[go: up one dir, main page]

Next Issue
Volume 12, August
Previous Issue
Volume 12, June
 
 

Antioxidants, Volume 12, Issue 7 (July 2023) – 167 articles

Cover Story (view full-size image): Drug-induced liver injury (DILI) is one of the major causes of acute liver failure and drug attrition in preclinical and clinical phases of drug development. Idiosyncratic DILI (iDILI) occurs only in a low percentage of people exposed to a drug and usually cannot be predicted in preclinical studies since it is dose-independent and relies on different patient-specific factors. Cell-based models provide a relatively inexpensive and fast system that can offer valuable toxicological information in the early stages of drug development, although one of the most important gaps is iDILI detection. In order to assess the role of immune cells in iDILI, we developed a direct co-culture of HepG2 cells and activated macrophages to decipher the mechanisms induced by model iDILI drugs. 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, 1183 KiB  
Review
Astaxanthin as a Potent Antioxidant for Promoting Bone Health: An Up-to-Date Review
by Iswari Davan, Sharida Fakurazi, Ekram Alias, Nurul ‘Izzah Ibrahim, Ng Min Hwei and Haniza Hassan
Antioxidants 2023, 12(7), 1480; https://doi.org/10.3390/antiox12071480 - 24 Jul 2023
Cited by 3 | Viewed by 4427
Abstract
In recent years, bone loss and its associated diseases have become a significant public health concern due to increased disability, morbidity, and mortality. Oxidative stress and bone loss are correlated, where oxidative stress suppresses osteoblast activity, resulting in compromised homeostasis between bone formation [...] Read more.
In recent years, bone loss and its associated diseases have become a significant public health concern due to increased disability, morbidity, and mortality. Oxidative stress and bone loss are correlated, where oxidative stress suppresses osteoblast activity, resulting in compromised homeostasis between bone formation and resorption. This event causes upregulation of bone remodeling turnover rate with an increased risk of fractures and bone loss. Therefore, supplementation of antioxidants can be proposed to reduce oxidative stress, facilitate the bone remodeling process, suppress the initiation of bone diseases, and improve bone health. Astaxanthin (3,3′-dihydroxy-4-4′-diketo-β-β carotene), a potent antioxidant belonging to the xanthophylls family, is a potential ROS scavenger and could be a promising therapeutic nutraceutical possessing various pharmacological properties. In bone, astaxanthin enhances osteoblast differentiation, osteocytes numbers, and/or differentiation, inhibits osteoclast differentiation, cartilage degradation markers, and increases bone mineral density, expression of osteogenic markers, while reducing bone loss. In this review, we presented the up-to-date findings of the potential anabolic effects of astaxanthin on bone health in vitro, animal, and human studies by providing comprehensive evidence for its future clinical application, especially in treating bone diseases. Full article
(This article belongs to the Special Issue Women’s Special Issue Series: Antioxidants in Human Health)
Show Figures

Figure 1

Figure 1
<p>Bone formation by osteoblast and bone resorption by osteoclasts. Mesenchymal cells differentiate into osteoblasts transforming into bone lining cells and osteocytes embedded in mineralized bone matrix. Osteoblast also undergoes apoptosis, which is induced by oxidative stress. Receptor activator of NF-ΚB ligand (RANKL) on osteoblast binds to RANK expressed by osteoclasts, activating osteoclast cells, facilitating osteoclastogenesis by secreting hydrogen ions and lysosome enzyme (Cathepsin K) into the microenvironment under the ruffled border forming resorption pit. Figure generated by biorender.com.</p>
Full article ">Figure 2
<p>Molecular structure of astaxanthin. Figure generated by biorender.com.</p>
Full article ">Figure 3
<p>Schematic diagram showing the pathway of nuclear factor kappa beta (NF-Κβ), mitogen-activated protein kinase (MAPK), and c-Jun N-terminal kinases (JNK). This activates aggrecanase and collagenase, leading to ECM degradation and promoting osteoarthritis. Figure generated by biorender.com.</p>
Full article ">
24 pages, 1343 KiB  
Review
Urolithin A in Health and Diseases: Prospects for Parkinson’s Disease Management
by Olga Wojciechowska and Małgorzata Kujawska
Antioxidants 2023, 12(7), 1479; https://doi.org/10.3390/antiox12071479 - 24 Jul 2023
Cited by 3 | Viewed by 4752
Abstract
Parkinson’s disease (PD) is a chronic and progressive neurodegenerative disorder characterized by a complex pathophysiology and a range of symptoms. The prevalence increases with age, putting the ageing population at risk. Disease management includes the improvement of symptoms, the comfort of the patient’s [...] Read more.
Parkinson’s disease (PD) is a chronic and progressive neurodegenerative disorder characterized by a complex pathophysiology and a range of symptoms. The prevalence increases with age, putting the ageing population at risk. Disease management includes the improvement of symptoms, the comfort of the patient’s life, and palliative care. As there is currently no cure, growing evidence points towards the beneficial role of polyphenols on neurodegeneration. Numerous studies indicate the health benefits of the family of urolithins, especially urolithin A (UA). UA is a bacterial metabolite produced by dietary ellagitannins and ellagic acid. An expanding body of literature explores the involvement of the compound in mitochondrial health, and its anti-inflammatory, anti-oxidant, and anti-apoptotic properties. The review organizes the existing knowledge on the role of UA in health and diseases, emphasizing neurodegenerative diseases, especially PD. We gathered data on the potential neuroprotective effect in in vivo and in vitro models. We discussed the possible mechanisms of action of the compound and related health benefits to give a broader perspective of potential applications of UA in neuroprotective strategies. Moreover, we projected the future directions of applying UA in PD management. Full article
(This article belongs to the Special Issue Natural Antioxidants and Their Sources in Food)
Show Figures

