R2, have been identified. Notably, IP3R1 plays a pivotal role in cardiac ischemia and diabetes-induced arrhythmias, while IP3R2 is implicated in sepsis-induced cardiomyopathy and cardiac hypertrophy. Furthermore, IP3Rs have been reported to be involved in various programmed cell death (PCD) pathways, such as apoptosis, pyroptosis, and ferroptosis underscoring their multifaceted roles in cardiac pathophysiology. Based on these findings, it is evident that exploring potential therapeutic avenues becomes crucial. Both genetic ablation and pharmacological intervention using IP3R antagonists have emerged as promising strategies against IP3R-related pathologies suggesting their potential therapeutic potency. This review summarizes the roles of IP3Rs in cardiac physiology and pathology and establishes a foundational understanding with a particular focus on their involvement in the various PCD pathways within the context of cardiovascular diseases. Full article
(This article belongs to the Section Cellular Biochemistry)
Show Figures

Figure 1

Figure 1
<p>A schematic diagram illustrating the regulatory mechanisms of IP<sub>3</sub>Rs in cardiomyocytes under both physiological and pathological conditions. (<b>A</b>) During physiological conditions, external stimuli such as adenosine, angiotensin II, bradykinin, endothelin, vasopressin, dopamine, epinephrine, norepinephrine, etc., bind and activate GPCRs on the plasma membrane. Subsequently, the Gqα-subunit dissociates from the trimeric GPCR structure, activating PLC to produce IP<sub>3</sub> from PIP2. IP<sub>3</sub> then binds to the IP<sub>3</sub> binding core domain of IP<sub>3</sub>Rs leading to the release of Ca<sup>2+</sup> from the SR. However, the amount of Ca<sup>2+</sup> flux via IP<sub>3</sub>Rs is relatively lower compared to the Ca<sup>2+</sup> transients generated by CICR and other major voltage-operated channels and ligand-gated channels. IP<sub>3</sub>R-mediated Ca<sup>2+</sup> release causes Ca<sup>2+</sup> translocation from the SR to the cytosol, mitochondria, and nucleus. In the cytosol, Ca<sup>2+</sup> serves as a secondary messenger playing modulatory roles. Ca<sup>2+</sup><sub>i</sub> binds to cTnC and initiates cardiac contraction. In mitochondria, Ca<sup>2+</sup><sub>Mito</sub> influences mitochondrial energy production and metabolic activity. Mitochondria also act as secondary storage sites and modulate [Ca<sup>2+</sup>]<sub>i</sub> levels. In the nucleus, Ca<sup>2+</sup><sub>Nuc</sub> regulates gene expression and transcription. (<b>B</b>) During cardiac pathologies (e.g., ischemia, I/R injuries, arrhythmias, sepsis-induced cardiomyopathy, cardiac hypertrophy, and heart failure), neurohormonal hyperactivation leads to exacerbated IP<sub>3</sub> production from GPCR. Additionally, these pathological conditions cause IP<sub>3</sub>Rs overexpression leading to Ca<sup>2+</sup><sub>i</sub>, Ca<sup>2+</sup><sub>Mito</sub>, and Ca<sup>2+</sup><sub>Nuc</sub> overload. Increased Ca<sup>2+</sup><sub>i</sub> binds with calmodulin thus leading to CaMKII overactivation. Ca<sup>2+</sup><sub>i</sub> overload causes cardiac hypercontracture, contractile dysfunction, cardiac cell injuries, cardiac cell death, and arrhythmias. Ca<sup>2+</sup><sub>Mito</sub> overload causes mitochondrial dysfunction and impaired mitochondrial ATP production. Ca<sup>2+</sup><sub>Nuc</sub> overload enhances the function of transcription factors that control the expression of pro-hypertrophic and pro-arrhythmic genes. It should be noted that the remodeling of IP<sub>3</sub>Rs and Ca<sup>2+</sup> signaling contribute to different cardiovascular diseases and pathophysiological conditions. Endothelin-induced excessive IP<sub>3</sub>R1 expression and overactivation lead to Ca<sup>2+</sup><sub>i</sub> and Ca<sup>2+</sup><sub>Nuc</sub> overload, subsequently contributing to the development of atrial arrhythmias [<a href="#B6-biomolecules-14-01334" class="html-bibr">6</a>,<a href="#B10-biomolecules-14-01334" class="html-bibr">10</a>,<a href="#B21-biomolecules-14-01334" class="html-bibr">21</a>]. In sepsis-induced cardiomyopathy, the LPS-induced overexpression of IP<sub>3</sub>R2 leads to Ca<sup>2+</sup><sub>i</sub> overload triggering myocardial cell death through pyroptosis and apoptosis [<a href="#B14-biomolecules-14-01334" class="html-bibr">14</a>]. In ischemic heart disease, the upregulation of IP<sub>3</sub>R1 in ventricular cardiomyocytes results in Ca<sup>2+</sup><sub>i</sub> and Ca<sup>2+</sup><sub>Mito</sub> overload leading to myocardial cell death through pyroptosis [<a href="#B6-biomolecules-14-01334" class="html-bibr">6</a>,<a href="#B13-biomolecules-14-01334" class="html-bibr">13</a>]. In the failing heart, persistent increased cardiac workload leads to chronic neurohormonal hyperactivation thereby triggering IP<sub>3</sub>R2/calcineurin/CaMKII-mediated NFAT nuclear translocation, which contributes to the transcription of hypertrophic genes [<a href="#B6-biomolecules-14-01334" class="html-bibr">6</a>,<a href="#B22-biomolecules-14-01334" class="html-bibr">22</a>,<a href="#B23-biomolecules-14-01334" class="html-bibr">23</a>]. This figure was created using BioRender. Abbreviations: ADP, adenosine diphosphate; ATP, adenosine triphosphate; CaMKII, Ca<sup>2+</sup>/calmodulin-dependent protein kinase II; CICR, calcium-induced calcium release; cTnC, cardiac troponin C; GPCR, G protein-coupled receptors; IP<sub>3</sub>, inositol 1,4,5-trisphosphate; IP<sub>3</sub>Rs, inositol 1,4,5-trisphosphate receptors; LTCC, L-type calcium channel; PIP2, phosphatidylinositol 4,5-bisphosphate (PIP2); PLB, phospholamban; PLC, phospholipase C; MCU, mitochondrial calcium uniporter; mPTP, mitochondrial permeability transition pore; mtROS, mitochondrial ROS production; NCX, sodium–calcium exchanger; RyR, ryanodine receptors; SERCA, sarcoplasmic/endoplasmic reticulum Ca<sup>2+</sup>-ATPase; SR, sarcoplasmic reticulum; [Ca<sup>2+</sup>], calcium ion concentration; [Ca<sup>2+</sup>]<sub>i</sub>, calcium ion concentration in the cytosol; [Ca<sup>2+</sup>]<sub>Mito</sub>, calcium ion concentration in mitochondria; [Ca<sup>2+</sup>]<sub>Nuc</sub> calcium ion concentration in the nucleus; Δ<span class="html-italic">Ψ<sub>m</sub></span> mitochondrial membrane potential.</p>
Full article ">Figure 2
<p>A schematic diagram illustrating the roles of IP<sub>3</sub>Rs in apoptosis, pyroptosis, and ferroptosis. (<b>A</b>) Apoptosis: Several stress conditions (e.g., high glucose condition) mediate ER stress resulting in the upregulation of IP<sub>3</sub>Rs and GRP75 expression. Formation of the IP<sub>3</sub>Rs-GRP75-VDAC complex facilitates ER-mediated Ca<sup>2+</sup><sub>Mito</sub> overload. Excessive [Ca<sup>2+</sup>]<sub>Mito</sub> causes mitochondrial dysfunction and facilitates mPTP opening, leading to Cytochrome C being released. The access of Cytochrome C to the cytosol initiates mitochondrial-mediated apoptosis by activating caspase cascades, including the cleavage of caspase 9 and caspase 3. (<b>B</b>) Pyroptosis: Stressors such as I/R or H/R induce the overexpression of IP<sub>3</sub>Rs and the downregulation of ERP44, thereby mediating Ca<sup>2+</sup><sub>Mito</sub> and Ca<sup>2+</sup><sub>i</sub> overload. Ca<sup>2+</sup><sub>Mito</sub> overload impairs mitochondrial functions, disrupts oxidative phosphorylation, and increases mtROS production. The resultant overproduction of mtROS and Ca<sup>2+</sup><sub>i</sub> overload lead to NLRP3 activation and inflammasome assembly. Once activated, the NLRP3 inflammasome cleaves pro-caspase 1 to its activated form. Then, cleaved caspase 1 further cleaves and activates GSDMD into its <span class="html-italic">N</span>-terminal fragment (GSDMD-NT). As a result, GSDMD-NT translocates and oligomerizes at the inner leaflet of the plasma membrane resulting in the formation of transmembrane pores. (<b>C</b>) Ferroptosis: It is proposed that ER stress could mediate ferroptosis through the overactivation of IP<sub>3</sub>Rs, thereby, leading to Ca<sup>2+</sup><sub>Mito</sub> overload and mtROS overproduction. The administration of ferroptosis activator RSL3 inhibited the antioxidant property of GPX4 exacerbating mitochondrial and intracellular ROS levels. The subsequent augmentation of ROS leads to membrane lipid peroxidation and ultimately causes membrane rupture. In addition, Ca<sup>2+</sup><sub>i</sub> overload caused by IP<sub>3</sub>Rs also enhances the catalytic activity of LOX, further promoting lipid membrane peroxidation. However, the underlying mechanism of whether and how Ca<sup>2+</sup><sub>i</sub> impacts LOX remains unclear. This figure was created using BioRender. Abbreviations: BAX, Bcl-2-associated protein X; Bcl-2, B-cell lymphoma 2; GSDMD, gasdermin D; GSDMD-NT, gasdermin D <span class="html-italic">N</span>-terminus; H/R, hypoxia-reoxygenation; IP<sub>3</sub>Rs, inositol 1,4,5-trisphosphate receptors; I/R, ischemia–reperfusion; LOX, lipoxygenases; MCU, mitochondrial calcium uniporter; MFRN, mitoferrin 2; mPTP, mitochondrial permeability transition pore; mtROS, mitochondrial ROS; NLRP3, nod-like receptor protein-3; RSL3, RAS-selective lethal 3; [Ca<sup>2+</sup>], calcium ion concentration; VDAC, voltage-dependent anion channel; [Ca<sup>2+</sup>]<sub>i</sub>, calcium ion concentration in the cytosol; [Ca<sup>2+</sup>]<sub>Mito</sub>, calcium ion concentration in mitochondria; [Ca<sup>2+</sup>]<sub>Nuc</sub> calcium ion concentration in the nucleus; Δ<span class="html-italic">Ψ<sub>m</sub></span>, mitochondrial membrane potential.</p>
Full article ">
15 pages, 1695 KiB  
Review
Stress-Induced Evolution of the Nucleolus: The Role of Ribosomal Intergenic Spacer (rIGS) Transcripts
by Anastasia A. Gavrilova, Margarita V. Neklesova, Yuliya A. Zagryadskaya, Irina M. Kuznetsova, Konstantin K. Turoverov and Alexander V. Fonin
Biomolecules 2024, 14(10), 1333; https://doi.org/10.3390/biom14101333 - 20 Oct 2024
Viewed by 1681
Abstract
It became clear more than 20 years ago that the nucleolus not only performs the most important biological function of assembling ribonucleic particles but is also a key controller of many cellular processes, participating in cellular adaptation to stress. The nucleolus’s multifunctionality is [...] Read more.
It became clear more than 20 years ago that the nucleolus not only performs the most important biological function of assembling ribonucleic particles but is also a key controller of many cellular processes, participating in cellular adaptation to stress. The nucleolus’s multifunctionality is due to the peculiarities of its biogenesis. The nucleolus is a multilayered biomolecular condensate formed by liquid–liquid phase separation (LLPS). In this review, we focus on changes occurring in the nucleolus during cellular stress, molecular features of the nucleolar response to abnormal and stressful conditions, and the role of long non-coding RNAs transcribed from the intergenic spacer region of ribosomal DNA (IGS rDNA). Full article
(This article belongs to the Section Biological Factors)
Show Figures

Figure 1

Figure 1
<p>The ribosomal DNA cassette (rDNA cassette). The rDNA cassette contains sequences encoding the pre-RNA and the ribosomal intergenic spacer (rIGS). In humans, the cassette size is approximately 43 kb; they are located on the p-arms of five chromosomes. Distal Junction is a sequence approximately 400 kb long that flanks the ribosomal gene repeat. Polymerase I transcribes several functional noncoding RNAs from rIGS. rIGS 16 RNA and rIGS 22 RNA are synthesized during heat shock, and rIGS 28 RNA is synthesized under acidosis conditions. pRNA is transcribed from spacer promoters upstream of the pre-rRNA transcription start site. PAPAS are a set of antisense RNAs that are synthesized by Pol II. PAPAS transcripts do not have a common promoter; their transcription begins at random sites and can span both pre-rRNA coding and IGS regions longer than 10 kb.</p>
Full article ">Figure 2
<p>Nucleolar transformation in response to stress. The interphase nucleolus with characteristic tripartite structure is shown in the center. Panels (<b>A</b>–<b>D</b>) show structures that arise under different stressful influences. (<b>A</b>) Nucleolar segregation or nucleolar caps are formed when RNA pol I transcription is inactivated. Actinomycin D or ionizing radiation induces rDNA double-strand breaks (DSBs), resulting in the formation of nucleolar capsules adjacent to their DJ anchor. (<b>B</b>) Upon DRB treatment, RNA pol I transcription is active, but rRNA processing is converted to form a nucleolar necklace. (<b>C</b>) When exposed to heat shock and acidosis, the nucleolus transforms into an electron-dense fibrillar organization, the A-body. The fibers contain immobilized proteins in an amyloid-like state. (<b>D</b>) Nucleolar aggresomes are formed upon proteotoxic insults such as proteasome inhibition and heat shock. This may or may not involve inhibited RNA pol I activity.</p>
Full article ">Figure 3
<p>Stress-induced transcription of non-coding RNAs from the IGS region. (<b>A</b>) pRNA recruits the Nucleolar Remodeling Complex (NoRC) to the promoter and, due to its hairpin structure, binds to one of the NoRC proteins, TIP5, thereby mediating the nucleolar localization of the entire complex. NoRC moves the nucleosome to a repressive position, preventing transcription initiation. (<b>B</b>) During cellular quiescence and starvation, the amount of PAPAS increases. The transcripts recruit the histone methyltransferase Suv4-20h2 to the rDNA locus, which installs the repressive H4K20me3 mark on the rDNA, resulting in immediate suppression of rDNA expression. (<b>C</b>) During heat shock and acidosis, rIGS 16 RNA, rIGS 22 RNA, and rIGS 28 RNA are transcribed, respectively. These transcripts likely mediate the nucleolar sequestration of VHL, CDC73, MDM2, POLD1, and many other proteins possessing amyloid-converting motifs. The local concentration of proteins with an amyloidogenic propensity in the foci triggers physiological amyloidogenesis and generates nascent amyloid bodies (A-bodies).</p>
Full article ">
22 pages, 7650 KiB  
Article
Identification of Multifunctional Putative Bioactive Peptides in the Insect Model Red Palm Weevil (Rhynchophorus ferrugineus)
by Carmen Scieuzo, Roberta Rinaldi, Fabiana Giglio, Rosanna Salvia, Mohammed Ali AlSaleh, Jernej Jakše, Arnab Pain, Binu Antony and Patrizia Falabella
Biomolecules 2024, 14(10), 1332; https://doi.org/10.3390/biom14101332 - 19 Oct 2024
Cited by 1 | Viewed by 1631
Abstract
Innate immunity, the body’s initial defense against bacteria, fungi, and viruses, heavily depends on antimicrobial peptides (AMPs), which are small molecules produced by all living organisms. Insects, with their vast biodiversity, are one of the most abundant and innovative sources of AMPs. In [...] Read more.
Innate immunity, the body’s initial defense against bacteria, fungi, and viruses, heavily depends on antimicrobial peptides (AMPs), which are small molecules produced by all living organisms. Insects, with their vast biodiversity, are one of the most abundant and innovative sources of AMPs. In this study, AMPs from the red palm weevil (RPW) Rhynchophorus ferrugineus (Coleoptera: Curculionidae), a known invasive pest of palm species, were examined. The AMPs were identified in the transcriptomes from different body parts of male and female adults, under different experimental conditions, including specimens collected from the field and those reared in the laboratory. The RPW transcriptomes were examined to predict antimicrobial activity, and all sequences putatively encoding AMPs were analyzed using several machine learning algorithms available in the CAMPR3 database. Additionally, anticancer, antiviral, and antifungal activity of the peptides were predicted using iACP, AVPpred, and Antifp server tools, respectively. Physicochemical parameters were assessed using the Antimicrobial Peptide Database Calculator and Predictor. From these analyses, 198 putatively active peptides were identified, which can be tested in future studies to validate the in silico predictions. Genome-wide analysis revealed that several AMPs have predominantly emerged through gene duplication. Noticeably, we detect a newly originated defensin allele from an ancestral defensin via the deletion of two amino acids following gene duplication in RPW, which may confer an enhanced resilience to microbial infection. Our study shed light on AMP gene families and shows that high duplication and deletion rates are essential to achieve a diversity of antimicrobial mechanisms; hence, we propose the RPW AMPs as a model for exploring gene duplication and functional variations against microbial infection. Full article
(This article belongs to the Special Issue State of the Art and Perspectives in Antimicrobial Peptides)
Show Figures