Figure 1

Figure 1
<p>Catabolic pathway of ellagitannins and ellagic acid to urolithins. 5-OH, 4-OH, 3-OH, 2-OH, and 1-OH refer to the number of hydroxyl groups for each urolithin group—penta-, tetra-, tri-, di- and monohydroxy urolithins, respectively. The blue font refers to new urolithins generated by a bacterial 3-dehydroxylase. The purple and red circles designate the final urolithins produced in UM-A and UM-B, respectively. Uro-AR can be found in both metabotypes. Adapted from [<a href="#B7-antioxidants-12-01479" class="html-bibr">7</a>].</p>
Full article ">Figure 2
<p>Urolithin A mechanisms of action. Ellagitannins and ellagic acid are polyphenols that occur naturally in dietary products like pomegranates, berries, and nuts. The compounds are substrates of colonic bacteria transformed into urolithins. However, it is estimated that only 40% of individuals could naturally convert the polyphenolic precursors to UA. Thus, UA administration is proposed as an answer for urolithin non-producers. In vivo and in vitro experiments suggest the health-promoting activity of UA. The effects of the compound are related to different mechanisms of action, including engagement in mitochondrial function and the process of mitophagy, inflammation, oxidative stress, and the modulation of the apoptosis process. Created with BioRender.com.</p>
Full article ">Figure 3
<p>Urolithin A’s activity in Parkinson’s disease model studies. UA’s beneficial activity may include potential favorable effects of UA on brain health. The current research explores the potential neuroprotective effects of the compound in PD models. The beneficial role of the compound can be related to the reduction in neuroinflammation, loss of dopaminergic neurons, a-synuclein aggregation, and apoptosis, as well as improved mitochondrial, motor, and cognitive function. Created with BioRender.com.</p>
Full article ">
17 pages, 3707 KiB  
Article
Teneligliptin Co-Infusion Alleviates Morphine Tolerance by Inhibition of Spinal Microglial Cell Activation in Streptozotocin-Induced Diabetic Rats
by Yaswanth Kuthati, Vaikar Navakanth Rao, Wei-Hsiu Huang, Prabhakar Busa and Chih-Shung Wong
Antioxidants 2023, 12(7), 1478; https://doi.org/10.3390/antiox12071478 - 24 Jul 2023
Cited by 2 | Viewed by 1407
Abstract
Morphine (MOR) is a commonly prescribed drug for the treatment of moderate to severe diabetic neuropathic pain (DNP). However, long-term MOR treatment is limited by morphine analgesic tolerance (MAT). The activation of microglial cells and the release of glia-derived proinflammatory cytokines are known [...] Read more.
Morphine (MOR) is a commonly prescribed drug for the treatment of moderate to severe diabetic neuropathic pain (DNP). However, long-term MOR treatment is limited by morphine analgesic tolerance (MAT). The activation of microglial cells and the release of glia-derived proinflammatory cytokines are known to play an important role in the development of MAT. In this study, we aimed to investigate the effects of the dipeptidyl peptidase-4 inhibitor (DPP-4i) teneligliptin (TEN) on MOR-induced microglial cell activation and MAT in DNP rats. DNP was induced in four groups of male Wistar rats through a single intraperitoneal injection of streptozotocin (STZ) (50 mg/kg, freshly dissolved in 5 mmol/L citrate buffer, pH 4.5). Sham rats were administered with the vehicle. Seven days after STZ injection, all rats were implanted with an intrathecal (i.t) catheter connected to a mini-osmotic pump, divided into five groups, and infused with the following combinations: sham + saline (1 µL/h, i.t), DNP + saline (1 µL/h, i.t), DNP + MOR (15 µg/h, i.t), DNP + TEN (2 µg/h, i.t), and DNP + MOR (15 µg/h, i.t) + TEN (2 µg/h, i.t) for 7 days at a rate of 1 μL/h. The MAT was confirmed through the measurement of mechanical paw withdrawal threshold and tail-flick tests. The mRNA expression of neuroprotective proteins nuclear factor erythroid 2-related factor (Nrf2) and heme oxygenase-1 (HO-1) in the dorsal horn was evaluated by quantitative PCR (qPCR). Microglial cell activation and mononucleate cell infiltration in the spinal cord dorsal horn were assessed by immunofluorescence assay (IFA) and Western blotting (WB). The results showed that co-infusion of TEN with MOR significantly attenuated MAT in DNP rats through the restoration of neuroprotective proteins Nrf2 and HO-1 and suppression of microglial cell activation in the dorsal horn. Though TEN at a dose of 2 μg has mild antinociceptive effects, it is highly effective in limiting MAT. Full article
(This article belongs to the Section Health Outcomes of Antioxidants and Oxidative Stress)
Show Figures

Figure 1

Figure 1
<p>Changes in (<b>a</b>) body weight, (<b>b</b>) glycemic levels, and (<b>c</b>) paw withdrawal sensitivity in sham vs. DNP rats. Red curves indicate the mean ± SD of diabetic rats, and blue curves correspond to the mean ± SD of non-diabetic rats. Asterisk denotes a statistically significant difference when comparing sham rats vs. STZ DNP 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">n</span> = 6 animals per group).</p>
Full article ">Figure 2
<p>A single intrathecal injection of MOR was given in diabetic rats and non-diabetic rats at different doses. In diabetic rats, the drug was administered after 7 days of STZ injection. (<b>a</b>) Paw withdrawal response to mechanical allodynia in diabetic rats and non-diabetic rats, (<b>b</b>) tail-flick latency in response to hot water immersion test in diabetic rats and non-diabetic rats. Asterisk denotes a statistically significant difference when comparing saline rats vs. diabetic rats, saline diabetic rats vs. diabetic 2 mg MOR, saline diabetic rats vs. diabetic 5 mg MOR, and saline diabetic rats vs. diabetic 10 mg g MOR and a statistically significant difference when comparing saline non-diabetic rats vs. non-diabetic 2 mg MOR, saline non-diabetic rats vs. non-diabetic 5 mg MOR, and saline non-diabetic rats vs. non-diabetic 10 mg MOR. * <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> = 6 animals per group).</p>
Full article ">Figure 3
<p>Changes in mechanical paw withdrawal thresholds of rats after installation of osmotic pump after establishment of DNP in (<b>a</b>) sham + saline rats, DNP + saline rats, and rats infused with DNP + TEN. (<b>b</b>) DNP + saline, DNP + MOR, and DNP + MOR + TEN. Before pump installation readings denote day 7 readings after STZ injection (<span class="html-italic">n</span> = 6). The asterisk in (<b>a</b>) denotes a statistically significant difference when comparing sham vs. DNP + saline and DNP + saline vs. DNP + TEN. Asterisk in (<b>b</b>) denotes a statistically significant difference when comparing DNP + saline vs. DNP + MOR and DNP + MOR vs. DNP + TEN + MOR. * <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> = 6 animals per group).</p>
Full article ">Figure 4
<p>Changes in blood glucose level of rats after installation of the osmotic pump in (<b>a</b>) sham + saline, DNP + saline, and DNP + TEN and (<b>b</b>) DNP + saline, DNP + MOR, and DNP + MOR + TEN. The asterisk in (<b>a</b>) denotes a statistically significant difference when comparing sham vs. DNP + saline and DNP + saline vs. DNP + TEN. Asterisk in Figure (<b>b</b>) denotes a statistically significant difference when comparing DNP + saline vs. DNP + MOR and DNP + MOR vs. DNP + TEN + MOR. ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001 (<span class="html-italic">n</span> = 6 animals per group).</p>
Full article ">Figure 5
<p>Changes in body weight level of rats after installation of the osmotic pump in (<b>a</b>) sham + saline, DNP + saline, and DNP + TEN and (<b>b</b>) DNP + saline, DNP + MOR, and DNP + MOR + TEN. The asterisk in (<b>a</b>) denotes a statistically significant difference when comparing sham + saline vs. DNP + saline and DNP + saline vs. DNP + TEN. Asterisk in Figure (<b>b</b>) denotes a statistically significant difference when comparing DNP + saline vs. DNP + MOR and DNP + MOR vs. DNP + TEN + MOR. ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001 (<span class="html-italic">n</span> = 6 animals per group).</p>
Full article ">Figure 6
<p>Relative mRNA expression levels of (<b>a</b>) Nrf2 and (<b>b</b>) HO-1 were determined by Q-PCR. The asterisk denotes a statistically significant difference when comparing sham + saline vs. DNP + saline and DNP + saline vs. DNP + TEN and DNP + saline vs. DNP + MOR. # denotes a statistically significant difference when comparing DNP + MOR vs. DNP + MOR + TEN. * <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> = 6 animals per group).</p>
Full article ">Figure 7
<p>The expression and activation of CD-11b. (<b>a</b>) Western blot and (<b>b</b>) the quantitative analysis of CD11b in sham and DNP rat models receiving i.t infusion with saline (1 µL/h), MOR (15 μg/h), TEN (2 μg/h), or MOR (15 μg/h) + TEN (2 μg/h) for 7 days. The asterisk denotes a statistically significant difference when comparing sham + saline vs. DNP + saline, DNP + saline vs. DNP + TEN, and DNP + saline vs. DNP + MOR. # denotes a statistically significant difference when comparing DNP + MOR vs. DNP + MOR + TEN. * <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> = 6 animals per group).</p>
Full article ">Figure 8
<p>Activated microglial cells using IBA-1 antibody in (<b>a</b>) sham + saline-, (<b>b</b>) DNP + saline-, (<b>c</b>) DNP + MOR-, (<b>d</b>) DNP + TEN-, and (<b>e</b>) DNP + MOR + TEN-infused rats. (<b>f</b>) The quantitative analysis of activated microglial cells. There was a significant increase in activated microglia after MOR infusion which was ameliorated by TEN co-infusion. All spinal cord samples were collected on day 7 after the osmotic pump infusion. The asterisk denotes a statistically significant difference when comparing sham vs. DNP + saline, DNP + saline vs. DNP + TEN, and DNP + saline vs. DNP + MOR. # denotes a statistically significant difference when comparing DNP + MOR vs. DNP + MOR + TEN. **/## <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001 (<span class="html-italic">n</span> = 6 animals per group). Scale bar = 20 µm.</p>
Full article ">Figure 9
<p>H&amp;E staining showing mononucleate cell infiltration, a marker of inflammatory response in (<b>a</b>) sham + saline-, (<b>b</b>) DNP + saline-, (<b>c</b>) DNP + MOR-, (<b>d</b>) DNP + TEN-, and (<b>e</b>) DNP + MOR + TEN-infused rats. (<b>f</b>) The quantitative analysis of infiltrates. MOR infusion aggravated mononuclear cell infiltration. TEN infusion in combination with MOR alleviated the inflammatory response. All spinal cord samples were collected on day 7 after the osmotic pump infusion. * denotes a statistically significant difference when comparing sham + saline vs. DNP + saline and DNP + saline vs. DNP + TEN. # denotes a statistically significant difference when comparing DNP + MOR vs. DNP + MOR + TEN and DNP + saline vs. DNP + MOR. **/## <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001 (<span class="html-italic">n</span> = 6 animals per group). Scale bar = 100 µm.</p>
Full article ">
11 pages, 1015 KiB  
Review
Complex II Biology in Aging, Health, and Disease