Figure 1

Figure 1
<p>Heatmap showing the expression levels of duplicated genes in the different body parts of <span class="html-italic">Rhynchophorus ferrugineus</span> lab-reared and field-collected male and female adults. The heatmap colors represent transcript abundance in transcripts per million (TPM) from highest (red) to lowest (blue) expression levels. The data represented as log-transformed TPM values were tabulated and converted into heatmaps using R and R Studio software (version 2023.06.2+561). (<b>A</b>) Wings; (<b>B</b>) Legs; (<b>C</b>) Abdomen; (<b>D</b>) Thorax; (<b>E</b>) Head; (<b>F</b>) Antennae; (<b>G</b>) Snout; (<b>H</b>) Gut; (<b>I</b>) Fat Body.</p>
Full article ">Figure 2
<p>Classification of the 198 total AMPs detected in adult transcriptomes related to their family classification.</p>
Full article ">Figure 3
<p>Graphical display for the cecropin genes mapped in scaffold_405 (NCBI acc no. JAACXV010000404.1) showing functional cecropins gene length, CDS (coding region) length, and protein length, which was generated using the NCBI graphical sequence viewer available in the Genome workbench. The visual code shows green, red, and purple, indicating gene, coding region, and mRNA, respectively. The line shows the introns and boxes for the exons.</p>
Full article ">Figure 4
<p>Graphical display for the defensin genes mapped in the six scaffolds (NCBI acc no. JAACXV010014362.1, JAACXV010000413.1, JAACXV010014484.1, JAACXV010014200.1, JAACXV010014575.1, and JAACXV010014362.1) showing functional gene length, CDS length, and protein length generated using the NCBI graphical sequence viewer available in the Genome workbench. The visual code shows green, red, and purple, indicating gene, coding region, and mRNA.</p>
Full article ">Figure 5
<p>Two allelic variants in the locus tag “GWI33_019784” and deduced amino acids predict 82aa and 84-aa proteins (NCBI acc nos. KAF7266949 and KAF7266950), with a conserved DEFL_defensin-like domain at the C-terminal region. Dots denote identical amino acid residues, and conserved DEFL_defensin-like domain at the C-terminal region are underlined.</p>
Full article ">Figure 6
<p>Graphical display for the hypothetical AMP genes mapped in the two scaffolds (66363 and 66088) (NCBI acc nos. JAACXV010014549.1 and JAACXV010014301.1) showing the functional AMP gene length, CDS length, and protein length generated using the NCBI graphical sequence viewer available in the Genome workbench. The visual code shows green, red, and purple, indicating gene, coding region, and mRNA, respectively. The line shows the introns and boxes for the exons.</p>
Full article ">Figure 7
<p>Graphical display for the lysozyme genes mapped in the two scaffolds (66335 and 66281) (NCBI acc nos. JAACXV010014523.1 and JAACXV010014472.1) showing functional lysozyme gene length, CDS length, and protein length generated using the NCBI graphical sequence viewer available in the Genome workbench. The visual code shows green, red, and purple, indicating gene, coding region, and mRNA, respectively. The line shows the introns and boxes for the exons.</p>
Full article ">
17 pages, 4282 KiB  
Article
A Novel Peptide from VP1 of EV-D68 Exhibits Broad-Spectrum Antiviral Activity Against Human Enteroviruses
by Xiaojing Lin, Qiang Sun, Yang Cao, Zi Li, Cuiling Xu, Jun Liu, Jingdong Song, Kun Qin, Yong Zhang and Jianfang Zhou
Biomolecules 2024, 14(10), 1331; https://doi.org/10.3390/biom14101331 - 19 Oct 2024
Viewed by 1219
Abstract
Enteroviruses have been a historical concern since the identification of polioviruses in humans. Wild polioviruses have almost been eliminated, while multiple species of non-polio enteroviruses and their variants co-circulate annually. To date, at least 116 types have been found in humans and are [...] Read more.
Enteroviruses have been a historical concern since the identification of polioviruses in humans. Wild polioviruses have almost been eliminated, while multiple species of non-polio enteroviruses and their variants co-circulate annually. To date, at least 116 types have been found in humans and are grouped into the species Enterovirus A–D and Rhinovirus A–C. However, there are few available antiviral drugs, especially with a universal pharmaceutical effect. Here, we demonstrate that peptide P25 from EV-D68 has broad antiviral activity against EV A–D enteroviruses in vitro. P25, derived from the HI loop and β-I sheet of VP1, operates through a conserved hydrophilic motif -R---K-K--K- and the hydrophobic F near the N-terminus. It could prevent viral infection of EV-A71 by competing for the heparan sulfate (HS) receptor, binding and stabilizing virions by suppressing the release of the viral genome. P25 also inhibited the generation of infectious viral particles by reducing viral protein synthesis. The molecular docking revealed that P25 might bind to the pocket opening area, a potential target for broad-spectrum antivirals. Our findings implicate the multiple antiviral effects of peptide P25, including blocking viral binding to the HS receptor, impeding viral genome release, and reducing progeny particles, which could be a novel universal anti-enterovirus drug candidate. Full article
(This article belongs to the Section Biomacromolecules: Proteins)
Show Figures

Figure 1

Figure 1
<p>Peptide P11 and P25 exhibit antiviral potency to human enteroviruses. (<b>a</b>) Antiviral effects of peptide P1-P30 on EV-A71/FY0805, Echo 30/WZ16, and EV-D68/BCH895A. RD cells were infected with 100 TCID<sub>50</sub>/50 µL human enteroviruses co-incubated with 50 µL peptide P1-P30 at a concentration of 125 µg/mL (about 56 µM). 24 h post-infection, RD cells were stained using crystal violet and measured at 550 nm. The CPE was normalized to the only virus control (0%) and 0.5% DMSO mock (100%), and then converted to a white and blue heatmap. (<b>b</b>) The antiviral effects of the P25 homologous segments of VP1 from EV-A71 (P25.A71), Echo 30 (P25.E30), Poliovirus 3 (P25.PV3), Rhinovirus A81 (P25.A81), and Rhinovirus B70 (P25.B70). (<b>c</b>) Location of P11 and P25 at VP1 of EV-D68 (PDB: 6CRR). P11 and P25 are shown in yellow and magenta, respectively. (<b>d</b>) Peptide parameters of P11 and P25 calculated using the ProtParam tool. The positively charged amino acids were marked in red. P25 has a longer estimated half-life than P11. Sequences of P1-P30 are provided in <a href="#app1-biomolecules-14-01331" class="html-app">Supplementary Table S1</a>.</p>
Full article ">Figure 2
<p>Antiviral activities of P25 mutants and its truncated peptides. (<b>a</b>) Sequences and parameters of the mutant P25s, parameters were calculated using the ProtParam tool. The positively charged amino acids were marked in red; “ + ” and “-” indicated the antiviral activity positive and negative, respectively; “ ++ ” indicated a broad spectrum of antiviral activity. The peptide P25.A81, was derived from Rhinovirus A81 and only inhibited EV-A71/FY0805 infection. The replacement of the N-terminal with G and F of P25.A81GF extended its antiviral profile. (<b>b</b>) RD cells were treated with 1 mg/mL of P25, P25.8, P25.9, P25.M, and P25.R, with serial 2-fold dilution for 24 h, and CC<sub>50</sub> values were assayed using CCK8 reagents. P25.R was the peptide with a reverse sequence of P25 and had a stronger cytotoxic effect. So, the IC<sub>50</sub> was not further tested as listed in <a href="#biomolecules-14-01331-t001" class="html-table">Table 1</a>. (<b>c</b>–<b>f</b>) RD cells were infected with enteroviruses co-incubated with serial 2-fold diluted P25 mutants and its truncated peptides, stained using crystal violet and measured at 550 nm at 24 h post-infection. The CPE was normalized to the only virus control (0%) and a 0.5% DMSO mock (100%), and then converted to a white and blue heatmap. (<b>c</b>) Anti-EV-A71/FY0805 infection. (<b>d</b>) Anti-Echo 30/WZ16 infection. (<b>e</b>) Anti-Poliovirus 3/nOPV3 infection. (<b>f</b>) Anti-EV-D68/BCH895A infection.</p>
Full article ">Figure 3
<p>P25 reduced the CPE of EV-A71/FY0805 via blocking viral binding to HS. (<b>a</b>) P25 at 62.5 µg/mL was used at the time points of pre-treatment (−6 h, −4 h, −2 h), co-treatment (0 h), and post-treatment (2 h, 4 h) with RD cells, and then the cells were infected with 100 TCID<sub>50</sub>/50 µL of EV-A71/FY0805. The cells were stained using crystal violet at 24 h post-infection. (<b>b</b>) The nonlinear curve fit of P25 pre-treatment with RD cells at −6 h, −4 h, −2 h compared with the co-treatment at 0 h. Pre-treatment enhanced the antiviral activity along with increased incubation time. The IC<sub>50</sub> of P25 at 6 h of pre-treatment is 3.9 µg/mL. (<b>c</b>) Co-treatment of P25, P25.8, P25.9, and P25.M provides complete protection against 100 TCID<sub>50</sub>/50 µL of EV-A71/FY0805 with an IC<sub>50</sub> of 32.35 ± 7.23 µg/mL, 24.49 ± 2.96 µg/mL, 13.02 ± 0.81 µg/mL, and 9.11 ± 2.52 µg/mL, respectively. P25.5 served as the negative control. (<b>d</b>) The antiviral activities of P25, P25.8, P25.9, and P25.M were interfered with by HS, which was a binding receptor for EV-A71/FY0805. Peptide P25s at a concentration of 62.5 µg/mL was pre-incubated with 500 µg/mL HS at 35°C for 1 h, then mixed with the virus and infected RD cells for 24 h. The cells were stained using crystal violet and measured at 550 nm. Data are presented as mean ± SD by three independent experiments. Statistical analysis was performed using a one-way ANOVA comparison. * indicates <span class="html-italic">p</span> &lt; 0.05; ** indicates <span class="html-italic">p</span> &lt; 0.01; *** indicates <span class="html-italic">p</span> &lt; 0.001; ns indicates no statistical difference. (<b>e</b>,<b>f</b>) Antiviral performance of the P25s interrupted by serial diluted HS was shown using crystal violet and by the measurement at 550 nm. No significant interference was observed between HS and P25.5 with truncated G and F at the N-terminal, together with P25.11 removing the positive-charged amino acids. The findings suggested that positively charged amino acids were not the sole residues for binding with negatively charged HS.</p>
Full article ">Figure 4
<p>Co-treatment of P25, P25.8, P25.9, and P25.M reduced CPE caused by human enteroviruses. P25, P25.8, P25.9, and P25.M co-treatment with enteroviruses from species A, B, C, and D at 35 °C for 1 h demonstrated anti-CPE effects. P25.8, P25.9, and P25.M have a broad spectrum of antiviral activity. P25.5 served as the negative control. Anti-CPE effects were nonlinear curve-fitted on four represented enteroviruses at 100 TCID<sub>50</sub>/50 µL. (<b>a</b>) EV-A71/SZK2021, which is not a HS-related strain. (<b>b</b>) Echo 30/WZ16. (<b>c</b>) Poliovirus 3/nOPV3. (<b>d</b>) EV-D68/BCH895A. Experiments were performed in triplicate. IC<sub>50</sub> values are shown at <a href="#biomolecules-14-01331-t001" class="html-table">Table 1</a>. (<b>e</b>) 5 × 10<sup>4</sup> RD cells were seeded 12 h before infection, infected with 10 TCID<sub>50</sub>/50 µL EV-D68 in the presence of P25.5, P25.8, P25.9, and P25.M at serial concentration for 1 h, and washed with DMEM twice, then replaced with DMEM for 24 h incubation. Viral VP1 protein was immune-stained and the stained focus was calculated using ImageJ (Version 1.54g).</p>
Full article ">Figure 5
<p>P25s binding and thermostabilization of the virion. (<b>a</b>) EV-D68/BCH895A captured using a P25s coating ELISA and detected using anti-EV-D68 polyclonal Abs with a secondary antibody conjugated with HRP, binding affinities were compared by the fold increase normalized to the blank baseline (PBS). The procedure was described as in the Materials and Methods section. (<b>b</b>–<b>e</b>) The viral thermostabilization in the presence of P25, P25.5, P25.8, P25.9, and P25.M. About 4 µg of virus were mixed with 1.875 µg of P25, P25.5, P25.8, P25.9, and P25.M in 20 µL at 37 °C for 15 min and the temperature was subsequently increased to 90 °C, recording 10 points of the fluorescence signal at 1˚C intervals. The normalized genome release fluorescence dynamics and the first derivatives of EV-D68 (<b>b</b>,<b>c</b>) and Echo 30 (<b>d</b>,<b>e</b>) are shown. (<b>f</b>) The breakpoint temperature for genome release was calculated using the derivative of the fluorescence signal as the peak value. P25.8, P25.9, and P25.M increased the breakpoint temperature for the release of viral genome compared with P25.5 by approximately 5 °C for EV-D68/BCH895A and by 2–5 °C for Echo 30/WZ16. (<b>g</b>) P25.9 and P25.M retained the infectivity of Echo 30/WZ16. 10<sup>6</sup> TCID<sub>50</sub>/mL of Echo 30/WZ16 was co-incubated with an equal volume of the peptide at a concentration of 62.5 µg/mL at 37 °C for 15 min and 45 °C for 2 min, respectively, followed by rapid cooling on ice. The virus titer was determined using TCID<sub>50</sub> as described in the Materials and Methods section. The experiment was repeated in triplicate. Statistical analysis was performed using paired two-tailed <span class="html-italic">t</span>-test. ** indicates <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 6
<p>P25 mutants docking to Echo 30 (PDB: 7C9S). Molecular docking analysis of P25s and the pentamer of Echo 30 using LeDock. The pocket opening of Echo 30 was marked with a black asterisk (*) and a single protomer was colored light blue to distinguish the other two neighboring protomers. The best scored pose was visualized using Pymol. (<b>a</b>) Pocket opening. (<b>b</b>) P25.9 (magenta). (<b>c</b>) P25.M (black). (<b>d</b>) Vacuum electrostatics on the surface of the pentamer with the peptide of P25.8 (green), P25.9 (magenta), and P25.M (black), where the pocket opening area has a higher potency for protein contact. Red indicates negative electrostatic potential energy; blue indicates positive electrostatic potential energy.</p>
Full article ">Figure 7
<p>P25.M reduced the production of infectious virions. RD cells were infected with 100 TCID<sub>50</sub> of enteroviruses for 1 h and washed using DMEM twice, then replaced with P25.M and P25.5 at a concentration of 62.5 µg/mL for a 24 h treatment, respectively. P25.5 served as the control. Data were presented in three independent experiments. Statistical analysis was performed using a paired two-tailed <span class="html-italic">t</span>-test. * indicates <span class="html-italic">p</span> &lt; 0.05; ** indicates <span class="html-italic">p</span> &lt; 0.01; (<b>a</b>) EV-A71/SZK2021. (<b>b</b>) Echo 30/WZ16. (<b>c</b>) Poliovirus 3/nOPV3. (<b>d</b>) EV-D68/BCH895A. (<b>e</b>) 5 × 10<sup>4</sup> RD cells were seeded 12 h before infection, infected with 10 TCID<sub>50</sub>/50 µL of EV-D68 for 1 h and washed with DMEM twice, then replaced with P25.5, P25.8, P25.9, and P25.M at serial concentrations for a 24 h treatment. Viral VP1 was immune-stained and the stained area was calculated using ImageJ (Version 1.54g). P25.9 and P25.M at a concentration of 125 µg/mL, significantly inhibited the synthesis of the viral protein. (<b>f</b>) Western blotting for viral proteins. The infected cells in the presence of 62.5 µg/mL peptides were harvested, and the density of the viral protein band was calculated using ImageJ and normalized to β-actin as 100%. (The original image can be found in <a href="#app1-biomolecules-14-01331" class="html-app">Supplementary File S1</a>). The grey, green, pink and black color denotes the treatment with p25.5, p25.8, p25.9 and P25.M, respectively.</p>
Full article ">
26 pages, 1530 KiB  
Review
A Survey on Computational Methods in Drug Discovery for Neurodegenerative Diseases
by Caterina Vicidomini, Francesco Fontanella, Tiziana D’Alessandro and Giovanni N. Roviello
Biomolecules 2024, 14(10), 1330; https://doi.org/10.3390/biom14101330 - 19 Oct 2024
Cited by 4 | Viewed by 1907
Abstract
Currently, the age structure of the world population is changing due to declining birth rates and increasing life expectancy. As a result, physicians worldwide have to treat an increasing number of age-related diseases, of which neurological disorders represent a significant part. In this [...] Read more.
Currently, the age structure of the world population is changing due to declining birth rates and increasing life expectancy. As a result, physicians worldwide have to treat an increasing number of age-related diseases, of which neurological disorders represent a significant part. In this context, there is an urgent need to discover new therapeutic approaches to counteract the effects of neurodegeneration on human health, and computational science can be of pivotal importance for more effective neurodrug discovery. The knowledge of the molecular structure of the receptors and other biomolecules involved in neurological pathogenesis facilitates the design of new molecules as potential drugs to be used in the fight against diseases of high social relevance such as dementia, Alzheimer’s disease (AD) and Parkinson’s disease (PD), to cite only a few. However, the absence of comprehensive guidelines regarding the strengths and weaknesses of alternative approaches creates a fragmented and disconnected field, resulting in missed opportunities to enhance performance and achieve successful applications. This review aims to summarize some of the most innovative strategies based on computational methods used for neurodrug development. In particular, recent applications and the state-of-the-art of molecular docking and artificial intelligence for ligand- and target-based approaches in novel drug design were reviewed, highlighting the crucial role of in silico methods in the context of neurodrug discovery for neurodegenerative diseases. Full article
Show Figures