by Eric Goetzman, Zhenwei Gong, Bob Zhang and Radhika Muzumdar
Antioxidants 2023, 12(7), 1477; https://doi.org/10.3390/antiox12071477 - 24 Jul 2023
Cited by 9 | Viewed by 2449
Abstract
Aging is associated with a decline in mitochondrial function which may contribute to age-related diseases such as neurodegeneration, cancer, and cardiovascular diseases. Recently, mitochondrial Complex II has emerged as an important player in the aging process. Mitochondrial Complex II converts succinate to fumarate [...] Read more.
Aging is associated with a decline in mitochondrial function which may contribute to age-related diseases such as neurodegeneration, cancer, and cardiovascular diseases. Recently, mitochondrial Complex II has emerged as an important player in the aging process. Mitochondrial Complex II converts succinate to fumarate and plays an essential role in both the tricarboxylic acid (TCA) cycle and the electron transport chain (ETC). The dysfunction of Complex II not only limits mitochondrial energy production; it may also promote oxidative stress, contributing, over time, to cellular damage, aging, and disease. Intriguingly, succinate, the substrate for Complex II which accumulates during mitochondrial dysfunction, has been shown to have widespread effects as a signaling molecule. Here, we review recent advances related to understanding the function of Complex II, succinate signaling, and their combined roles in aging and aging-related diseases. Full article
(This article belongs to the Special Issue Mitochondrial Oxidative Stress in Aging and Disease)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Structure and functional role of mitochondrial Complex II. Complex II is composed of four nuclear-encoded subunits SDHA, SDHB, SDHC, and SDHD (labeled “A”, “B”, “C”, and “D” in the figure).Under normal physiological conditions, SDHA serves to oxidize succinate to fumarate, transporting electrons first to the FAD cofactor on SDHA (depicted in yellow rings), then to a series of three iron-sulfur clusters (Fe-S) embedded in SDHB, and ending with transfer to ubiquinone (Q) at the interface of SDHC/D and the inner mitochondrial membrane (IMM). Malonate and oxaloacetate are naturally occurring inhibitors that compete with succinate for binding to the active site. A series of fungicides are known Complex II inhibitors via the ubiquinone binding site. Other abbreviations: OMM, outer mitochondrial membrane; IMS, inter-membrane space; III, respiratory chain Complex III; IV, respiratory chain Complex IV; V, respiratory chain Complex V (ATP synthase); CytC, cytochrome C; TCA, tricarboxylic acid cycle. Figure created with <a href="http://BioRender.com" target="_blank">BioRender.com</a> (accessed on 9 June 2023).</p>
Full article ">Figure 2
<p>Complex II produces ROS under three contexts. (1) When succinate is at normal physiological concentrations, the binding site is often left unoccupied while the FAD cofactor is in the reduced state (electrons indicated as pink circles). This allows oxygenated solvent access to steal electrons from the FAD cofactor. (2) Inborn errors of Complex II genes, Complex II assembly factors, or acquired mutations in cancer can disrupt the functional integrity of the complex, promoting electron leak from either the FAD or from the iron-sulfur (Fe-S) clusters. (3) Reverse electron transport can be a robust source of ROS under conditions of high succinate concentrations. Here, succinate saturates the SDHA biding site, preventing oxygen from stealing electrons, but the ubiquinone system cannot keep up and becomes highly reduced (QH2), leading to electrons flowing upstream to Complex I, where they react with oxygen to form ROS. Other abbreviations: IMM, inner mitochondrial membrane; III, respiratory chain Complex III; A,B,C,D, succinate dehydrogenase subunits SDHA, SDHB, SDHC, and SDHD. Figure created with <a href="http://BioRender.com" target="_blank">BioRender.com</a> (accessed on 9 June 2023).</p>
Full article ">
19 pages, 5469 KiB  
Article
Effects of Tannic Acid Supplementation on the Intestinal Health, Immunity, and Antioxidant Function of Broilers Challenged with Necrotic Enteritis
by Huiping Xu, Xiaodan Zhang, Peng Li, Yimeng Luo, Jianyang Fu, Lu Gong, Zengpeng Lv and Yuming Guo
Antioxidants 2023, 12(7), 1476; https://doi.org/10.3390/antiox12071476 - 24 Jul 2023
Cited by 5 | Viewed by 2200
Abstract
Clostridium perfringens causes necrotic enteritis (NE) after proliferation in the intestine of poultry, resulting in considerable losses to the poultry industry. This study aimed to investigate the impact of tannic acid on the antioxidant, immunity, and gut health of broilers with NE. In [...] Read more.
Clostridium perfringens causes necrotic enteritis (NE) after proliferation in the intestine of poultry, resulting in considerable losses to the poultry industry. This study aimed to investigate the impact of tannic acid on the antioxidant, immunity, and gut health of broilers with NE. In the experiment, 630 one-day-old Cobb500 male chicks were randomly divided into six treatment groups, with seven replicate cages and with fifteen birds in each cage. The treatment groups were as follows: control group (NC), challenged group (PC), and challenged NE chickens treated with 250, 500, 750, and 1000 mg/kg tannic acid (PTA1, PTA2, PTA3, and PTA4, respectively). To induce NE, coccidia vaccine and Clostridium perfringens were administered on day 19 and days 22–28, respectively. Indexes related to antioxidant, immune, and intestinal health were measured on days 28 and 35. During the infection period, we observed significant increases in fecal water content, D-LA, TNF-α, and malondialdehyde concentrations (p < 0.05). Conversely, significant decreases were noted in chyme pH and in T-AOC, IL-4, and IL-10 concentrations (p < 0.05). The addition of tannic acid exhibited a linear decrease in fecal water content and TNF-α concentration (p < 0.05). Furthermore, tannic acid supplementation resulted in a quadratic curve decrease in D-LA concentration and linear increases in T-AOC, IL-4, and IL-10 (p < 0.05). Cecal microbiological analysis revealed that Ruminococcaceae and Butyricimona were dominant in PTA3. In conclusion, the dietary addition of tannic acid may reduce the negative effects of NE by increasing antioxidant and anti-inflammatory capacity, improving the intestinal barrier, and regulating the intestinal flora. Full article
(This article belongs to the Special Issue Natural Antioxidants in Animal Immunity—2nd Edition)
Show Figures

Figure 1

Figure 1
<p>Flowchart of the animal experiment. The type of <span class="html-italic">C. perfringens</span> in this test was chicken-derived <span class="html-italic">C. perfringens</span> type A CVCC52.</p>
Full article ">Figure 2
<p>Rationalization of cecum microbial sequencing data volume and number of OTUs in the NC, PC, and PTA3 groups on d 28: (<b>A</b>) shows the rarefaction curve of cecal microorganisms, (<b>B</b>) shows the rank abundance of cecal microorganisms, and (<b>C</b>) shows the OTUs specific to cecal microorganisms in the NC, PC, and PTA3 groups. NC: unchallenged and untreated; PC: challenged with NE and untreated; PTA3: challenged with NE and treated with a control diet supplemented with 750 mg/kg tannic acid. <span class="html-italic">n =</span> 7.</p>
Full article ">Figure 3
<p>Rationalization of cecal microbial sequencing data volume and number of OTUs in the NC, PC, and PTA3 groups on d 35: (<b>A</b>) shows the rarefaction curve of cecal microorganisms, (<b>B</b>) shows the rank abundance of cecal microorganisms, and (<b>C</b>) shows the OTUs specific to cecal microorganisms in the NC, PC, and PTA3 groups. NC: unchallenged and untreated; PC: challenged with NE and untreated; PTA3: challenged with NE and treated with a control diet supplemented with 750 mg/kg tannic acid. <span class="html-italic">n =</span> 7.</p>
Full article ">Figure 4
<p>Effects of tannic acid addition on cecal microorganisms in broiler chickens with NE on d 28: (<b>A</b>) shows the effects of tannic acid on cecal microbial α-diversity; (<b>B</b>) shows the PCoA plot; (<b>C</b>) shows the relative abundance of the top fifteen microorganisms at the class level; (<b>D</b>) shows the relative abundance of the top fifteen microorganisms at the genus level; (<b>E</b>) is the result of LEfSe analysis for differential microorganisms; and (<b>F</b>) shows the Cladogram plots of the differential microorganisms. NC: unchallenged and untreated; PC: challenged with NE and untreated; PTA3: challenged with NE and treated with a control diet supplemented with 750 mg/kg tannic acid. <span class="html-italic">n =</span> 7. ** indicates <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 5
<p>Effects of tannic acid addition on the cecal microorganisms in broiler chickens with NE on d 35: (<b>A</b>) shows the effects of tannic acid on cecal microbial α-diversity; (<b>B</b>) shows the PCoA plot; (<b>C</b>) shows the relative abundance of the top fifteen microorganisms at the class level; (<b>D</b>) shows the relative abundance of the top fifteen microorganisms at the genus level; (<b>E</b>) shows the result of the LEfSe analysis of the differential microorganisms; and (<b>F</b>) shows the Cladogram plots of the differential microorganisms. NC: unchallenged and untreated; PC: challenged with NE and untreated; PTA3: challenged with NE and treated with a control diet supplemented with 750 mg/kg tannic acid. <span class="html-italic">n =</span> 7.</p>
Full article ">Figure 6
<p>Correlation analysis of antioxidance, immunity, intestinal barrier, and chyme pH with differential bacteria: (<b>A</b>) shows the results of the correlation analysis on d 28 and (<b>B</b>) shows the results of the correlation analysis on d 35. Probability values are indicated as follows: * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">
19 pages, 3297 KiB  
Article
Ultrasound-Assisted Deep Eutectic Solvent Extraction of Phenolic Compounds from Thinned Young Kiwifruits and Their Beneficial Effects
by Ding-Tao Wu, Wen Deng, Jie Li, Jin-Lei Geng, Yi-Chen Hu, Liang Zou, Yi Liu, Hong-Yan Liu and Ren-You Gan
Antioxidants 2023, 12(7), 1475; https://doi.org/10.3390/antiox12071475 - 23 Jul 2023
Cited by 5 | Viewed by 1627
Abstract
Fruit thinning is a common practice employed to enhance the quality and yield of kiwifruits during the growing period, and about 30–50% of unripe kiwifruits will be thinned and discarded. In fact, these unripe kiwifruits are rich in nutrients and bioactive compounds. Nevertheless, [...] Read more.
Fruit thinning is a common practice employed to enhance the quality and yield of kiwifruits during the growing period, and about 30–50% of unripe kiwifruits will be thinned and discarded. In fact, these unripe kiwifruits are rich in nutrients and bioactive compounds. Nevertheless, the applications of thinned young kiwifruits and related bioactive compounds in the food and functional food industry are still limited. Therefore, to promote the potential applications of thinned young kiwifruits as value-added health products, the extraction, characterization, and evaluation of beneficial effects of phenolic compounds from thinned young fruits of red-fleshed Actinidia chinensis cv ‘HY’ were examined in the present study. A green and efficient ultrasound-assisted deep eutectic solvent extraction (UADE) method for extracting phenolic compounds from thinned young kiwifruits was established. A maximum yield (105.37 ± 1.2 mg GAE/g DW) of total phenolics extracted from thinned young kiwifruits by UADE was obtained, which was significantly higher than those of conventional organic solvent extraction (CSE, about 14.51 ± 0.26 mg GAE/g DW) and ultrasound-assisted ethanol extraction (UAEE, about 43.85 ± 1.17 mg GAE/g DW). In addition, 29 compounds, e.g., gallic acid, chlorogenic acid, neochlorogenic acid, catechin, epicatechin, procyanidin B1, procyanidin B2, quercetin-3-rhamnoside, and quercetin-3-O-glucoside, were identified in the kiwifruit extract by UPLC-MS/MS. Furthermore, the contents of major phenolic compounds in different kiwifruit extracts prepared by conventional organic solvent extraction (EE), ultrasound-assisted ethanol extraction (UEE), and ultrasound-assisted deep eutectic solvent extraction (UDE) were compared by HPLC analysis. Results revealed that the content of major phenolics in UDE (about 15.067 mg/g DW) was significantly higher than that in EE (about 2.218 mg/g DW) and UEE (about 6.122 mg/g DW), suggesting that the UADE method was more efficient for extracting polyphenolics from thinned young kiwifruits. In addition, compared with EE and UEE, UDE exhibited much higher antioxidant and anti-inflammatory effects as well as inhibitory effects against α-glucosidase and pancreatic lipase, which were closely associated with its higher content of phenolic compounds. Collectively, the findings suggest that the UADE method can be applied as an efficient technique for the preparation of bioactive polyphenolics from thinned young kiwifruits, and the thinned young fruits of red-fleshed A. chinensis cv ‘HY’ have good potential to be developed and utilized as functional foods and nutraceuticals. Full article
Show Figures