Figure 1

Figure 1
<p>Molecular docking pose views of the complex between solasodine (yellow) and N-type calcium channel (PDB code 3DVE; receptor style: cartoon) as obtained by blind docking with HDOCK server (top-1 pose <a href="http://hdock.phys.hust.edu.cn/" target="_blank">http://hdock.phys.hust.edu.cn/</a>, accessed on 6 July 2023). The above pictures are property of the authors of the present review article.</p>
Full article ">Figure 2
<p>Applications of AI in drug discovery.</p>
Full article ">Scheme 1
<p>Structural representation of some putative neurodrugs investigated by molecular docking.</p>
Full article ">
17 pages, 971 KiB  
Review
Aspirin Hypersensitivity in Patients with Coronary Artery Disease: An Updated Review and Practical Recommendations
by Luigi Cappannoli, Stefania Colantuono, Francesco Maria Animati, Francesco Fracassi, Mattia Galli, Cristina Aurigemma, Enrico Romagnoli, Rocco Antonio Montone, Mattia Lunardi, Lazzaro Paraggio, Carolina Ierardi, Ilaria Baglivo, Cristiano Caruso, Carlo Trani and Francesco Burzotta
Biomolecules 2024, 14(10), 1329; https://doi.org/10.3390/biom14101329 - 19 Oct 2024
Viewed by 1822
Abstract
Acetylsalicylic acid (ASA) represents a cornerstone of antiplatelet therapy for the treatment of atherosclerotic coronary artery disease (CAD). ASA is in fact indicated in case of an acute coronary syndrome or after a percutaneous coronary intervention with stent implantation. Aspirin hypersensitivity is frequently [...] Read more.
Acetylsalicylic acid (ASA) represents a cornerstone of antiplatelet therapy for the treatment of atherosclerotic coronary artery disease (CAD). ASA is in fact indicated in case of an acute coronary syndrome or after a percutaneous coronary intervention with stent implantation. Aspirin hypersensitivity is frequently reported by patients, and this challenging situation requires a careful evaluation of the true nature of the presumed sensitivity and of its mechanisms, as well as to differentiate it from a more frequent (and more easily manageable) aspirin intolerance. Two main strategies are available to allow ASA administration for patients with CAD and suspected ASA hypersensitivity: a low-dose ASA challenge, aimed at assessing the tolerability of ASA at the antiplatelet dose of 100 mg, and desensitization, a therapeutic procedure which aims to induce tolerance to ASA. For those patients who cannot undergo ASA challenge and desensitization due to previous serious adverse reactions, or for those in whom desensitization was unsuccessful, a number of further alternative strategies are available, even if these have not been validated and approved by guidelines. The aim of this state-of-the-art review is therefore to summarize the established evidence regarding pathophysiology, clinical presentation, diagnosis, and management of aspirin hypersensitivity and to provide a practical guide for cardiologists (and clinicians) who have to face the not uncommon situation of a patient with concomitant coronary artery disease and aspirin hypersensitivity. Full article
(This article belongs to the Special Issue New Discoveries in Biological Functions of Platelet)
Show Figures

Figure 1

Figure 1
<p>Arachidonic acid pathway in inflammation. The Figure shows the arachidonic acid pathway and the formation of its metabolites (leukotrienes, prostaglandins, and thromboxane) through cyclooxygenase (COX-1 and COX-2) action. Acetylsalicylic acid, blocking COXs, inhibits metabolites effects and exerts anti-inflammatory and antiplatelet function. ASA: acetylsalicylic acid; PG: prostaglandin; TxA<sub>2</sub>: thromboxane A2.</p>
Full article ">Figure 2
<p>ASA hypersensitivity: diagnostic and therapeutic algorithm. When ASA hypersensitivity is suspected in a patient with atherosclerotic coronary artery disease, a diagnostic workout to confirm or exclude hypersensitivity is mandatory in order to perform percutaneous coronary interventions safely. A detailed clinical history can distinguish ASA intolerance from hypersensitivity signs and symptoms, above all in the presence of comorbidities (chronic rhinosinusitis, nasal polyps, asthma, food allergy). If ASA hypersensitivity is confirmed (or still suspected) in an emergency setting, PCI should be performed by administering an alternative antiplatelet drug. If patient conditions allow for 24–48 h waiting and allergologist consultation is available, LDAC and/or desensitization should be performed. ASA: acetylsalicylic acid; CAD: coronary artery disease; LDAC: low-dose ASA challenge; PCI: percutaneous coronary interventions.</p>
Full article ">
17 pages, 2211 KiB  
Article
The Biological Effect of Enriching the Plasma Content in Platelet-Rich Plasma: An In Vitro Study
by Eduardo Anitua, Mar Zalduendo, Roberto Prado, María Troya, Roberto Tierno, María de la Fuente and Mohammad H. Alkhraisat
Biomolecules 2024, 14(10), 1328; https://doi.org/10.3390/biom14101328 - 18 Oct 2024
Viewed by 1006
Abstract
BACKGROUND: Platelet-rich plasma (PRP) formulations have become valuable therapeutic tools in regenerative medicine. In addition, these blood derivates have been successfully included in cell therapy as fetal bovine serum substitutes, due to the real need to avoid the risk of host immunologic reactions [...] Read more.
BACKGROUND: Platelet-rich plasma (PRP) formulations have become valuable therapeutic tools in regenerative medicine. In addition, these blood derivates have been successfully included in cell therapy as fetal bovine serum substitutes, due to the real need to avoid the risk of host immunologic reactions and the animal disease transmission associated with reagents from animal origin. However, the protocols for obtaining them should be optimized to improve their biological potential. METHODS: PRP-derived preparations with different concentrations of the platelet and plasma components were obtained from the blood of five donors by freeze-drying. Measurements of the pH, protein, and growth factor concentration were performed. Moreover, their biological effects on cell proliferation and migration and their angiogenic potential were assessed. RESULTS: An increased plasma component concentration resulted in an augmented quantity of the total protein content, a significative variation in the hepatocyte growth factor concentration, and an experimental but clinically irrelevant alteration of the pH value. No significant changes were induced in their potential to enhance proliferative and migratory responses in epithelial cells, with the latter being reduced for dermal fibroblasts. The endothelial cell capacity for tube formation was significatively reduced. CONCLUSIONS: An increased blood plasma content did not improve the biological potential of the formulations. However, they have emerged as a promising approach for regenerative therapies where neovascularization must be avoided. Full article
Show Figures