Figure 1

Figure 1
<p>Effects of water content in DES solution (<b>A</b>), ultrasonic power (<b>B</b>), ultrasonic time (<b>C</b>), and liquid–solid ratio (<b>D</b>) on the yield of total phenolics extracted from of thinned young kiwifruits by ultrasound-assisted deep eutectic solvent extraction. TPC indicates total phenolic content; different letters (a–e) indicate significant differences at <span class="html-italic">p</span> &lt; 0.05 determined by ANOVA.</p>
Full article ">Figure 2
<p>Three-dimensional response surface plots (<b>A</b>,<b>C</b>,<b>E</b>) and 2D contour plots (<b>B</b>,<b>D</b>,<b>F</b>) of ultrasound-assisted deep eutectic solvent extraction. (<b>A</b>–<b>F</b>) indicate interactions among ultrasonic time, water content in DES, and liquid–solid ratio, respectively.</p>
Full article ">Figure 3
<p>UPLC-MS/MS extracted ions chromatogram (<b>A</b>) and HPLC chromatograms of mixed standards (<b>B</b>–<b>D</b>) and the thinned young kiwifruit extract (<b>E</b>–<b>G</b>). Compounds <b>1</b>–<b>29</b> are the same as in <a href="#antioxidants-12-01475-t003" class="html-table">Table 3</a>.</p>
Full article ">Figure 4
<p>Antioxidant capacities and inhibitory effects on digestive enzymes of the thinned young kiwifruit extracts: (<b>A</b>), the ferric-reducing antioxidant power; (<b>B</b>), the ABTS radical scavenging ability; (<b>C</b>), the DPPH radical scavenging ability; (<b>D</b>), the hydroxyl radical scavenging ability; (<b>E</b>), the inhibitory effect against pancreatic lipase; (<b>F</b>), the inhibitory effect against α-glucosidase. UDE, the thinned kiwifruit extract prepared by ultrasound-assisted deep eutectic solvent extraction; UEE, the thinned kiwifruit extract prepared by ultrasound-assisted ethanol extraction; EE, the thinned kiwifruit extract prepared by conventional organic solvent extraction; different letters (a–c) indicate statistically significant differences (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 5
<p>Pearson correlation matrix of phenolics, antioxidant capacities, inhibitory effects on digestive enzymes, and anti-inflammatory effects. The correlation coefficients are proportional to the circular size and color intensity; * Stands for significant correlation at <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 6
<p>In vitro anti-inflammatory effects of the thinned young kiwifruit extracts: (<b>A</b>), the effect of different thinned young kiwifruit extracts on the cell viability of RAW 264.7 cells; (<b>B</b>), the effect of different thinned young kiwifruit extracts on the NO production from LPS-stimulated RAW 264.7 cells. UDE, the thinned kiwifruit extract prepared by ultrasound-assisted deep eutectic solvent extraction; UEE, the thinned kiwifruit extract prepared by ultrasound-assisted ethanol extraction; EE, the thinned kiwifruit extract prepared by conventional organic solvent extraction; different letters (a, b) indicate statistically significant differences (<span class="html-italic">p</span> &lt; 0.05) among different kiwifruit extracts; significant differences in cell viability and NO production in LPS and kiwifruit extracts vs. control are shown by ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">
19 pages, 3700 KiB  
Article
Natural Extracts Mitigate the Deleterious Effects of Prolonged Intense Physical Exercise on the Cardiovascular and Muscular Systems
by Marc Yehya, Doria Boulghobra, Pierre-Edouard Grillet, Pablo R. Fleitas-Paniagua, Patrice Bideaux, Sandrine Gayrard, Pierre Sicard, Jérome Thireau, Cyril Reboul and Olivier Cazorla
Antioxidants 2023, 12(7), 1474; https://doi.org/10.3390/antiox12071474 - 22 Jul 2023
Cited by 3 | Viewed by 1510
Abstract
Muscle fatigue is a common symptom induced by exercise. A reversible loss of muscle force is observed with variable rates of recovery depending on the causes or underlying mechanisms. It can not only affect locomotion muscles, but can also affect the heart, in [...] Read more.
Muscle fatigue is a common symptom induced by exercise. A reversible loss of muscle force is observed with variable rates of recovery depending on the causes or underlying mechanisms. It can not only affect locomotion muscles, but can also affect the heart, in particular after intense prolonged exercise such as marathons and ultra-triathlons. The goal of our study was to explore the effect of four different natural extracts with recognized antioxidant properties on the contractile function of skeletal (locomotion) and cardiac muscles after a prolonged exhausting exercise. Male Wistar rats performed a bout of exhausting exercise on a treadmill for about 2.5 h and were compared to sedentary animals. Some rats received oral treatment of a natural extract (rosemary, buckwheat, Powergrape®, or rapeseed) or the placebo 24 h and 1 h before exercise. Experiments were performed 30 min after the race and after 7 days of recovery. All natural extracts had protective effects both in cardiac and skeletal muscles. The extent of protection was different depending on muscle type and the duration post-exercise (just after and after one-week recovery), including antiarrhythmic effect and anti-diastolic dysfunction for the heart, and faster recovery of contractility for the skeletal muscles. Moreover, the muscular protective effect varied between natural extracts. Our study shows that an acute antioxidant supplementation can protect against acute abnormal endogenous ROS toxicity, induced here by prolonged exhausting exercise. Full article
(This article belongs to the Section Health Outcomes of Antioxidants and Oxidative Stress)
Show Figures