Figure 1

Figure 1
<p>Schematic illustration showing the process for obtaining the 6 supernatant preparations prepared from plasma rich in growth factors (PRGF). PRGF: Plasma Rich in Growth Factors. 1-0 formulation: preparation with 1× plasma content and no platelets. 1-2 PRP: Plasma Rich in Platelets with 1× and 2× concentration factors in plasma and platelet contents, respectively. 1-4 PRP: Plasma Rich in Platelets with 1× and 4× concentration factors in plasma and platelet contents, respectively. Snt: supernatant for cell culture medium supplementation. 1-2 Snt: supernatant with 1× and 2× plasma and platelet content, respectively. 2-2 Snt: supernatant with 2× plasma and platelet content. 3-2 Snt: supernatant with 3× and 2× plasma and platelet content, respectively. 1-4 Snt: supernatant with 1× and 4× plasma and platelet content, respectively. 2-4 Snt: supernatant with 2× and 4× plasma and platelet content, respectively. 3-4 Snt: supernatant with 3× and 4× plasma and platelet content, respectively. Created with BioRender.com.</p>
Full article ">Figure 2
<p>Characterization of the six supernatants obtained from formulations with different plasma and platelet contents. Values of pH (<b>A</b>), protein concentration (<b>B</b>), and growth factors concentrations (<b>C</b>) for the six preparations are graphically represented. Moreover, the effects of both the plasmatic and the platelet factors on these parameters were individually analyzed (<b>D</b>–<b>F</b>). 1-2 Snt: supernatant with the same plasma content (one times the plasma) and twice the platelet content of the peripheral blood (two times the platelets). 2-2 Snt: supernatant with twice the platelet and plasma contents of the peripheral blood. 3-2 Snt: supernatant with three- and two-times higher concentrations of plasma and platelet contents, respectively, than the peripheral blood. 1-4 Snt: supernatant with the same plasma content and a platelet concentration four times higher than the peripheral blood. 2-4 Snt: supernatant with twice the plasma content and a four-times higher concentration of platelet-derived factors, with respect to the blood. 3-4 Snt: supernatant with three and four times the concentrations of plasma and platelets, respectively, compared to the peripheral blood. <span class="html-italic">ØStatistically significant differences with respect to the 2-4 Snt, 3-2 Snt, and 3-4 Snt groups. βStatistically significant differences with respect to the supernatants derived from formulations with different plasmatic factors. ¶Statistically significant differences with respect to the 1-2 Snt and 1-4 Snt groups. γStatistically significant differences with respect to the two times and three times plasmatic factor groups. ‡Statistically significant differences with respect to the four times platelet factor group. ¥Statistically significant differences with respect to the 3-4 Snt group (p ≤ 0.05) (n = 5)</span>.</p>
Full article ">Figure 3
<p>Cell proliferation analysis. Cells from two phenotypes were treated with supernatants (Snts) derived from six PRP-derived formulations. The code of these supernatants consisted of two numbers referring to the plasma and platelet contents, respectively, compared to the peripheral blood. A double statistical analysis was performed to determine the effect of these supernatants on the proliferation rate induced by the specific composition of both plasma and platelet-derived factors (<b>A</b>), and, in addition, that induced by the plasma- or platelet-derived factors considered separately (<b>B</b>). The results are expressed as a percentage of the proliferation achieved by the cells maintained with the routine culture medium specific for each cell phenotype (the positive control). HDFs: human dermal fibroblasts; and HCE-1: human corneal epithelial cell. <span class="html-italic"><span>$</span>Statistically significant differences with respect to the three times plasmatic factor group</span> (<span class="html-italic">p ≤ 0.05</span>) (<span class="html-italic">n = 5</span>).</p>
Full article ">Figure 4
<p>Cell chemotaxis analysis. Cells from two phenotypes were treated with supernatants (Snts) derived from six PRP-derived formulations. The code of these PRPs consisted of two numbers referring to the plasma and platelet contents, respectively, compared to the peripheral blood. A double statistical analysis was performed to determine the effect of these supernatants on the migration rate induced by the specific composition of both plasma- and platelet-derived factors (<b>A</b>) and, in addition, that induced by plasma or platelet-derived factors considered individually (<b>B</b>). The results are expressed as a percentage of the proliferation achieved by the cells maintained with the routine culture medium specific for each cell phenotype (the positive control). HDFs: human dermal fibroblasts; and HCE-1: human corneal epithelial cell (<span class="html-italic">n = 5</span>).</p>
Full article ">Figure 5
<p>Endothelial cell proliferation analysis and angiogenesis assay. The endothelial cells were treated with supernatants derived from PRP with increasing concentrations of plasmatic factors and setting the platelet content at twice that of the peripheral blood (1-2 Snt, 2-2 Snt, and 3-2 Snt). The proliferation results are expressed as a percentage of the proliferation achieved by the cells under optimal conditions, maintained with the routine culture medium (the positive control) (<b>A</b>). For the angiogenesis assay, the endothelial cells were seeded in a µ-plate with 96 wells for a high-throughput 3D cell culture and tube formation assays in which a solubilized basement membrane preparation was previously added. The covered area (<b>B</b>), the number of loops (<b>C</b>), the total tube length (<b>D</b>), the total tubes (<b>E</b>), and the total branching points (<b>F</b>), considered as the main parameters of the angiogenic process, were measured. The results are expressed as a percentage with respect to the positive control (the routine culture medium). <span class="html-italic">&amp;Statistically significant differences with respect to the 3-2 Snt group. (p ≤ 0.05).</span> HUVECs: human umbilical vein endothelial cells <span class="html-italic">(n = 5)</span>.</p>
Full article ">
15 pages, 2592 KiB  
Article
Pulling Forces Differentially Affect Refolding Pathways Due to Entangled Misfolded States in SARS-CoV-1 and SARS-CoV-2 Receptor Binding Domain
by Pham Dang Lan, Edward P. O’Brien and Mai Suan Li
Biomolecules 2024, 14(10), 1327; https://doi.org/10.3390/biom14101327 - 18 Oct 2024
Viewed by 1335
Abstract
Single-molecule force spectroscopy (SMFS) experiments can monitor protein refolding by applying a small force of a few piconewtons (pN) and slowing down the folding process. Bell theory predicts that in the narrow force regime where refolding can occur, the folding time should increase [...] Read more.
Single-molecule force spectroscopy (SMFS) experiments can monitor protein refolding by applying a small force of a few piconewtons (pN) and slowing down the folding process. Bell theory predicts that in the narrow force regime where refolding can occur, the folding time should increase exponentially with increased external force. In this work, using coarse-grained molecular dynamics simulations, we compared the refolding pathways of SARS-CoV-1 RBD and SARS-CoV-2 RBD (RBD refers to the receptor binding domain) starting from unfolded conformations with and without a force applied to the protein termini. For SARS-CoV-2 RBD, the number of trajectories that fold is significantly reduced with the application of a 5 pN force, indicating that, qualitatively consistent with Bell theory, refolding is slowed down when a pulling force is applied to the termini. In contrast, the refolding times of SARS-CoV-1 RBD do not change meaningfully when a force of 5 pN is applied. How this lack of a Bell response could arise at the molecular level is unknown. Analysis of the entanglement changes of the folded conformations revealed that in the case of SARS-CoV-1 RBD, an external force minimizes misfolding into kinetically trapped states, thereby promoting efficient folding and offsetting any potential slowdown due to the external force. These misfolded states contain non-native entanglements that do not exist in the native state of either SARS-CoV-1-RBD or SARS-CoV-2-RBD. These results indicate that non-Bell behavior can arise from this class of misfolding and, hence, may be a means of experimentally detecting these elusive, theoretically predicted states. Full article
(This article belongs to the Section Biomacromolecules: Proteins)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Representative native entanglements in the native structures of SARS-CoV-1 RBD and SARS-CoV-2 RBD. These structures were obtained using AlphaFold2 and MD simulations. The loops (colored red) are closed by native contacts (colored yellow). (<b>Left</b>) The loop is closed by the native contact between residue 99 and 136, with both N and C termini threading through the loop. Note that only the C-terminal thread is colored blue for more visibility. (<b>Right</b>) The loop is closed by the native contact between residue 108 and 146, with the C-terminal thread highlighted in blue.</p>
Full article ">Figure 2
<p>The root mean square deviation (RMSD) and the radius of gyration (Rg) of the backbone from three 500 ns all-atom MD simulations for (<b>a</b>) SARS-CoV-1 RBD and (<b>b</b>) SARS-CoV-2 RBD.</p>
Full article ">Figure 3
<p>Dependence of −ln(<span class="html-italic">P</span>) on <span class="html-italic">Q</span>, where <span class="html-italic">P</span> is the probability of sampling a particular <span class="html-italic">Q</span> value of (<b>a</b>) SARS-CoV-1 RBD and (<b>b</b>) SARS-CoV-2 RBD. An additional minimum is observed at <span class="html-italic">Q</span>~0.2, which occurs in the presence of the external force.</p>
Full article ">Figure 4
<p>Misfolded states were observed during the folding of the SARS-CoV-1 RBD. Force may promote the folding process of the SARS-CoV-1 RBD, resulting in an increase in the percentage of trajectories that follow paths leading to correct folding. (<b>a</b>,<b>b</b>) represent −ln(<span class="html-italic">P</span>) surfaces, where <span class="html-italic">P</span> is the probability of sampling <span class="html-italic">Q</span> and <span class="html-italic">G</span> values during refolding simulations with <span class="html-italic">F</span> = 0 and <span class="html-italic">F</span> = 5 pN, respectively. (<b>c</b>,<b>d</b>) are transition networks of discrete trajectories along metastable states represented by nodes. Blue, green and red nodes correspond to folded, intermediate and misfolded states, respectively, that trajectories visited at the end of simulations. Red numbers indicate the number of transitions between states, and black numbers represent nodes.</p>
Full article ">Figure 5
<p>Misfolded states were also observed in the folding of the SARS-CoV-2 RBD. The force effect results in 43% of trajectories remaining in the unfolded state 1 at the end of the simulation. (<b>a</b>,<b>b</b>) represent the −ln(<span class="html-italic">P</span>) surfaces, where <span class="html-italic">P</span> is the probability of sampling certain values of <span class="html-italic">Q</span> and <span class="html-italic">G</span> from refolding simulations with <span class="html-italic">F</span> = 0 and <span class="html-italic">F</span> = 5 pN, respectively. (<b>c</b>,<b>d</b>) are transition networks of discrete trajectories along metastable states represented by nodes. Blue, green and red nodes correspond to folded, intermediate and misfolded states, respectively, that trajectories visited at the end of the simulations. The black node indicates that trajectories remain in the unfolded state 1 at the end of the simulations. Red numbers indicate the number of transitions between states, and black numbers represent nodes.</p>
Full article ">Figure 6
<p>Conformational distribution along the <span class="html-italic">G</span> parameter of (<b>a</b>) SARS-CoV-1 RBD and (<b>b</b>) SARS-CoV-2 RBD with peaks are concise with characteristics of (<span class="html-italic">Q</span>, <span class="html-italic">G</span>) log probability surfaces, indicating the presence of structures with change in entanglement at different levels, namely <span class="html-italic">G</span>~0.05–0.1, 0.15–0.2, 0.2–0.25.</p>
Full article ">
23 pages, 3110 KiB  
Article
From Organotypic Mouse Brain Slices to Human Alzheimer’s Plasma Biomarkers: A Focus on Nerve Fiber Outgrowth
by Sakir Necat Yilmaz, Katharina Steiner, Josef Marksteiner, Klaus Faserl, Mathias Villunger, Bettina Sarg and Christian Humpel
Biomolecules 2024, 14(10), 1326; https://doi.org/10.3390/biom14101326 - 18 Oct 2024
Viewed by 968
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disease characterized by memory loss and progressive deterioration of cognitive functions. Being able to identify reliable biomarkers in easily available body fluids such as blood plasma is vital for the disease. To achieve this, we used a [...] Read more.
Alzheimer’s disease (AD) is a neurodegenerative disease characterized by memory loss and progressive deterioration of cognitive functions. Being able to identify reliable biomarkers in easily available body fluids such as blood plasma is vital for the disease. To achieve this, we used a technique that applied human plasma to organotypic brain slice culture via microcontact printing. After a 2-week culture period, we performed immunolabeling for neurofilament and myelin oligodendrocyte glycoprotein (MOG) to visualize newly formed nerve fibers and oligodendrocytes. There was no significant change in the number of new nerve fibers in the AD plasma group compared to the healthy control group, while the length of the produced fibers significantly decreased. A significant increase in the number of MOG+ dots around these new fibers was detected in the patient group. According to our hypothesis, there are factors in the plasma of AD patients that affect the growth of new nerve fibers, which also affect the oligodendrocytes. Based on these findings, we selected the most promising plasma samples and conducted mass spectrometry using a differential approach and we identified three putative biomarkers: aldehyde-dehydrogenase 1A1, alpha-synuclein and protein S100-A4. Our method represents a novel and innovative approach for translating research findings from mouse models to human applications. Full article
(This article belongs to the Section Molecular Biomarkers)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Experimental design of this study. Plasma samples from Alzheimer’s disease (AD) patients, mild cognitive impairment (MCI) patients and healthy controls (Co) were used for experiments. Mouse organotypic brain slices were coupled with plasma microcontact (µC) printed membranes and cultured for 2 weeks. At the end of the culture period, brain slices were fixed and labeled for neurofilament (NF), microtubule-associated protein-2 (MAP2) or myelin oligodendrocyte glycoprotein (MOG). For mass spectrometry analysis, selected plasma samples were used.</p>
Full article ">Figure 2
<p>Immunolabeling of neurofilament (NF) and microtubule-associated protein-2 (MAP2). (<b>A</b>) Organotypic brain slices from the hippocampal level were cultured on an extra membrane in semipermeable inserts. (<b>B</b>) The scheme shows that these slices were connected to microcontact prints loaded with plasma, allowing nerve fibers to grow along them. (<b>C</b>) The microcontact prints can be visualized by additionally loading a red-fluorescent Alexa 546 antibody to assess the efficiency, appearing as red lines perpendicular to the border of the slice (dashed line). (<b>D</b>) Nerve fibers are stained with neurofilament (green, Alexa 488) and (<b>E</b>) with MAP2 (red, Alexa 546). (<b>F</b>) This panel shows the merged staining of NF and MAP2. The slices were counterstained with nuclear DAPI (blue). Scale bar = 1 cm (<b>A</b>), 75 µm (<b>C</b>), 25 µm (<b>D</b>–<b>F</b>).</p>
Full article ">Figure 3
<p>Immunolabeling of neurofilament (NF)-positive nerve fibers and myelin oligodendrocyte glycoprotein (MOG)-positive dots. Organotypic brain slices were cultured on plasma microcontact prints for 2 weeks. (<b>A</b>) Following fixation, they were stained for NF (green, Alexa 488) and (<b>B</b>) MOG (red, Alexa 546). (<b>C</b>) This panel shows the merged staining of NF and MOG. The slices were counterstained with nuclear DAPI (blue). The dashed white line indicates the border of the brain slices. The arrows point to specific examples of MOG-positive dots (bottom arrow) and negative staining (top arrow). (<b>D</b>,<b>E</b>) This panel shows an example at higher magnification of MOG+ dots located along NF+ nerve fibers. Scale bar = 75 µm (<b>A</b>–<b>C</b>), 100 µm (<b>D</b>), 20 µm (<b>E</b>).</p>
Full article ">Figure 4
<p>Immunolabeling of neurofilament (NF)-positive nerve fibers and myelin oligodendrocyte glycoprotein (MOG)-positive staining on healthy control plasma microcontact prints compared to those from Alzheimer’s samples. Organotypic brain slices were cultured on plasma microcontact prints for 2 weeks. (<b>A</b>,<b>D</b>) Following fixation, they were stained for NF (green, Alexa 488) and (<b>B</b>,<b>E</b>) MOG (red, Alexa 546). (<b>C</b>,<b>F</b>) These panels show the merged staining of NF and MOG. Note the MOG+ dots along the nerve fibers (indicated by arrows), which are more prominent and larger in the Alzheimer’s group. Scale bar = 20 µm (<b>A</b>–<b>F</b>).</p>
Full article ">Figure 5
<p>The best six plasma samples (three healthy controls and three Alzheimer’s disease) were selected based on the results of measurements and counting on NF+ axons and MOG+ dot structures. Proteins identified after depletion of 14 proteins are shown. The keywords “oligodendrocyte”, or “axon” or “myelin” were searched in combination with differential mass spectrometry results and ultimately three possible biomarkers were identified.</p>
Full article ">
10 pages, 1292 KiB  
Article
Endometrial Dysbiosis: A Possible Association with Estrobolome Alteration
by Giorgia Scarfò, Simona Daniele, Elisa Chelucci, Francesca Papini, Francesco Epifani, Maria Ruggiero, Vito Cela, Ferdinando Franzoni and Paolo Giovanni Artini
Biomolecules 2024, 14(10), 1325; https://doi.org/10.3390/biom14101325 - 18 Oct 2024
Viewed by 984
Abstract
Background/Objectives: Microbiota modification at the endometrial level can favor gynecological diseases and impair women’s fertility. The overgrowth of pathogen microorganisms is related to the contemporary alteration of estrogen-metabolizing bacteria, including β-glucuronidase, thereby enhancing estrogen-related inflammatory states and decreasing anti-inflammatory cells. The possible connection [...] Read more.
Background/Objectives: Microbiota modification at the endometrial level can favor gynecological diseases and impair women’s fertility. The overgrowth of pathogen microorganisms is related to the contemporary alteration of estrogen-metabolizing bacteria, including β-glucuronidase, thereby enhancing estrogen-related inflammatory states and decreasing anti-inflammatory cells. The possible connection between estrobolome impairment and gynecological diseases has been suggested in animal models. Nevertheless, in humans, coherent evidence on the estrobolome alteration and functionality of the female reproductive tract is still lacking. The objective of this study was to explore alterations in estrogen-related signaling and the putative link with endometrial dysbiosis. Methods: Women with infertility and repeated implantation failure (RIF, N = 40) were enrolled in order to explore the putative link between estrogen metabolism and endometrial dysbiosis. Endometrial biopsies were used to measure inflammatory and growth factor molecules. β-glucuronidase enzyme activity and estrogen receptor (ER) expression were also assessed. Results: Herein, increased levels of inflammatory molecules (i.e., IL-1β and HIF-1α) and decreased levels of the growth factor IGF-1 were found in the endometrial biopsies of patients presenting dysbiosis compared to eubiotic ones. β-glucuronidase activity and the expression of ERβ were significantly enhanced in patients in the dysbiosis group. Interestingly, Lactobacilli abundance was inversely related to β-glucuronidase activity and to ERβ expression, thus suggesting that an alteration of the estrogen-activating enzyme may affect the expression of ERs as well. Conclusions. Overall, these preliminary data suggested a link between endometrial dysbiosis and estrobolome impairment as possible synergistic contributing factors to women infertility and RIF. Full article
(This article belongs to the Special Issue Molecular Aspects of Female Infertility)
Show Figures

Figure 1

Figure 1
<p>Correlations between ER β and IL−1 β (<b>A</b>); Correlations Between β glucuronidase and IL−1 β and IL−8 (<b>B</b>,<b>C</b>). All correlations between the selected variables were performed using simple linear regression analysis using the StatView program (Abacus Concepts, Inc., SAS Institute, Cary, NC, USA). <span class="html-italic">p</span> and R2 values obtained for each correlation are reported in the text.</p>
Full article ">Figure 2
<p>Correlations of Lactobacilli and IL−1 β (<b>A</b>), β glucuronidase (<b>B</b>), and ER β (<b>C</b>). All correlations between the selected variables were performed by simple linear regression analysis using the StatView program (Abacus Concepts, Inc., SAS Institute, Cary, NC, USA). <span class="html-italic">p</span> and R2 values obtained for each correlation are reported in the respective panel.</p>
Full article ">
15 pages, 577 KiB  
Review
Targeting Protein Aggregation in ALS
by Michele Perni and Benedetta Mannini
Biomolecules 2024, 14(10), 1324; https://doi.org/10.3390/biom14101324 - 18 Oct 2024
Viewed by 1598
Abstract
Proteinopathies involve the abnormal accumulation of specific proteins. Maintaining the balance of the proteome is a finely regulated process managed by a complex network of cellular machinery responsible for protein synthesis, folding, and degradation. However, stress and ageing can disrupt this balance, leading [...] Read more.
Proteinopathies involve the abnormal accumulation of specific proteins. Maintaining the balance of the proteome is a finely regulated process managed by a complex network of cellular machinery responsible for protein synthesis, folding, and degradation. However, stress and ageing can disrupt this balance, leading to widespread protein aggregation. Currently, several therapies targeting protein aggregation are in clinical trials for ALS. These approaches mainly focus on two strategies: addressing proteins that are prone to aggregation due to mutations and targeting the cellular mechanisms that maintain protein homeostasis to prevent aggregation. This review will cover these emerging drugs. Advances in ALS research not only offer hope for better outcomes for ALS patients but also provide valuable insights and methodologies that can benefit the broader field of neurodegenerative disease drug discovery. Full article
(This article belongs to the Special Issue The Role of Amyloid in Neurological Disorders)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Drugs targeting protein aggregation in ALS categorized according to their mode of action.</p>
Full article ">
12 pages, 687 KiB  
Review
The Emerging Role of Immunoglobulins and Complement in the Stimulation of Neuronal Activity and Repair: Not as Simple as We Thought
by Tatyana Veremeyko, Natasha S. Barteneva, Ivan Vorobyev and Eugene D. Ponomarev
Biomolecules 2024, 14(10), 1323; https://doi.org/10.3390/biom14101323 - 18 Oct 2024
Viewed by 1156
Abstract
Neurologic disorders such as traumatic brain injury, multiple sclerosis, Alzheimer’s disease, and drug-resistant epilepsy have a high socioeconomic impact around the world. Current therapies for these disorders are often not effective. This creates a demand for the development of new therapeutic approaches to [...] Read more.
Neurologic disorders such as traumatic brain injury, multiple sclerosis, Alzheimer’s disease, and drug-resistant epilepsy have a high socioeconomic impact around the world. Current therapies for these disorders are often not effective. This creates a demand for the development of new therapeutic approaches to treat these disorders. Recent data suggest that autoreactive naturally occurring immunoglobulins produced by subsets of B cells, called B1 B cells, combined with complement, are actively involved in the processes of restoration of neuronal functions during pathological conditions and remyelination. The focus of this review is to discuss the possibility of creating specific therapeutic antibodies that can activate and fix complement to enhance neuronal survival and promote central nervous system repair after injuries associated with many types of neurodegenerative diseases. Full article
(This article belongs to the Special Issue Recent Advances in Neurological Diseases)
Show Figures

Figure 1

Figure 1
<p>The proposed model of regulation of the CNS repair process by B1-derived NAAs (IgM) and complement subunits C1 and C4 by forming IgM-C1-C4b complexes that affect neuronal cells, microglia, and oligodendrocyte progenitors. The image was created with BioRender.com.</p>
Full article ">
20 pages, 1446 KiB  
Article
The Role of Keeving in Modulating Fermentation and the Flavour Profiles of Apple Brandy
by Magdalena Januszek, Paweł Satora, Aneta Pater and Łukasz Wajda
Biomolecules 2024, 14(10), 1322; https://doi.org/10.3390/biom14101322 - 18 Oct 2024
Cited by 1 | Viewed by 834
Abstract
Keeving is the removal of nutrients from apple musts due to their binding to pectin, resulting in a slower fermentation and spontaneous arrest. The aim of this study was to determine the effect of keeving on the chemical composition of fermented apple must [...] Read more.
Keeving is the removal of nutrients from apple musts due to their binding to pectin, resulting in a slower fermentation and spontaneous arrest. The aim of this study was to determine the effect of keeving on the chemical composition of fermented apple must and on the volatile profile and sensory analysis of apple brandies. We compared the application of keeving during spontaneous fermentation with fermentation carried out by Saccharomyces cerevisiae (SafSpirit HG-1). We evaluated the impact of adding different doses of calcium chloride on various parameters of fermented musts and distillates. Calcium chloride had a greater effect on the ethanol concentration, total extract, and fermentation efficiency than on the type of fermentation used. However, a different phenomenon was observed with respect to the volatiles. The concentration of most of the higher alcohols, acetaldehyde, dodecanal, and geranylaceton, decreased after spontaneous fermentation and increased during the fermentation carried out with Saccharomyces cerevisiae SafSpirit HG-1. In general, the application of keeving contributed to a decrease in the concentration of ethyl and methyl esters, but caused an increase in the concentration of all acetate esters and terpenoids. When the amount of nutrients in the environment is limited and starvation occurs, microorganisms use the available nutrients for basic metabolic processes that allow them to survive and limit the formation of side metabolites such as volatiles. However, most of the samples fermented after the faecal depletion achieved high scores for the floral, fruity, and “overall note” parameters in the sensory analysis. This means that this method, carried out with a properly selected yeast strain, could be feasible for the distilling industry. Full article
(This article belongs to the Section Natural and Bio-derived Molecules)
Show Figures

Figure 1

Figure 1
<p>Dissolution of the gel during the fermentation process after keeving.</p>
Full article ">Figure 2
<p>Principal component analysis showing the effect of different CaCl<sub>2</sub> additions and types of yeast on the chemical composition of apple musts. SF—spontaneous fermentation; Sc—<span class="html-italic">Saccharomyces cerevisiae</span> SafSpirit HG-1. Doses of CaCl<sub>2</sub>: 0—0 g/L; 01—0.1 g/L; 02—0.2 g/L; and 04—0.4 g/L.</p>
Full article ">Figure 3
<p>Principal component analysis for oenological parameters of fermented apple musts.</p>
Full article ">Figure 4
<p>Principal component analysis showing the effect of different CaCl<sub>2</sub> additions and types of yeast on the volatile compounds present in apple spirits. SF—spontaneous fermentation; Sc—<span class="html-italic">Saccharomyces cerevisiae</span> SafSpirit HG-1. Doses of CaCl<sub>2:</sub> 0—0 g/L; 01—0.1 g/L; 02—0.2 g/L; 04—0.4 g/L.</p>
Full article ">Figure 5
<p>Principal component analysis showing the effect of different CaCl<sub>2</sub> additions and types of yeast on the volatile compounds present in apple spirits. Esters (E); Methanol (M); Higher Alcohols (A); Aldehydes and ketones (Al), Terpenoids (T); Other compounds (O). The reference of the compound name to the symbol is summarized in <a href="#biomolecules-14-01322-t003" class="html-table">Table 3</a>.</p>
Full article ">Figure 6
<p>Characteristic aroma features of apple brandies obtained from musts fermented with the addition of CaCl<sub>2</sub>; n = 5, STD &lt; 5%. Sp—spontaneous fermentation; Sc—<span class="html-italic">Saccharomyces cerevisiae</span> SafSpirit HG-1 (0; 0.1; 0.2; 0.4 g/L—doses of CaCl<sub>2</sub> added); *, **, and ***—the significance at 0.05, 0.01, and 0.005 by least significant difference, respectively.</p>
Full article ">
22 pages, 7042 KiB  
Article
Glucocorticoid-Induced Leucine Zipper Protein and Yeast-Extracted Compound Alleviate Colitis and Reduce Fungal Dysbiosis
by Marco Gentili, Samuele Sabbatini, Emilia Nunzi, Eleonora Lusenti, Luigi Cari, Antonella Mencacci, Nathalie Ballet, Graziella Migliorati, Carlo Riccardi, Simona Ronchetti and Claudia Monari
Biomolecules 2024, 14(10), 1321; https://doi.org/10.3390/biom14101321 - 17 Oct 2024
Viewed by 1014
Abstract
Inflammatory bowel diseases (IBD) have a complex, poorly understood pathogenesis and lack long-lasting effective treatments. Recent research suggests that intestinal fungal dysbiosis may play a role in IBD development. This study investigates the effects of the glucocorticoid-induced leucine zipper protein (GILZp)”, known for [...] Read more.
Inflammatory bowel diseases (IBD) have a complex, poorly understood pathogenesis and lack long-lasting effective treatments. Recent research suggests that intestinal fungal dysbiosis may play a role in IBD development. This study investigates the effects of the glucocorticoid-induced leucine zipper protein (GILZp)”, known for its protective role in gut mucosa, and a yeast extract (Py) with prebiotic properties, either alone or combined, in DSS-induced colitis. Both treatments alleviated symptoms via overlapping or distinct mechanisms. In particular, they reduced the transcription levels of pro-inflammatory cytokines IL-1β and TNF-α, as well as the expression of the tight junction protein Claudin-2. Additionally, GILZp increased MUC2 transcription, while Py reduced IL-12p40 and IL-6 levels. Notably, both treatments were effective in restoring the intestinal burden of clinically important Candida and related species. Intestinal mycobiome analysis revealed that they were able to reduce colitis-associated fungal dysbiosis, and this effect was mainly the result of a decreased abundance of the Meyerozima genus, which was dominant in colitic mice. Overall, our results suggest that combined treatment regimens with GILZp and Py could represent a new strategy for the treatment of IBD by targeting multiple mechanisms, including the fungal dysbiosis. Full article
(This article belongs to the Section Molecular Medicine)
Show Figures

Figure 1

Figure 1
<p>Ameliorative effect of individual and combined GILZp and Py treatments on DSS-induced colitis symptoms in mice. (<b>A</b>) Schematic diagram of the experimental design and procedures. Colitis was induced with the administration of 3% DSS for 5 days. From day 0 to day 8, the four colitic groups received GILZp (0.2 mg/kg), Py (1000 mg/kg), a Py + GILZp combination, or PBS. The schematic was created with Biorender BioRender (HomeStars, Toronto, ON, Canada)). (<b>B</b>) Kaplan–Meier curve depicting mortality in mice with DSS-induced colitis. (<b>C</b>,<b>D</b>) Body weight loss and clinical score (DAI) were registered daily over the whole experimental period. (<b>E</b>) Mean weight/length ratio values of colons. Values are expressed as the means ± SEM value from two independent experiments (<span class="html-italic">n</span> = 5–7 for each group). * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001. Where not indicated, a non-significant difference was observed.</p>
Full article ">Figure 2
<p>Suppression of the expression of inflammatory cytokines and restoration of intestinal barrier integrity with individual and combined GILZp and Py treatments. Quantitative RT–PCR analysis of IL-1β and TNF-α (<b>A</b>) expression, (<b>B</b>) IL-6 and IL-12p40 expression, and (<b>C</b>) Claudin-2 and MUC2 expression in the colons of non-colitic and colitic mice. The colitic mice were treated as indicated. Values are expressed as means ± SEM. (<span class="html-italic">n</span> = 5–7) for each group. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 3
<p>Restoration of <span class="html-italic">Candida</span> and former <span class="html-italic">Candida</span> species burden to physiological levels in treated colitic mice. Quantitative RT-PCR for the detection of <span class="html-italic">Candida</span> spp. burden in stool samples using a custom TaqMan probe and the ITS1-2 primers to amplify fungal rDNA. Differences between the cycle threshold (Ct) of <span class="html-italic">Candida</span> spp. and ITS1-2 were calculated (ΔCts), and data are shown as 2<sup>−ΔCts</sup>. Values are expressed as mean ± SEM in panel B (<span class="html-italic">n</span> = 4 per group). * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 4
<p>Diversity analyses showing the peculiar fungal composition of non-colitic, control, and treated mice. (<b>A</b>) Boxplots of alpha diversity indexes (Shannon, Observed Features and Simpson) of fecal mycobiota evaluated for data rarefied at 4000 reads. Statistical significances were determined using the Wilcoxon multiple-comparison test with FDR adjustment. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 for non-colitic vs Crtl. group. (<b>B</b>) Boxplots of the beta diversity distances (Jaccard and Bray–Curtis) of fecal mycobiota evaluated for data rarefied at 4000 reads. Statistical significances were determined using the Wilcoxon multiple-comparison test with FDR adjustment. The asterisks (*) indicate the significance of differences for the Crtl or treated group versus the non-colitic group, and the hashtags (<sup>#</sup>) indicate the significance of differences for the non-colitic or treated versus the Crtl group. <sup>#,</sup>* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, ***<sup>,###</sup> <span class="html-italic">p</span> &lt; 0.001. (<b>C</b>) PCoA of Jaccard and Bray–Curtis distances. The first two PCoA components are represented in a scatter plot and in the marginal boxplots. The percentage of variance explained by each component is shown on each axis. Confidence ellipses assume a multivariate t-distribution. Asterisks (*) indicate the significance of differences for the control or treated versus non-colitic groups, and hashtags (#) indicate the significance of differences for the non-colitic or treated versus Crtl groups. <sup>#,</sup>* <span class="html-italic">p</span> &lt; 0.05, **<sup>,##</sup> <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 4 Cont.
<p>Diversity analyses showing the peculiar fungal composition of non-colitic, control, and treated mice. (<b>A</b>) Boxplots of alpha diversity indexes (Shannon, Observed Features and Simpson) of fecal mycobiota evaluated for data rarefied at 4000 reads. Statistical significances were determined using the Wilcoxon multiple-comparison test with FDR adjustment. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 for non-colitic vs Crtl. group. (<b>B</b>) Boxplots of the beta diversity distances (Jaccard and Bray–Curtis) of fecal mycobiota evaluated for data rarefied at 4000 reads. Statistical significances were determined using the Wilcoxon multiple-comparison test with FDR adjustment. The asterisks (*) indicate the significance of differences for the Crtl or treated group versus the non-colitic group, and the hashtags (<sup>#</sup>) indicate the significance of differences for the non-colitic or treated versus the Crtl group. <sup>#,</sup>* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, ***<sup>,###</sup> <span class="html-italic">p</span> &lt; 0.001. (<b>C</b>) PCoA of Jaccard and Bray–Curtis distances. The first two PCoA components are represented in a scatter plot and in the marginal boxplots. The percentage of variance explained by each component is shown on each axis. Confidence ellipses assume a multivariate t-distribution. Asterisks (*) indicate the significance of differences for the control or treated versus non-colitic groups, and hashtags (#) indicate the significance of differences for the non-colitic or treated versus Crtl groups. <sup>#,</sup>* <span class="html-italic">p</span> &lt; 0.05, **<sup>,##</sup> <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 5
<p>Mycobiome composition (phylum and genus levels) and mycobiome core (genus levels) analyses. (<b>A</b>) Heatmap depicting the abundances of phyla based on the sequencing data according to groups of interest. Features are sorted from top to bottom by the decreasing value in the average abundance calculated over the entire sequencing. (<b>B</b>) Heatmap depicting the abundances of genera present based on the sequencing data according to groups of interest. Features are sorted from top to bottom by decreasing value of the average abundance calculated over the entire sequencing. (<b>C</b>) Heatmaps of the mycobiome core of the genera in each sample group, as indicated in each heatmap, versus the per-sample relative abundance threshold (minimum detection threshold = 1%). The core of each group is composed of genera with a relative abundance of at least 1% and with prevalence greater than 50% in the group itself.</p>
Full article ">Figure 5 Cont.
<p>Mycobiome composition (phylum and genus levels) and mycobiome core (genus levels) analyses. (<b>A</b>) Heatmap depicting the abundances of phyla based on the sequencing data according to groups of interest. Features are sorted from top to bottom by the decreasing value in the average abundance calculated over the entire sequencing. (<b>B</b>) Heatmap depicting the abundances of genera present based on the sequencing data according to groups of interest. Features are sorted from top to bottom by decreasing value of the average abundance calculated over the entire sequencing. (<b>C</b>) Heatmaps of the mycobiome core of the genera in each sample group, as indicated in each heatmap, versus the per-sample relative abundance threshold (minimum detection threshold = 1%). The core of each group is composed of genera with a relative abundance of at least 1% and with prevalence greater than 50% in the group itself.</p>
Full article ">Figure 6
<p>Differential association analysis of genera in each treated or non-colitic group in comparison with the control group. (<b>A</b>–<b>D</b>) Cladograms and histograms showing taxonomies, up to the genus level, that were significantly (<span class="html-italic">p</span> &lt; 0.05) associated with one of the two groups considered, as indicated in the corresponding color legend. The yellow circles in the cladogram indicate non-significant taxa. The black arrows in the histograms emphasize the genera belonging to the mycobiome core of the group itself, from among the genera significantly associated with each group.</p>
Full article ">Figure 6 Cont.
<p>Differential association analysis of genera in each treated or non-colitic group in comparison with the control group. (<b>A</b>–<b>D</b>) Cladograms and histograms showing taxonomies, up to the genus level, that were significantly (<span class="html-italic">p</span> &lt; 0.05) associated with one of the two groups considered, as indicated in the corresponding color legend. The yellow circles in the cladogram indicate non-significant taxa. The black arrows in the histograms emphasize the genera belonging to the mycobiome core of the group itself, from among the genera significantly associated with each group.</p>
Full article ">
36 pages, 3132 KiB  
Review
The Ambivalence of Post COVID-19 Vaccination Responses in Humans
by Radha Gopalaswamy, Vivekanandhan Aravindhan and Selvakumar Subbian
Biomolecules 2024, 14(10), 1320; https://doi.org/10.3390/biom14101320 - 17 Oct 2024
Cited by 1 | Viewed by 2617
Abstract
The Coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), has prompted a massive global vaccination campaign, leading to the rapid development and deployment of several vaccines. Various COVID-19 vaccines are under different phases of clinical trials and include [...] Read more.
The Coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), has prompted a massive global vaccination campaign, leading to the rapid development and deployment of several vaccines. Various COVID-19 vaccines are under different phases of clinical trials and include the whole virus or its parts like DNA, mRNA, or protein subunits administered directly or through vectors. Beginning in 2020, a few mRNA (Pfizer-BioNTech BNT162b2 and Moderna mRNA-1273) and adenovirus-based (AstraZeneca ChAdOx1-S and the Janssen Ad26.COV2.S) vaccines were recommended by WHO for emergency use before the completion of the phase 3 and 4 trials. These vaccines were mostly administered in two or three doses at a defined frequency between the two doses. While these vaccines, mainly based on viral nucleic acids or protein conferred protection against the progression of SARS-CoV-2 infection into severe COVID-19, and prevented death due to the disease, their use has also been accompanied by a plethora of side effects. Common side effects include localized reactions such as pain at the injection site, as well as systemic reactions like fever, fatigue, and headache. These symptoms are generally mild to moderate and resolve within a few days. However, rare but more serious side effects have been reported, including allergic reactions such as anaphylaxis and, in some cases, myocarditis or pericarditis, particularly in younger males. Ongoing surveillance and research efforts continue to refine the understanding of these adverse effects, providing critical insights into the risk-benefit profile of COVID-19 vaccines. Nonetheless, the overall safety profile supports the continued use of these vaccines in combating the pandemic, with regulatory agencies and health organizations emphasizing the importance of vaccination in preventing COVID-19’s severe outcomes. In this review, we describe different types of COVID-19 vaccines and summarize various adverse effects due to autoimmune and inflammatory response(s) manifesting predominantly as cardiac, hematological, neurological, and psychological dysfunctions. The incidence, clinical presentation, risk factors, diagnosis, and management of different adverse effects and possible mechanisms contributing to these effects are discussed. The review highlights the potential ambivalence of human response post-COVID-19 vaccination and necessitates the need to mitigate the adverse side effects. Full article
Show Figures

Figure 1

Figure 1
<p>Summary of host response to COVID-19 vaccines. The COVID-19 vaccination-induced host responses can be broadly divided into immediate or delayed hypersensitivity. While the former response elicits allergic reactions and anaphylaxis, the latter response results in mild, moderate, or severe adverse events. The immediate hypersensitivity response is caused either by a classical, IgE-mediated activation of mast cells and basophils or an alternative non-classical pathway involving IgG and other antibodies activating neutrophils and basophils. Autoimmunity due to COVID-19 vaccination can be caused by molecular mimicry, bystander activation of immune cells, viral epitope spreading, or adjuvant-mediated immune response. The overall magnitude and durability of immune response as well as adverse effects mediated by COVID-19 vaccination are determined by several factors, including the age, sex, genetic makeup, immune status, and underlying health conditions of the host as well as the nature of the vaccine used. Image created in Biorender.</p>
Full article ">Figure 2
<p>Effects of COVID-19 vaccination-induced immunity. Following vaccination, the immune response against COVID-19 is mediated mainly by the development of Abs against SARS-CoV-2 proteins. The magnitude of immune response developed and its impact on the host protection is determined by the nature of Ab response elicited. An effective neutralizing Ab response neutralizes the virus, controls the infecting viral load and protects the vaccinated host against severe disease and/or death due to infection. However, a sub-optimal non-neutralizing Ab response leads to poor neutralization of the virus and ineffective control of viral load in the organs and may also contribute to Ab-mediated adverse effects (AE), which may enhance the disease manifestations. Image created in Biorender.</p>
Full article ">Figure 3
<p>Key pathways of COVID-19 vaccine-induced adverse immune reactions. The COVID-19 vaccine is comprised of the SARS-CoV-2 S protein (either as mRNA or protein) combined with an adjuvant such as polyethylene glycol (PEG). In the classical pathway, internalization of the viral and adjuvant-derived antigens (Ag) in the vaccine by antigen-presenting cells (APC) results in the presentation of antigenic epitopes to the T helper (Th) cells, which produces cytokines and activates Ag-specific B cells to produce various antibodies, such as IgG, IgE, IgM, etc. The Ag-specific IgE Abs binds to the FcεR1 and activates basophils and mast cells to produce histamine, which leads to allergy and/or anaphylaxis reactions. In the non-classical pathway, the antigens were taken up directly by the MRGPRX2 receptor on mast cells, which results in the induction of histamine and allergic responses. In addition, the immune complex formation by the Ag-specific and/or anti-idiotypic IgG, IgE, IgM Abs activates the C3a and C5a complement components, which ultimately results in complement activation-related pseudo-allergic reaction (CARPA). Finally, in the alternative/additional pathway, the antigen–IgG complex is taken up by neutrophils through FcγRs, which activates these polymorphonuclear cells to produce reactive oxygen species (ROS), proteases such as neutrophil-elastases (NE), Protease-3 (PR3), cathepsin G (CatG), and the formation of neutrophil extracellular traps (NETosis). The combined action of these pathways may contribute to the overall allergy and anaphylactic response due to COVID-19 vaccination. Image created in Biorender.</p>
Full article ">Figure 4
<p>Various mechanisms of adverse immune activation by COVID-19 vaccines. The viral S protein, either as mRNA or recombinant, adenovector-DNA, is endocytosed through Toll-like receptors (TLR) present on antigen-presenting cells (APCs). These endosomes trigger intracellular signaling pathways that result in the activation of Interferon regulatory factor-7 (IRF-7) and nuclear factor k B (NFkB) networks. Activated IRF7 and NFkB upregulate the production of proinflammatory cytokines IL-6 and TNFα. Alternatively, the viral components can escape from the endosome and trigger the cGAS signaling pathway, which activates STING/IRF3 network that ultimately results in the upregulation of proinflammatory type I interferons (IFN) response. Finally, the viral nucleic acids are translated into peptides and presented by the APC to activate T cells through the T cell receptor (TcR). Activation of naïve T cells results in the production of cytokines. Exposure to IL-4 skews the naïve T cells to an anti-inflammatory, Th2-type T cells that produce IL-3, IL-5, and IL-9, all of which can activate mast cells to elicit an allergic/anaphylactic reaction. In contrast, exposure to IL-12 and IFNγ polarizes the naïve T cells into Th1-type cells, which contributes to the proinflammatory response. Apart from the viral-derived molecules, vaccine adjuvants, such as CpG, can be recognized by TLR on the APC, with further activation of the NFkB pathway, leading to the production of inflammatory response. The viral nucleic acids also form a complex with platelet factor-4 (PF4) produced by the blood platelets. This complex activates Ag-specific B cells to produce anti-DNA/PF4 complex IgG, which binds with the FCγRIIa receptor on the platelets and activates these cells to form aggregates, leading to vaccine-induced thrombotic thrombocytopenia (VITT). Thus, both APCs and platelets play divergent roles in mounting immune dysregulation upon exposure to viral antigens and/or adjuvants. Image created in Biorender.</p>
Full article ">
12 pages, 1430 KiB  
Review
N6-Methyladenosine Methyltransferase Component KIAA1429 Is a Potential Target of Cancer Therapy
by Junjun Huang, Jihua Guo and Rong Jia
Biomolecules 2024, 14(10), 1319; https://doi.org/10.3390/biom14101319 - 17 Oct 2024
Viewed by 1147
Abstract
N6-methyladenosine (m6A), the most abundant RNA modification in eukaryotes, has a crucial impact on tumorigenesis. KIAA1429 is the key component of the m6A methyltransferase complex, in which KIAA1429 functions as a scaffold to bridge the catalytic core proteins. KIAA1429 [...] Read more.
N6-methyladenosine (m6A), the most abundant RNA modification in eukaryotes, has a crucial impact on tumorigenesis. KIAA1429 is the key component of the m6A methyltransferase complex, in which KIAA1429 functions as a scaffold to bridge the catalytic core proteins. KIAA1429 is often overexpressed in malignances, associated with patient prognosis, and required for tumorigenesis. KIAA1429 regulates the expression of a number of tumor-associated genes in an m6A -dependent manner, and thus, contributes to cell proliferation, migration, drug resistance, tumor formation and metastasis. This review focuses on recent progress in the understanding of roles and mechanisms of KIAA1429 in cancers, and offers ideas for potential anti-cancer therapeutic methods by targeting KIAA1429. Full article
(This article belongs to the Section Molecular Medicine)
Show Figures