Figure 1

Figure 1
<p>Effects of the 4 different natural extracts on the isometric strength of soleus after exhausting exercise and one week of recovery. (<b>A</b>) Soleus muscles were isolated from Sedentary (Base), 30 min post-exercise (Exer), and after one-week recovery (Recov). (<b>B</b>) Force–frequency relationships of soleus muscles measured in vitro from Sedentary (black square), 30 min post-exercise (black circle), and after one week of recovery (open circle, dash line) placebo groups. Results are expressed as mean ± SEM, (<span class="html-italic">n</span> = 10–13 animals/group). * <span class="html-italic">p</span> &lt; 0.05 Placebo post-exercise vs. Basal. (<b>C</b>,<b>D</b>) reveal the different contractile characteristics measured in vitro of the Soleus muscles in groups of animals fed with the 4 different natural extracts (rosemary, buckwheat, Powergrape, and rapeseed), either immediately post-exercise or one week later. (<b>C</b>) represents the force induced by a single twitch, and (<b>D</b>) represents the tetanic contraction or peak specific force. Results are expressed as mean ± SEM, (<span class="html-italic">n</span> = 7–13 in each group). <span>$</span> <span class="html-italic">p</span> &lt; 0.05 vs. basal, * <span class="html-italic">p</span> &lt; 0.05 recovery vs. post-exercise.</p>
Full article ">Figure 2
<p>Effects of the 4 different natural extracts on the soleus fatigability after exercise and recovery. (<b>A</b>) Test of fatigue in vitro on soleus muscles in sedentary rats (control, black square), placebo post-exercise (black circle), and placebo one-week recovery (open circle) groups. (<b>B</b>) Effects of rosemary (blue square), buckwheat (red triangle), Powergrape<sup>®</sup> (green triangle), and rapeseed (pink diamond) on in vitro post-exercise muscle fatigue test in soleus compared with placebo (open circle) and controls (black square). (<b>C</b>–<b>E</b>) Effects of the natural extracts on the in vitro muscle fatigue test one week after exercise compared with the placebo (open circle) and controls (black square). (<b>D</b>) Muscle fatigue was also expressed relative to the level of force of the placebo group at each time point. (<b>E</b>) Time to 1/2 loss of maximum strength during fatigue test in the different groups after one week of recovery. (<span class="html-italic">n</span> = 7–13 animals per group). * <span class="html-italic">p</span> &lt; 0.05 placebo vs. Control, rosemary, buckwheat, Powergrape<sup>®</sup>, and rapeseed groups.</p>
Full article ">Figure 3
<p>Effect of natural extracts on aortic endothelium-dependent and endothelium-independent vasorelaxation after prolonged exhausting exercise. (<b>A</b>) Schematic representation of the experimental design. Dose–response curves (<b>B</b>) and maximal response (<b>C</b>) to acetylcholine (ACh) obtained in aortic rings pre-contracted with phenylephrine (PE, 10<sup>−6</sup> M) (<span class="html-italic">n</span> = 13–15 aortic rings from 5–6 different rats per condition) for control (Ctrl, black square), placebo (open circle), Rosemary (blue circle), Buckwheat (red circle), grape (green circle), and rapeseed (pink circle). Each dot is one preparation. Dose–response curves (<b>D</b>) and maximal response (<b>E</b>) to sodium nitroprusside (SNO) obtained in aortic rings pre-contracted with phenylephrine (PE, 10<sup>−6</sup> M) (<span class="html-italic">n</span> = 13–15 aortic rings from 5–6 different rats per condition). All values are the mean ± SEM.</p>
Full article ">Figure 4
<p>Effects of natural extracts on in vivo cardiac function after exercise. (<b>A</b>) The cardiac function was evaluated in vivo by echocardiography at baseline (base, black square), 30 min after PEE (Exer, black circle), and 1 week after exercise (Recov, open circle) in placebo and extract-treated animals. (<b>B</b>) The function of contraction or systole was indexed by the ejection fraction. The phase of relaxation or diastole of the heart was indexed by the E/A ratios of early diastolic and atrial transmitral inflow velocities (<b>C</b>), as well as the E′/A′ ratios obtained by tissue Doppler imaging (<b>D</b>). (<span class="html-italic">n</span> = 10/group). * <span class="html-italic">p</span> &lt; 0.05 repeated measures ANOVA.</p>
Full article ">Figure 5
<p>Effects of natural extracts on ex vivo cardiac function after exhausting exercise. (<b>A</b>) Schematic representation of the experimental design. (<b>B</b>) Representative trace of the left ventricular (LV) pressure (upper panel) and its first derivative (lower panel) obtained from Langendorff hearts isolated from Ctrl and Placebo-exercised rats. (<b>C</b>) LV developed pressure obtained from hearts 30 min after exhausting exercise in rats treated or not treated with 4 different natural extracts (<span class="html-italic">n</span> = 7–13 hearts per condition). Maximal first derivative of LV pressure (dP/dtmax) (<b>D</b>) and minimal first derivative of LV pressure (dP/dtmin) (<b>E</b>) obtained from Langendorff isolated hearts 30 min after exhausting exercise in rats treated or not treated with 4 different natural extracts (<span class="html-italic">n</span> = 7–13 hearts per condition). * <span class="html-italic">p</span> &lt; 0.05 vs. Ctrl ANOVA followed by adjusted <span class="html-italic">t</span>-test; All values are the mean ± SEM.</p>
Full article ">Figure 6
<p>Effects of natural extracts on in vivo electrical cardiac function after exercise. (<b>A</b>) The ECG was recorded in vivo for the same rat at baseline (base, black square), 30 min after PEE (Exer, black circle), and 1 week after exercise (Recov, open circle) in placebo and extract-treated animals. (<b>B</b>) Heart rate (RR interval in ms) and (<b>C</b>) QT duration (in ms) were analyzed. (<b>D</b>) Example of ECG recording of a rat with ventricular extrasystole. (<b>E</b>) Number of ventricular extrasystoles (number/30 min) in extract-treated animals. (<span class="html-italic">n</span> = 8/group). * <span class="html-italic">p</span> &lt; 0.05 repeated measures ANOVA.</p>
Full article ">
16 pages, 4730 KiB  
Article
Synthesis, Characterization and Evaluation of a Novel Tetraphenolic Compound as a Potential Antioxidant
by Mengqi Xu, Pengcheng Meng, Hongyan Wang, Jun Liu, Tao Guo, Zhenjie Zhu and Yanlan Bi
Antioxidants 2023, 12(7), 1473; https://doi.org/10.3390/antiox12071473 - 22 Jul 2023
Cited by 3 | Viewed by 1773
Abstract
A novel antioxidant containing four hydroxyl groups, namely 2,2′-(2-methylpropane-1,3-diyl)bis(hydroquinone) (MPBHQ), was synthesized using hydroquinone and methylallyl alcohol as the raw materials, phosphoric acid as the catalyst, and toluene as the solvent system. The structure of MPBHQ was characterized by mass spectrometry, nuclear magnetic [...] Read more.
A novel antioxidant containing four hydroxyl groups, namely 2,2′-(2-methylpropane-1,3-diyl)bis(hydroquinone) (MPBHQ), was synthesized using hydroquinone and methylallyl alcohol as the raw materials, phosphoric acid as the catalyst, and toluene as the solvent system. The structure of MPBHQ was characterized by mass spectrometry, nuclear magnetic resonance, ultraviolet spectroscopy, and infrared spectroscopy. The results showed that MPBHQ has a good radical scavenging effect, as measured by the ORAC assay, DPPH radical scavenging assay, ABST radical scavenging assay, and Rancimat test. In fatty acid methyl ester and lard without exogenous antioxidants, MPBHQ showed better antioxidant performance than butylated hydroxytoluene (BHT), hydroquinone (HQ), tert-butyl hydroquinone (TBHQ), and propyl gallate (PG), meeting the need for a new antioxidant with better properties to ensure the oxidative stability of lipids and biodiesel. Full article
(This article belongs to the Special Issue Antioxidants and Oxidative Stability in Fats and Oils)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>(<b>a</b>) Possible products in the synthesis reaction. (<b>b</b>) Ultraviolet spectrogram of MPBHQ solution and the mixture of MPBHQ and KOH. (<b>c</b>) The FTIR spectra of MPBHQ, HQ and TBHQ.</p>