Figure 1

Figure 1
<p>Gene structure and isoforms of the <span class="html-italic">VIRMA</span> gene encoding the KIAA1429 protein. (<b>A</b>) The gene structure of the <span class="html-italic">VIRMA</span> gene. (<b>B</b>) The mRNA structures of the KIAA1429 long isoform (KIAA1429-L) and the KIAA1429 short isoform (KIAA1429-S). (<b>C</b>) The structures of proteins encoded by KIAA1429 isoforms.</p>
Full article ">Figure 2
<p>Roles of KIAA1429 in tumorigenesis.</p>
Full article ">Figure 3
<p>Molecular mechanisms of KIAA1429 in cancers. KIAA1429 destabilizes the mRNAs of some tumor-suppressive genes, including <span class="html-italic">GATA3</span>, <span class="html-italic">RND3</span>, <span class="html-italic">BTG2</span>, and <span class="html-italic">DAPK3</span>, but stabilizes the mRNAs of some oncogenic genes, such as <span class="html-italic">RAB27B</span>, <span class="html-italic">HAS2</span>, <span class="html-italic">FOXM1</span>, <span class="html-italic">SMC1A</span>, and <span class="html-italic">JUN</span>. In addition, KIAA1429 can stabilize the mRNAs of the <span class="html-italic">SLC7A11</span> gene, a ferroptosis inhibitor, and some enzymes for aerobic glycolysis, like <span class="html-italic">HK2</span> and <span class="html-italic">ENO1</span>. The down-arrow represents the downregulation of the protein level. The up-arrow represents the upregulation of the protein level.</p>
Full article ">Figure 4
<p>The regulation of KIAA1429 expression.</p>
Full article ">
19 pages, 1565 KiB  
Review
The Role of Mitochondrial Permeability Transition in Bone Metabolism, Bone Healing, and Bone Diseases
by Xiting Zhu, Ziqi Qin, Min Zhou, Chen Li, Junjun Jing, Wushuang Ye and Xueqi Gan
Biomolecules 2024, 14(10), 1318; https://doi.org/10.3390/biom14101318 - 17 Oct 2024
Viewed by 1589
Abstract
Bone is a dynamic organ with an active metabolism and high sensitivity to mitochondrial dysfunction. The mitochondrial permeability transition pore (mPTP) is a low-selectivity channel situated in the inner mitochondrial membrane (IMM), permitting the exchange of molecules of up to 1.5 kDa in [...] Read more.
Bone is a dynamic organ with an active metabolism and high sensitivity to mitochondrial dysfunction. The mitochondrial permeability transition pore (mPTP) is a low-selectivity channel situated in the inner mitochondrial membrane (IMM), permitting the exchange of molecules of up to 1.5 kDa in and out of the IMM. Recent studies have highlighted the critical role of the mPTP in bone tissue, but there is currently a lack of reviews concerning this topic. This review discusses the structure and function of the mPTP and its impact on bone-related cells and bone-related pathological states. The mPTP activity is reduced during the osteogenic differentiation of mesenchymal stem cells (MSCs), while its desensitisation may underlie the mechanism of enhanced resistance to apoptosis in neoplastic osteoblastic cells. mPTP over-opening triggers mitochondrial swelling, regulated cell death, and inflammatory response. In particular, mPTP over-opening is involved in dexamethasone-induced osteoblast dysfunction and bisphosphonate-induced osteoclast apoptosis. In vivo, the mPTP plays a significant role in maintaining bone homeostasis, with many bone disorders linked to its excessive opening. Genetic deletion or pharmacological inhibition of the over-opening of mPTP has shown potential in enhancing bone injury recovery and alleviating bone diseases. Here, we review the findings on the relationship of the mPTP and bone at both the cellular and disease levels, highlighting novel avenues for pharmacological approaches targeting mitochondrial function to promote bone healing and manage bone-related disorders. Full article
(This article belongs to the Section Cellular Biochemistry)
Show Figures

Figure 1

Figure 1
<p>mPTP structure and the consequences of mPTP opening. The mitochondrial permeability transition pore (mPTP) has different conductance states. In its low-conductance state, the mPTP, potentially formed by the adenine nucleotide translocator (ANT), allows for the passage of ions and small metabolites. Ca<sup>2+</sup> efflux in this state limits the Ca<sup>2+</sup>-dependent tricarboxylic acid (TCA) cycle. In its high-conductance state, the mPTP is formed by a rearrangement of the F<sub>1</sub>F<sub>o</sub> ATP synthase complex. This state has more detrimental effects on cellular function. The release of mitochondrial DNA (mtDNA) through the mPTP triggers inflammatory responses. Extensive water influx causes mitochondrial swelling and subsequently induces outer membrane permeabilisation and the release of pro-apoptotic cofactors, leading to apoptosis or necrosis and TCA cycle collapse. ↓(orange downward arrow) represents downregulation of the TCA cycle.</p>
Full article ">Figure 2
<p>Diverse roles of mPTP opening in bone-related cells. The mitochondrial permeability transition pore (mPTP) is crucial in various functions within bone-related cells. In bone marrow-derived mesenchymal stem cells (BMSCs), cyclophilin D (CypD) downregulation and subsequent mPTP activity decline are required for osteogenic differentiation. In osteoblasts, mPTP over-opening is involved in several drug-induced regulated cell deaths (RCDs) in an ROS-dependent or ROS-independent manner. The mechanism by which osteosarcoma cells are resistant to apoptosis is, at least in part, due to desensitisation to the mPTP. Several studies have indicated that the mPTP is also involved in osteoclast apoptosis. ↑(black upward arrow) and ↓(black downward arrow) represents upregulation and downregulation of mPTP opening, respectively.</p>
Full article ">
16 pages, 2015 KiB  
Review
The Roles of Mitochondria in Human Being’s Life and Aging
by Hiroko P. Indo, Moragot Chatatikun, Ikuo Nakanishi, Ken-ichiro Matsumoto, Motoki Imai, Fumitaka Kawakami, Makoto Kubo, Hiroshi Abe, Hiroshi Ichikawa, Yoshikazu Yonei, Hisashi J. Beppu, Yukiko Minamiyama, Takuro Kanekura, Takafumi Ichikawa, Atthaphong Phongphithakchai, Lunla Udomwech, Suriyan Sukati, Nurdina Charong, Voravuth Somsak, Jitbanjong Tangpong, Sachiyo Nomura and Hideyuki J. Majimaadd Show full author list remove Hide full author list
Biomolecules 2024, 14(10), 1317; https://doi.org/10.3390/biom14101317 - 17 Oct 2024
Viewed by 2314
Abstract
The universe began 13.8 billion years ago, and Earth was born 4.6 billion years ago. Early traces of life were found as soon as 4.1 billion years ago; then, ~200,000 years ago, the human being was born. The evolution of life on earth [...] Read more.
The universe began 13.8 billion years ago, and Earth was born 4.6 billion years ago. Early traces of life were found as soon as 4.1 billion years ago; then, ~200,000 years ago, the human being was born. The evolution of life on earth was to become individual rather than cellular life. The birth of mitochondria made this possible to be the individual life. Since then, individuals have had a limited time of life. It was 1.4 billion years ago that a bacterial cell began living inside an archaeal host cell, a form of endosymbiosis that is the development of eukaryotic cells, which contain a nucleus and other membrane-bound compartments. The bacterium started to provide its host cell with additional energy, and the interaction eventually resulted in a eukaryotic cell, with both archaeal (the host cell) and bacterial (mitochondrial) origins still having genomes. The cells survived high concentrations of oxygen producing more energy inside the cell. Further, the roles of mitochondria in human being’s life and aging will be discussed. Full article
(This article belongs to the Special Issue Mitochondrial Quality Control in Aging and Neurodegeneration)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Schema of the electron transport chain. ATP production by oxidative phosphorylation with an electron transport chain (ETC).</p>
Full article ">Figure 2
<p>A scheme of mitochondrial generation of ROS. O<sub>2</sub><b><sup>•</sup></b><sup>−</sup> generated from mitochondria binds with NO and further produces peroxynitrous acid (ONOOH), and further ONOOH produces NO<sub>2</sub><b><sup>•</sup></b> and <b><sup>•</sup></b>OH. In the figure red arrow of up means up-regulations, and stable means no expression change. (Figure transferred from Reference [<a href="#B1-biomolecules-14-01317" class="html-bibr">1</a>]).</p>
Full article ">Figure 3
<p>Illustration of the 4.6-billion-year-old Earth, acknowledging “Andrée Valley, University of Wisconsin—Madison”. (Reference [<a href="#B6-biomolecules-14-01317" class="html-bibr">6</a>]). The author, H.J.M. added mitochondria to the figure.</p>
Full article ">Figure 4
<p>Increase of atmospheric oxygen in the history of Earth through 4.6 billion years. The figure was transferred from Reference [<a href="#B11-biomolecules-14-01317" class="html-bibr">11</a>] with a license.</p>
Full article ">Figure 5
<p>Conclusion: relationship between aging and mitochondria. Upon aging, arrow stable means no change, arrow down means down regulation, and arrow up means up regulation.</p>
Full article ">
13 pages, 1968 KiB  
Article
Gintonin Stimulates Glucose Uptake in Myocytes: Involvement of Calcium and Extracellular Signal-Regulated Kinase Signaling
by Rami Lee, Kyung-Jong Won, Ji-Hun Kim, Byung-Hwan Lee, Sung-Hee Hwang and Seung-Yeol Nah
Biomolecules 2024, 14(10), 1316; https://doi.org/10.3390/biom14101316 - 17 Oct 2024
Viewed by 1009
Abstract
Ginseng has anti-hyperglycemic effects. Gintonin, a glycolipoprotein derived from ginseng, also stimulates insulin release from pancreatic beta cells. However, the role of gintonin in glucose metabolism within skeletal muscle is unknown. Here, we showed the effect of gintonin on glucose uptake, glycogen content, [...] Read more.
Ginseng has anti-hyperglycemic effects. Gintonin, a glycolipoprotein derived from ginseng, also stimulates insulin release from pancreatic beta cells. However, the role of gintonin in glucose metabolism within skeletal muscle is unknown. Here, we showed the effect of gintonin on glucose uptake, glycogen content, glucose transporter (GLUT) 4 expression, and adenosine triphosphate (ATP) content in C2C12 myotubes. Gintonin (3–30 μg/mL) dose-dependently stimulated glucose uptake in myotubes. The expression of GLUT4 on the cell membrane was increased by gintonin treatment. Treatment with 1–3 μg/mL of gintonin increased glycogen content in myotubes, but the content was decreased at 30 μg/mL of gintonin. The ATP content in myotubes increased following treatment with 10–100 μg/mL gintonin. Gintonin transiently elevated intracellular calcium concentrations and increased the phosphorylation of extracellular signal-regulated kinase (ERK). Gintonin-induced transient calcium increases were inhibited by treatment with the lysophosphatidic acid receptor inhibitor Ki16425, the phospholipase C inhibitor U73122, and the inositol 1,4,5-trisphosphate receptor antagonist 2-aminoethoxydiphenyl borate. Gintonin-stimulated glucose uptake was decreased by treatment with U73122, the intracellular calcium chelator 1,2-bis(o-aminophenoxy)ethane-N,N,N′,N′-tetraacetic acid tetra(acetoxymethyl) ester, and the ERK inhibitor PD98059. These results show that gintonin plays a role in glucose metabolism by increasing glucose uptake through transient calcium increases and ERK signaling pathways. Thus, gintonin may be beneficial for glucose metabolism control. Full article
(This article belongs to the Special Issue Therapeutic Potential of Natural Products in Metabolic Diseases)
Show Figures

Figure 1

Figure 1
<p>Effect of gintonin on the cell viability of myotubes and transient intracellular calcium increases. (<b>A</b>) Cell viability. C2C12 myotubes were treated with gintonin (GT, 0.1–100 μg/mL) or lysophosphatidic acid (10 μM) for 24 h. Then, WST assay was performed. All data are shown as the mean ± SEM (<span class="html-italic">n</span> = 6). (<b>B</b>,<b>C</b>) Transient intracellular calcium increases. Fura-2-AM-incorporated C2C12 myotubes were treated with gintonin (GT, 0.3–30 μg/mL), and intracellular calcium levels were measured by spectrofluorophotometry and calculated. Each arrow in panel (<b>B</b>) represents time points of treatment with gintonin at indicated concentrations. The horizontal length of the upper scale bar corresponds to 100 s (100 s). (<b>D</b>) Inhibitory effects of inhibitors on GT-induced intracellular calcium increase. Fura-2-AM-loaded C2C12 myotubes were pretreated with PTX (100 ng/mL), Ki16425 (10 μM), U73122 (5 μM), or 2-APB (100 μM) for 5 min and then treated with gintonin (GT, 1 μg/mL). All data are shown as the mean ± SEM (<span class="html-italic">n</span> = 3–5); * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001 vs. untreated control cells (Con). PTX, pertussis toxin; 2-APB, the inositol 1,4,5-trisphosphate receptor antagonist 2-aminoethoxydiphenyl borate.</p>
Full article ">Figure 2
<p>Effect of gintonin on glucose uptake in C2C12 myotubes. (<b>A</b>) C2C12 myotubes were treated with gintonin (GT, 3 μg/mL) and 2-NBDG (100 μM) for 0–24 h. (<b>B</b>) The myotubes were treated with gintonin (0.1–30 μg/mL) for 24 h. (<b>C</b>–<b>F</b>) The myotubes were treated with gintonin (GT, 3 μg/mL) and 2-NBDG (100 μM) for 24 h, with or without the addition of inhibitors (Ki16425, 10 μM; U73122, 5 μM; BAPTA-AM, 50 μM; PD98059, 10 μM). Then, 2-NBDG uptake was measured using spectrofluorophotometry. All data are shown as the mean ± SEM. (<span class="html-italic">n</span> = 6); * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001 vs. time 0 or untreated control cells (Con). <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05; <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01; <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 vs. GT alone. BAPTA-AM, 1,2-bis(o-aminophenoxy)ethane-N,N,N′,N′-tetraacetic acid tetra(acetoxymethyl) ester.</p>
Full article ">Figure 3
<p>Effect of gintonin on ATP and glycogen content in C2C12 myotubes. (<b>A</b>,<b>B</b>) ATP content. (<b>A</b>) C2C12 myotubes were treated with gintonin (GT, 10 μg/mL) for 0–24 h. (<b>B</b>) The myotubes were treated with gintonin (0.1–100 μg/mL) or lysophosphatidic acid (LPA, 10 μM) for 8 h. The ATP content of myotubes was measured using an ATP assay kit. (<b>C</b>) Glycogen content. The myotubes were treated with gintonin (GT, 0.1–30 μg/mL) or insulin (INS, 100 nM), and the glycogen content was measured using a glycogen assay kit. All data are presented as the mean ± SEM (<span class="html-italic">n</span> = 6); * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001 vs. time 0 or untreated control cells (Con).</p>
Full article ">Figure 4
<p>Effect of gintonin on ERK phosphorylation in C2C12 myotubes. (<b>A</b>) C2C12 myotubes were treated with gintonin (GT, 10 μg/mL) for 0–60 min. (<b>B</b>) The myotubes were treated with gintonin (0.3–100 μg/mL) for 10 min. (<b>C</b>) The myotubes were pretreated with inhibitors (Ki16425, 10 μM; PD98059, 10 μM; U73122, 5 μM) for 1 h and then treated with gintonin (3 μg/mL) for 10 min. Phosphorylated ERK and ERK were detected by immunoblotting. All data are shown as the mean ± SEM (n = 4); * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001 vs. time 0 or untreated control cells (Con). <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05; <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01; <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 vs. GT alone. p-ERK, phospho-ERK. Original western blot images can be found in <a href="#app1-biomolecules-14-01316" class="html-app">Supplementary File S1</a>.</p>
Full article ">Figure 5
<p>Effect of gintonin on GLUT4 expression in total lysates and plasma membrane fractions of C2C12 myotubes. (<b>A</b>) GLUT4 expression in total lysates. (<b>B</b>) GLUT4 expression in the plasma membrane fraction. C2C12 myotubes were treated with gintonin (GT, 10 μg/mL) for 120 min or insulin (INS, 100 nM) for 30 min. GLUT4 expression in total lysates and plasma membrane fraction of C2C12 myotubes was detected by immunoblotting. β-actin and Na<sup>+</sup>/K<sup>+</sup> ATPase were also detected as loading controls. All data are shown as the mean ± SEM (<span class="html-italic">n</span> = 4); ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001 vs. untreated control cells (Con). Original western blot images can be found in <a href="#app1-biomolecules-14-01316" class="html-app">Supplementary File S1</a>.</p>
Full article ">Figure 6
<p>Possible signaling pathways of gintonin (GT)-induced glucose uptake in C2C12 myotubes. Gintonin induces transient increases in intracellular calcium concentrations and ERK activation via LPA receptor (LPAR) activation. These may lead to increases in the expression and translocation of GLUT4, subsequently increasing glucose uptake. PLC, phospholipase C; ERK, extracellular signal-regulated kinase; GLUT4, glucose transporter type 4.</p>
Full article ">
14 pages, 2418 KiB  
Review
Fungal L-Methionine Biosynthesis Pathway Enzymes and Their Applications in Various Scientific and Commercial Fields
by Kamila Rząd, Aleksandra Kuplińska and Iwona Gabriel
Biomolecules 2024, 14(10), 1315; https://doi.org/10.3390/biom14101315 - 17 Oct 2024
Viewed by 1336
Abstract
L-methionine (L-Met) is one of the nine proteinogenic amino acids essential for humans since, in human cells, there are no complete pathways for its biosynthesis from simple precursors. L-Met plays a crucial role in cellular function as it is required for proper protein [...] Read more.
L-methionine (L-Met) is one of the nine proteinogenic amino acids essential for humans since, in human cells, there are no complete pathways for its biosynthesis from simple precursors. L-Met plays a crucial role in cellular function as it is required for proper protein synthesis, acting as an initiator. Additionally, this amino acid participates in various metabolic processes and serves as a precursor for the synthesis of S-adenosylmethionine (AdoMet), which is involved in the methylation of DNA molecules and phospholipids, as well as in maintaining genome stability. Due to its importance, fungal L-methionine biosynthesis pathway enzymes are being intensively studied. This review presents the current state of the art in terms of their cellular function, usefulness as molecular markers, antifungal targets, or industrial approaches. Full article
(This article belongs to the Section Enzymology)
Show Figures

Figure 1

Figure 1
<p>The biosynthesis pathway of L-Met in <span class="html-italic">S. cerevisiae</span> cells. Met2p <span class="html-italic">O</span>-acetyl-L-homoserine <span class="html-italic">O</span>-acetyltransferase (EC 2.3.1.31); Met17p bifunctional <span class="html-italic">O</span>-acetyl-L-homoserine/<span class="html-italic">O</span>-acetyl-L-serine sulfhydrylase (EC 2.5.1.49, EC 2.5.1.47); Str2p cystathionine γ-synthase (EC 2.5.1.48); Str3p cystathionine β-lyase (EC 4.4.1.8); Cys4p cystathionine β-synthase (EC 4.2.1.22); Cys3p cystathionine γ-lyase (EC 4.4.1.1); Met6p methionine synthase (EC 2.1.1.13).</p>
Full article ">Figure 2
<p>The reaction catalyzed by L-homoserine <span class="html-italic">O</span>-acetyltransferase (EC 2.3.1.31).</p>
Full article ">Figure 3
<p>Structures of L-homoserine and its analog L-penicillamine, an inhibitor of <span class="html-italic">C. albicans</span> Met2p.</p>
Full article ">Figure 4
<p>The reaction catalyzed by the bi-functional <span class="html-italic">O</span>-acetyl-L-homoserine/<span class="html-italic">O</span>-acetyl-L-serine sulfhydrylase enzyme (EC 2.5.1.49, EC 2.5.1.47) from <span class="html-italic">S. cerevisiae</span>. ‘Ac’ denotes acetate.</p>
Full article ">Figure 5
<p>The reaction catalyzed by cystathionine-γ-synthase (EC 2.5.1.48). ‘Ac’ denotes acetate.</p>
Full article ">Figure 6
<p>The reaction catalyzed by cystathionine β-lyase (EC 4.4.1.8). ‘Pyr’ denotes pyruvate.</p>
Full article ">Figure 7
<p>The reaction catalyzed by cystathionine β-synthase (EC 4.2.1.22).</p>
Full article ">Figure 8
<p>Structures of EAOA and PA, inhibitors of Cy4p from <span class="html-italic">C. albicans</span> [<a href="#B45-biomolecules-14-01315" class="html-bibr">45</a>].</p>
Full article ">Figure 9
<p>The L-cystathionine conversion reaction catalyzed by cystathionine γ-lyase (EC4.4.1.1). ‘2-OB’ denotes 2-oxybutanoate.</p>
Full article ">Figure 10
<p>The reaction catalyzed by methionine synthase (EC 2.1.1.13). 5-MTHF and THF denote 5-methyltetrahydrofolate and tetrahydrofolate, respectively.</p>
Full article ">
12 pages, 2239 KiB  
Article
Some Glycoproteins Expressed on the Surface of Immune Cells and Cytokine Plasma Levels Can Be Used as Potential Biomarkers in Patients with Colorectal Cancer
by Tsvetelina Batsalova, Denitsa Uzunova, Gergana Chavdarova, Tatyana Apostolova and Balik Dzhambazov
Biomolecules 2024, 14(10), 1314; https://doi.org/10.3390/biom14101314 - 16 Oct 2024
Viewed by 1124
Abstract
Colorectal cancer (CRC) is a leading cause of mortality worldwide. Its incidence holds a major position among the most common life-threatening diseases. Hence, the early identification and precise characterization of disease activity based on proper biomarkers are of utmost importance for therapeutic strategy [...] Read more.
Colorectal cancer (CRC) is a leading cause of mortality worldwide. Its incidence holds a major position among the most common life-threatening diseases. Hence, the early identification and precise characterization of disease activity based on proper biomarkers are of utmost importance for therapeutic strategy and patient survival. The identification of new biomarkers for colorectal cancer or disease-specific levels/combinations of biomarkers will significantly contribute to precise diagnosis and improved personalized treatment of patients. Therefore, the present study aims to identify colorectal cancer-specific immunological biomarkers. The plasma levels of several cytokines (interleukin-1β /IL-1β/, IL-2, IL-4, IL-10, IL-12, IL-15, TGFβ and IFNγ) of 20 patients with colorectal cancer and 21 healthy individuals were determined by ELISA. The expression of several types of glycoproteins on the surface of peripheral blood leukocytes isolated from CRC patients and healthy volunteers was evaluated by flow cytometry. Correlations between cytokine levels and cell surface glycoprotein expression were analyzed. The obtained results demonstrated significantly elevated levels of CD80, CD86, CD279 and CD274 expressing leukocyte populations in the cancer patient group, while the numbers of NK cells and CD8- and CD25-positive cells were decreased. Based on these data and the correlations with cytokine levels, it can be concluded that CD25, CD80, CD86, CD274 and CD279 glycoproteins combined with specific plasma levels of IL-1β, IL-2, IL-15 and TGFβ could represent potential biomarkers for colorectal cancer. Full article
(This article belongs to the Special Issue Immune-Related Biomarkers: 2nd Edition)
Show Figures

Figure 1

Figure 1
<p>Plasma concentrations of inflammatory cytokines in stage IV CRC patients and control healthy volunteers. Levels of IL-1β (<b>A</b>), IL-2 (<b>B</b>), IL-4 (<b>C</b>), IL-10 (<b>D</b>), IL-12 (<b>E</b>), IL-13 (<b>F</b>), IL-15 (<b>G</b>), TGFβ (<b>H</b>) and IFNγ (<b>I</b>). Data represent ± standard error of the mean (±SEM). A Mann–Whitney <span class="html-italic">U</span> test was used for statistical analyses. ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 2
<p>Flow cytometry analyses of the expression of T-cell-specific markers on peripheral blood leukocytes derived from stage IV CRC patients and healthy individuals. CD4<sup>+</sup> T cells (<b>A</b>), CD8<sup>+</sup> T cells (<b>B</b>), CD25<sup>+</sup> T cells (<b>C</b>), CD28<sup>+</sup> T cells (<b>D</b>), CD152<sup>+</sup> T cells (<b>E</b>), and CD279<sup>+</sup> T cells (<b>F</b>). The results are presented as ±SEM. Statistical significance was defined by the Mann–Whitney <span class="html-italic">U</span> test. * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 3
<p>Levels of CD20<sup>+</sup> B cells, NK cells and expression of some B7 family molecules in the peripheral blood of stage IV CRC patients and healthy controls. Percentage of CD20<sup>+</sup> B cells (<b>A</b>), CD80<sup>+</sup> B cells (<b>B</b>), CD86<sup>+</sup> B cells (<b>C</b>), CD273<sup>+</sup> lymphocytes (<b>D</b>), CD274<sup>+</sup> lymphocytes (<b>E</b>), and CD56<sup>+</sup>/CD16<sup>+</sup> NK cells (<b>F</b>). Data are shown as ±SEM. Statistical analyses were performed using the Mann–Whitney <span class="html-italic">U</span> test; ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 4
<p>Correlations between IL-2 and IL-15 plasma concentrations with other cytokines. First column: IL-2 vs. IL-1β (<b>A</b>), IL-2 vs. TGFβ (<b>C</b>), IL-2 vs. IFNγ (<b>E</b>), IL-2 vs. IL-15 (<b>G</b>), IL-2 vs. IL-12 (<b>I</b>); Second column: IL-15 vs. IL-12 (<b>B</b>), IL-15 vs. IL-1β (<b>D</b>), IL-15 vs. IL-13 (<b>F</b>), IL-15 vs. TGFβ (<b>H</b>), and IL-2 vs. IL-12 only for CRC patients (<b>J</b>). The charts indicate Spearman’s rank correlation coefficient (ρ). * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 5
<p>Correlations between circulating cytokines and immunomodulatory molecule expression levels determined for the stage IV CRC patient group. IL-15 vs. CD279<sup>+</sup> cells (<b>A</b>), IL-1β vs. CD86<sup>+</sup> cells (<b>B</b>) and CD8<sup>+</sup> vs. CD279<sup>+</sup> cells (<b>C</b>). The graphs show Spearman’s rank correlation coefficient (ρ). * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">
16 pages, 685 KiB  
Article
Altered Gut Microbiota Patterns in Young Children with Recent Maltreatment Exposure
by Gergana Karaboycheva, Melanie L. Conrad, Peggy Dörr, Katja Dittrich, Elena Murray, Karolina Skonieczna-Żydecka, Mariusz Kaczmarczyk, Igor Łoniewski, Heiko Klawitter, Claudia Buss, Sonja Entringer, Elisabeth Binder, Sibylle M. Winter and Christine Heim
Biomolecules 2024, 14(10), 1313; https://doi.org/10.3390/biom14101313 - 16 Oct 2024
Viewed by 1247
Abstract
Background: The brain and the intestinal microbiota are highly interconnected and especially vulnerable to disruptions in early life. Emerging evidence indicates that psychosocial adversity detrimentally impacts the intestinal microbiota, affecting both physical and mental health. This study aims to investigate the gut microbiome [...] Read more.
Background: The brain and the intestinal microbiota are highly interconnected and especially vulnerable to disruptions in early life. Emerging evidence indicates that psychosocial adversity detrimentally impacts the intestinal microbiota, affecting both physical and mental health. This study aims to investigate the gut microbiome in young children in the immediate aftermath of maltreatment exposure. Methods: Maltreatment exposure was assessed in 88 children (ages 3–7) using the Maternal Interview for the Classification of Maltreatment [MICM]. Children were allocated to three groups according to the number of experienced maltreatment categories: no maltreatment, low maltreatment, and high maltreatment exposures. Stool samples were collected and analyzed by 16S rRNA sequencing. Results: Children subjected to high maltreatment exposure exhibited lower alpha diversity in comparison to those with both no and low maltreatment exposure (Simpson Index, Tukey post hoc, p = 0.059 and p = 0.007, respectively). No significant distinctions in beta diversity were identified. High maltreatment exposure was associated with the enrichment of several genera from the class Clostridia (Clostridium, Intestinibacter, Howardella and Butyrivibrio) and the depletion of the genus Phocaeicola (class Bacteriodia). Conclusions: Severe maltreatment exposure is associated with alterations in the gut microbiota of young children. Longitudinal trajectories of intestinal microbiota composition in the context of maltreatment may reveal important insights related to psychiatric and somatic health outcomes. Full article
Show Figures

Figure 1

Figure 1
<p>Alpha diversity in the no-, low- and high-maltreatment-exposure groups, according to the Simpson Index. The high-maltreatment-exposure group shows a significantly lower alpha diversity compared to the no- and low-maltreatment-exposure groups. ANCOVA with post hoc Tukey analysis. Significance is represented by ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 2
<p>Volcano plots representing bacterial differential abundance between (<b>A</b>) the low- and the no-maltreatment-exposure group, and (<b>B</b>) the high and the no-maltreatment-exposure group. The horizontal line marks the FDR adjusted <span class="html-italic">p</span> = 0.05 cut off. The vertical lines mark a log fold change of 1 and −1, respectively. Positive log fold change values indicate taxa that are relatively more abundant in the multitype maltreatment group while negative log fold change values taxa, which are relatively depleted. We have labeled only genera with significant differential abundance.</p>
Full article ">
16 pages, 2591 KiB  
Article
Short Link N Modulates Inflammasome Activity in Intervertebral Discs Through Interaction with CD14
by Muskan Alad, Michael P. Grant, Laura M. Epure, Sunny Y. Shih, Geraldine Merle, Hee-Jeong Im, John Antoniou and Fackson Mwale
Biomolecules 2024, 14(10), 1312; https://doi.org/10.3390/biom14101312 - 16 Oct 2024
Cited by 1 | Viewed by 1104
Abstract
Intervertebral disc degeneration and pain are associated with the nucleotide-binding domain, leucine-rich repeat, and pyrin domain-containing 3 (NLRP3) inflammasome activation and the processing of interleukin-1 beta (IL-1β). Activation of thehm inflammasome is triggered by Toll-like receptor stimulation and requires the cofactor receptor cluster [...] Read more.
Intervertebral disc degeneration and pain are associated with the nucleotide-binding domain, leucine-rich repeat, and pyrin domain-containing 3 (NLRP3) inflammasome activation and the processing of interleukin-1 beta (IL-1β). Activation of thehm inflammasome is triggered by Toll-like receptor stimulation and requires the cofactor receptor cluster of differentiation 14 (CD14). Short Link N (sLN), a peptide derived from link protein, has been shown to modulate inflammation and pain in discs in vitro and in vivo; however, the underlying mechanisms remain elusive. This study aims to assess whether sLN modulates IL-1β and inflammasome activity through interaction with CD14. Disc cells treated with lipopolysaccharides (LPS) with or without sLN were used to assess changes in Caspase-1, IL-1β, and phosphorylated nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB). Peptide docking of sLN to CD14 and immunoprecipitation were performed to determine their interaction. The results indicated that sLN inhibited LPS-induced NFκB and Caspase-1 activation, reducing IL-1β maturation and secretion in disc cells. A significant decrease in inflammasome markers was observed with sLN treatment. Immunoprecipitation studies revealed a direct interaction between sLN and the LPS-binding pocket of CD14. Our results suggest that sLN could be a potential therapeutic agent for discogenic pain by mitigating IL-1β and inflammasome activity within discs. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

Figure 1
<p>Immunohistochemistry of NLRP3 expression in a degenerative IVD model. Rabbit IVDs from a puncture model were given a single injection of either saline or sLN. Sham animals were used as controls. Twelve weeks following injection, IVDs were processed for immunohistochemistry and the detection of NLRP3. Higher-magnification inset images are included and demarcated to highlight specific areas of interest. Immunohistochemistry of NLRP3 in (<b>A</b>) sham and degenerative IVDS treated with (<b>B</b>) saline or (<b>C</b>) sLN. (<b>D</b>) Densitometry of NLRP3 expression in IVDs presented in (<b>A</b>). Statistical significance was assessed using Student’s <span class="html-italic">t</span>-test (comparison between saline and sLN); ***, <span class="html-italic">p</span> &lt; 0.001; <span class="html-italic">n</span> = 4. (IVD—intervertebral disc, NLRP3—nucleotide-binding domain, leucine-rich repeat, and pyrin domain-containing 3, yellow dotted line—endplate).</p>
Full article ">Figure 2
<p>Evaluation of NF-κB activation in human nucleus pulposus cells treated with sLN and LPS. (<b>A</b>) Human nucleus pulposus (NP) cells were treated with LPS alone or in combination with sLN with the indicated concentrations (0.05, 0.5, or 5.0 μg/mL) for 45 min. Western blotting was performed to detect activation of P-NF-κB. Blots were normalized to GAPDH for loading by densitometry and calculated as a fold increase over control. The densitometry of the blots is presented in the graph below. (<b>B</b>) HNP cells were incubated with LPS alone or (<b>C</b>) LPS and 0.5 μg/mL sLN for the indicated times (0–180 min). P-NF-κB signal was normalized to GAPDH and calculated as a fold-over control. Statistical significance was assessed using ANOVA and post hoc Dunnett’s test (comparison to control); ****, <span class="html-italic">p</span> &lt; 0.0001; <span class="html-italic">n</span> = 4. Original Western blot images are available in <a href="#app1-biomolecules-14-01312" class="html-app">Supplementary Materials</a>.</p>
Full article ">Figure 3
<p>Suppression of inflammasome markers by sLN. HNP cells were incubated for 48 h in medium supplemented with LPS or LPS and 0.5 μg/mL sLN. Control samples were incubated with medium alone. RNA expression is shown for (<b>A</b>) <span class="html-italic">NLRP3</span>, (<b>B</b>) <span class="html-italic">PYC</span>, (<b>C</b>) <span class="html-italic">CASP1</span>, (<b>D</b>) <span class="html-italic">IL1B</span>, and (<b>E</b>) <span class="html-italic">TNFA</span>. Treatments were compared to controls. ANOVA and post hoc Dunnett’s test; *, <span class="html-italic">p</span> &lt; 0.05; **, <span class="html-italic">p</span> &lt; 0.01; ***, <span class="html-italic">p</span> &lt; 0.001; ****, <span class="html-italic">p</span> &lt; 0.0001; <span class="html-italic">n</span> = 4.</p>
Full article ">Figure 4
<p>Assessment of Caspase-1 activation and IL-1β secretion in hNP cells. HNP cells were treated with LPS alone or in combination with sLN at varying concentrations (0.05, 0.5 and 5.0 μg/mL) for 48 h. (<b>A</b>) Western blotting and densitometry of Caspase-1 demonstrating Pro-Caspase-1, Caspase-1, and GAPDH as loading control. (<b>B</b>) Western blotting and densitometry of pro- and mature forms of IL-1β. Data are expressed as fold-over controls. Statistical significance was assessed using ANOVA and post hoc Dunnett’s test (treatments compared to LPS alone); **, <span class="html-italic">p</span> &lt; 0.01; ***, <span class="html-italic">p</span> &lt; 0.001; <span class="html-italic">n</span> = 4. Original Western blot images are available in <a href="#app1-biomolecules-14-01312" class="html-app">Supplementary Materials</a>.</p>
Full article ">Figure 5
<p>Inflammasome activation and hNP-induced macrophage polarization. RAW macrophages and hNP cells were co-cultured to determine the effects of hNP on macrophage polarization. HNP cells were stimulated with LPS and incubated for 48 hrs with RAW cells. Expression of M1 (<b>A</b>–<b>C</b>) and M2 (<b>D</b>–<b>F</b>) markers in RAW macrophages were measured by qPCR. Plots represent fold-over control. ANOVA and post hoc Dunnett’s test (treatments compared to control); *, <span class="html-italic">p</span> &lt; 0.05; **, <span class="html-italic">p</span> &lt; 0.01; ***, <span class="html-italic">p</span> &lt; 0.001; <span class="html-italic">n</span> = 4.</p>
Full article ">Figure 6
<p>Interaction and immunoprecipitation of sLN and CD14. (<b>A</b>) Schematic on the co-immunoprecipitation of CD14 and sLN and dot-blot showing enrichment of sLN following pull-down with CD14. (<b>B</b>) Schematic demonstrating the competitive co-immunoprecipitation of biotinylated LPS for CD14 in the presence of unlabeled LPS and sLN. Western blot showing the detection of CD14 following the indicated pull-downs. Lanes from left to right: control (CD14 only), LPS-treated, sLN-treated. Original Western blot images are available in <a href="#app1-biomolecules-14-01312" class="html-app">Supplementary Materials</a>.</p>
Full article ">Figure 7
<p>Mechanism of sLN inhibition of inflammasome activation in disc cells. This schematic illustrates the process by which sLN inhibits inflammasome activation in nucleus pulposus cells, detailing the interactions and molecular pathways involved.</p>
Full article ">
17 pages, 3440 KiB  
Article
Caution for Multidrug Therapy: Significant Baroreflex Afferent Neuroexcitation Coordinated by Multi-Channels/Pumps Under the Threshold Concentration of Yoda1 and Dobutamine Combination
by Yin-zhi Xu, Zhao-yuan Xu, Hui-xiao Fu, Mao Yue, Jia-qun Li, Chang-peng Cui, Di Wu and Bai-yan Li
Biomolecules 2024, 14(10), 1311; https://doi.org/10.3390/biom14101311 - 16 Oct 2024
Viewed by 796
Abstract
Multi-drug therapies are common in cardiovascular disease intervention; however, io channel/pump coordination has not been tested electrophysiologically. Apparently, inward currents were not elicited by Yoda1/10 nM or Dobutamine/100 nM alone in Ah-type baroreceptor neurons, but were by their combination. To verify this, electroneurography [...] Read more.
Multi-drug therapies are common in cardiovascular disease intervention; however, io channel/pump coordination has not been tested electrophysiologically. Apparently, inward currents were not elicited by Yoda1/10 nM or Dobutamine/100 nM alone in Ah-type baroreceptor neurons, but were by their combination. To verify this, electroneurography and the whole-cell patch-clamp technique were performed. The results showed that Ah- and C-volley were dramatically increased by the combination at 0.5 V and 5 V, in contrast to A-volley, as consistent with repetitive discharge elicited by step and ramp with markedly reduced current injection/stimulus intensity. Notably, a frequency-dependent action potential (AP) duration was increased with Iberiotoxin-sensitive K+ component. Furthermore, an increased peak in AP measured in phase plots suggested enhanced Na+ influx, cytoplasmic Ca2+ accumulation through reverse mode of Na+/Ca2+ exchanger, and, consequently, functional KCa1.1 up-regulation. Strikingly, the Yoda1- or Dbtm-mediated small/transient Na+/K+-pump currents were robustly increased by their combination, implying a quick ion equilibration that may also be synchronized by hyperpolarization-induced voltage-sag, enabling faster repetitive firing. These novel findings demonstrate multi-channel/pump collaboration together to integrate neurotransmission at the cellular level for baroreflex, providing an afferent explanation in sexual dimorphic blood pressure regulation, and raising the caution regarding the individual drug concentration in multi-drug therapies to optimize efficacy and minimize toxicity. Full article
(This article belongs to the Section Molecular Medicine)
Show Figures

Figure 1

Figure 1
<p>Inward currents recorded in the identified Ah-type baroreceptor neurons using gap-free protocol under voltage-clamp mode before and after Yoda1 (10, 30, 100 nM), Dobutamine (Dbtm, 100, 300, 1000 nM), and the combination. The recording was held at −60 mV for 120 s. (<b>A</b>,<b>D</b>,<b>G</b>): Concentration-dependent effects of Yoda1 on inward currents. (<b>B</b>,<b>E</b>,<b>H</b>): Concentration-dependent effects of Dbtm on inward currents. (<b>C</b>,<b>F</b>,<b>I</b>): Concentration-dependent effect of Yoda1/Dbtm combination on inward currents. The black dot dash line: time to application; red dot dash line: the peak time of Yoda1; green dot dash line: peak time of the combination.</p>
Full article ">Figure 2
<p>Changes in compound action potential (AP) recorded from aortic depressor nerve (ADN) in intact adult female SD rats in the presence of Yoda1 or Yoda1 alone with Dobutamine (Dbtm). Compound AP was elicited by bi-polar electrode using a series of voltage ranged from 0.1 to 20 V and averaged root mean square (RMS, μV) was calculated. A-volley, Ah-volley, and C-volley represented the composition of all A-, Ah-, and C-type baroreceptor afferents, respectively, and were identified according to the afferent fiber conduction velocity (time from stimulation to the waveform/length of ADN, m/s). (<b>A</b>): Representative recording with 2.0 V stimulation. (<b>B</b>–<b>D</b>): Representative A-volley (&gt;10 m/s), Ah-volley (2–10 m/s), and C-volley (&lt;2 m/s) between each paired vertical dash dot line; downward arrowhead shown in (<b>B</b>) means the stimulus artifact. (<b>E</b>–<b>G</b>): Averaged (Avg.) RMS for (<b>B</b>–<b>D</b>). Unpaired <span class="html-italic">t</span>-test was used between groups, data were presented as mean ± SD, and <span class="html-italic">n</span> = 4 and 6 for Yoda1 (10 nM) and Yoda1 + Dbtm (100 nM); * <span class="html-italic">p</span> &lt; 0.05 and <sup>#</sup> <span class="html-italic">p</span> &lt; 0.01 vs. Yoda1 at the same time point.</p>
Full article ">Figure 3
<p>Changes in discharge capability of action potential (AP) recorded from identified Ah-type baroreceptor neurons (BRNs) isolated from adult female SD rats in the presence of Yoda1 or Yoda1 + Dobutamine (Dbtm). AP was elicited by stepped current injection under voltage-clamp mode of whole-cell patch configuration. (<b>A1</b>,<b>A2</b>): Representative recordings were obtained from two different Ah-type BRNs (Cell #1 @ the top two rows and Cell #2 @ the bottom two rows with three steps for each treatment) in the presence of Yoda1 10 nM (as control) or Yoda1 + Dbtm 100 nM. (<b>B</b>): Summarized data of the number of AP elicited within each step before (black/step #1, blue/step #2, and yellow/step #3) and after treatment (red/step #1, light blue/step #2, and green/step #3). (<b>C</b>): Summarized data of the step current applied for step. Repetitive discharges were collected before and after Dbtm and paired <span class="html-italic">t</span>-test was applied. Averaged data were expressed as mean ± SD, <span class="html-italic">n</span> = 7. ** <span class="html-italic">p</span> &lt; 0.01 vs. Yoda1. Scaled bars also applied for other step recordings of the same cell.</p>
Full article ">Figure 4
<p>Changes in the ramp current applied for evoking similar AP discharge in identified Ah-type BRNs in the presence of Yoda1 or Yoda1 + Dbtm. To determine the excitability under similar AP discharge by ramp protocol, the ramp current was quantified in identified Ah-type BRNs. (<b>A</b>): Representative recordings in the presence of Yoda1 (10 nM, black) and Yoda1 + Dbtm (100 nM, red). (<b>B</b>): Summarized data for the resting membrane potential (RMP). (<b>C</b>): Summarized ramp current applied. (<b>D</b>) Summarized APFT. Unpaired <span class="html-italic">t</span>-test was selected between groups and averaged data were expressed as mean ± SD, <span class="html-italic">n</span> = 20 for Yoda1, and <span class="html-italic">n</span> = 24 for Yoda1 + Dbtm. ** <span class="html-italic">p</span> &lt; 0.01 vs. Yoda1.</p>
Full article ">Figure 5
<p>Alternative changes in action potential duration (APD<sub>50</sub>) of identified Ah-type BRNs in the presence of Yoda1 or Yoda1 + Dbtm. The first (1st) and the last APs in the spike trains of repetitive discharges in the presence of Yoda1 and Yoda1 + Dbtm shown were superimposed; resting membrane potential (RMP), APD<sub>50</sub>, and the peak of AP were measured accordingly: (<b>A</b>): the 1st APs, (<b>B</b>): the last APs. Averaged data were expressed as mean ± SD, <span class="html-italic">n</span> = 6 complete recordings. * <span class="html-italic">p</span> &lt; 0.05 or ** <span class="html-italic">p</span> &lt; 0.01 vs. Yoda1. The horizontal bar in the (<b>B</b>) was also applied for (<b>A</b>).</p>
Full article ">Figure 6
<p>Iberiotoxin/KCa1.1 inactivation abolishes AP widening induced by Yoda1 + Dbtm in identified Ah-type BRNs. Repetitive discharge of AP was elicited by step depolarization in the presence of Yoda1 and Yoda1 + Dbtm without/with Iberiotoxin, and complete recordings in one cell were included for further analysis. (<b>A</b>): The last APs in the spike trains were superimposed. (<b>B</b>): Phase plots: the total membrane current plotted as a function of membrane voltage from each AP shown in (<b>A</b>), and α, β, χ, δ, and ε were represented for the AP firing threshold, the maximal up-stroke velocity of total inward current/depolarization phase (negative portion), the peak of AP, the maximal down-stroke velocity of total outward current/repolarization phase (positive portion), and the peak of hyperpolarization, respectively; blue arrowhead means the location of the repolarization humps: <span class="html-italic">Inset/left and inset/right</span>: changes in total inward and outward. Averaged data were presented as mean ± SD, <span class="html-italic">n</span> = 6 complete recordings from at least four preparations, ** <span class="html-italic">p</span> &lt; 0.01 vs. Yoda1, <sup>#</sup> <span class="html-italic">p</span> &lt; 0.01 vs. Yoda1 + Dbtm.</p>
Full article ">Figure 7
<p>Effects of Iberiotoxin (IbTx) on total K<sup>+</sup> currents in the presence of Yoda1 + Dbtm recorded from identified Ah-BRNs. After AP was recorded for afferent fiber type identification, the extracellular solution was changed with bath perfusion to the one for potassium current recording; the cell was clamped at a holding potential of −80 mV and stepped from −70 mV up to +40 mV with 5 mV increment with an interval of 1 s between sweeps. Potassium currents were recorded in the presence of Yoda1/10 nM, Dbtm/100 nM, and Yoda1 + Dbtm, respectively. IbTx 100 nM was micropurfused to the tested cell to avoid contaminating other cells in the chamber after successfully recording the total K<sup>+</sup> currents, and IbTx-sensitive components were obtained by subtraction. Total current was divided by its whole-cell capacitance and current density was presented as pA/pF. (<b>A</b>): Representative recording of Ah-type BRN identified by waveform characters, the vertical dash dot line means the presence of repolarization hump. (<b>B</b>,<b>C</b>): Representative tracings of total and IbTx-sensitive K<sup>+</sup> currents. Scale bars also applied for (<b>B</b>). (<b>D</b>): Summarized data for comparisons of total and IbTx-sensitive components. Averaged results were presented as mean ± SD, <span class="html-italic">n</span> = 19 recordings from at least nine preparations; ** <span class="html-italic">p</span> &lt; 0.01 vs. Yoda1.</p>
Full article ">Figure 8
<p>Changes in Na<sup>+</sup>-K<sup>+</sup>-ATPase currents recorded in identified Ah-type BRNs in the presence of Yoda1 or Yoda1 + Dbtm. Na<sup>+</sup>-K<sup>+</sup>-ATPase currents were recorded under physiological condition with intracellular concentration of 8.9 mM and extracellular concentration of 145 mM. The order of the recording was Yoda1 (10 nM, black, <span class="html-italic">n</span> = 6), Dbtm (100 nM, red, <span class="html-italic">n</span> = 6), Yoda1 + 30 nM Dbtm (green, <span class="html-italic">n</span> = 7), and Yoda1 + 100 nM Dbtm (blue, <span class="html-italic">n</span> = 7). Inset showing the summarized analysis. Unpaired <span class="html-italic">t</span>-test was applied between groups, and one-way ANOVA was also tested among groups with post-hoc Tukey test. Averaged data were expressed as mean ± SD; * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 vs. Yoda1; <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 vs. Dbtm.</p>
Full article ">Figure 9
<p>Schematic outline of up-regulated baroreflex afferent neuroexcitation through coordination of multi-ion channel and pump activation over the course of action potential by the combination use of Yoda1 and Dobutamine under threshold concentration.</p>
Full article ">
Previous Issue
Next Issue
Back to TopTop