Full article ">Figure 2
<p>The primary (<b>a</b>) and secondary (<b>b</b>) ion mass spectra of MPBHQ.</p>
Full article ">Figure 3
<p>NMR spectra of MPBHQ, including <sup>1</sup>H NMR (<b>a</b>), <sup>13</sup>C NMR (<b>b</b>), <sup>1</sup>H-<sup>1</sup>H COSY-NMR (<b>c</b>), <sup>13</sup>C-<sup>1</sup>H HSQC-NMR (<b>d</b>), and <sup>13</sup>C-<sup>1</sup>H HMBC-NMR (<b>e</b>).</p>
Full article ">Figure 3 Cont.
<p>NMR spectra of MPBHQ, including <sup>1</sup>H NMR (<b>a</b>), <sup>13</sup>C NMR (<b>b</b>), <sup>1</sup>H-<sup>1</sup>H COSY-NMR (<b>c</b>), <sup>13</sup>C-<sup>1</sup>H HSQC-NMR (<b>d</b>), and <sup>13</sup>C-<sup>1</sup>H HMBC-NMR (<b>e</b>).</p>
Full article ">Figure 4
<p>(<b>a</b>) DPPH radical scavenging activity of different antioxidants. (<b>b</b>) ABTS radical scavenging activity of different antioxidants. (<b>c</b>) The attenuation curve of fluorescence intensity of Trolox solution. (<b>d</b>) ORAC values of different antioxidants. The different lowercase letters in the graph, including a, b, c, d, e, and f, indicate the significant difference of EC<sub>50</sub> values of the same substance at different concentrations (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 5
<p>(<b>a</b>) Changes in Pf values of lard containing 0.02 wt.% of antioxidants at different temperatures. (<b>b</b>) Changes in Pf values of lard containing different antioxidants concentrations at 120 °C. (<b>c</b>) Changes in Pf values of fatty acid methyl ester containing 0.02 wt.% of antioxidants at different temperatures. (<b>d</b>) Changes in Pf values of fatty acid methyl ester containing different antioxidants concentrations at 120 °C. The chemical structure of the five antioxidants is shown below. The different lowercase letters in the graph, including a, b, c, d, e, and f, indicate the significant difference of Pf values of the different substances at same condition (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Scheme 1
<p>Flowchart on the synthesis and harvest process of MPBHQ crude products.</p>
Full article ">
25 pages, 367 KiB  
Review
Olive Oil Phenolic Compounds’ Activity against Age-Associated Cognitive Decline: Clinical and Experimental Evidence
by Anna Boronat, Gabriele Serreli, Jose Rodríguez-Morató, Monica Deiana and Rafael de la Torre
Antioxidants 2023, 12(7), 1472; https://doi.org/10.3390/antiox12071472 - 22 Jul 2023
Cited by 2 | Viewed by 2374
Abstract
Epidemiological studies have shown that consuming olive oil rich in phenolic bioactive compounds is associated with a lower risk of neurodegenerative diseases and better cognitive performance in aged populations. Since oxidative stress is a common hallmark of age-related cognitive decline, incorporating exogenous antioxidants [...] Read more.
Epidemiological studies have shown that consuming olive oil rich in phenolic bioactive compounds is associated with a lower risk of neurodegenerative diseases and better cognitive performance in aged populations. Since oxidative stress is a common hallmark of age-related cognitive decline, incorporating exogenous antioxidants could have beneficial effects on brain aging. In this review, we firstly summarize and critically discuss the current preclinical evidence and the potential neuroprotective mechanisms. Existing studies indicate that olive oil phenolic compounds can modulate and counteract oxidative stress and neuroinflammation, two relevant pathways linked to the onset and progression of neurodegenerative processes. Secondly, we summarize the current clinical evidence. In contrast to preclinical studies, there is no direct evidence in humans of the bioactivity of olive oil phenolic compounds. Instead, we have summarized current findings regarding nutritional interventions supplemented with olive oil on cognition. A growing body of research indicates that high consumption of olive oil phenolic compounds is associated with better preservation of cognitive performance, conferring an additional benefit, independent of the dietary pattern. In conclusion, the consumption of olive oil rich in phenolic bioactive compounds has potential neuroprotective effects. Further research is needed to understand the underlying mechanisms and potential clinical applications. Full article
15 pages, 2348 KiB  
Article
1,3-Butanediol Administration Increases β-Hydroxybutyrate Plasma Levels and Affects Redox Homeostasis, Endoplasmic Reticulum Stress, and Adipokine Production in Rat Gonadal Adipose Tissue
by Giuliana Panico, Gianluca Fasciolo, Vincenzo Migliaccio, Rita De Matteis, Lillà Lionetti, Gaetana Napolitano, Claudio Agnisola, Paola Venditti and Assunta Lombardi
Antioxidants 2023, 12(7), 1471; https://doi.org/10.3390/antiox12071471 - 22 Jul 2023
Cited by 1 | Viewed by 1837
Abstract
Ketone bodies (KBs) are an alternative energy source under starvation and play multiple roles as signaling molecules regulating energy and metabolic homeostasis. The mechanism by which KBs influence visceral white adipose tissue physiology is only partially known, and our study aimed to shed [...] Read more.
Ketone bodies (KBs) are an alternative energy source under starvation and play multiple roles as signaling molecules regulating energy and metabolic homeostasis. The mechanism by which KBs influence visceral white adipose tissue physiology is only partially known, and our study aimed to shed light on the effects they exert on such tissue. To this aim, we administered 1,3-butanediol (BD) to rats since it rapidly enhances β-hydroxybutyrate serum levels, and we evaluated the effect it induces within 3 h or after 14 days of treatment. After 14 days of treatment, rats showed a decrease in body weight gain, energy intake, gonadal-WAT (gWAT) weight, and adipocyte size compared to the control. BD exerted a pronounced antioxidant effect and directed redox homeostasis toward reductive stress, already evident within 3 h after its administration. BD lowered tissue ROS levels and oxidative damage to lipids and proteins and enhanced tissue soluble and enzymatic antioxidant capacity as well as nuclear erythroid factor-2 protein levels. BD also reduced specific mitochondrial maximal oxidative capacity and induced endoplasmic reticulum stress as well as interrelated processes, leading to changes in the level of adipokines/cytokines involved in inflammation, macrophage infiltration into gWAT, adipocyte differentiation, and lipolysis. Full article
(This article belongs to the Special Issue The 10th Anniversary of Antioxidants: Past, Present and Future)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Effect of BD treatment on (D) β-OHB serum levels, detected after 3 h from BD i.p administration (BD 3 h) and after 14 days of oral administration (BD 14 d). Values represent the mean ± SE of 10 different animals. * <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 2
<p>Effect of 14 days BD administration on gWAT morphology and lipolysis. (<b>A</b>) shows a representative histological analysis of gWAT from control (C) and BD-treated rats (BD 14 d). Below is the enlargement (100×) of the framed areas. Note the presence of capillaries (arrow) and infiltrating (inflammatory) cells/macrophages (arrowhead) around the adipocytes in BD 14 d-compared to C-gWAT. Hematoxylin and Eosin staining. (<b>B</b>) represents Basal and isoprotenerol-stimulated lipolysis. Values represent the mean ± ES of 8 different animals * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 3
<p>Effect of BD on gWAT redox homeostasis. Total ROS (<b>A</b>), lipid hydroperoxides (<b>B</b>), protein oxidative damage (<b>C</b>), and tissue soluble antioxidant capacity (<b>D</b>). Values are means ± SEM of 8 different rats. * <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. Enzymatic antioxidant capacity (<b>E</b>). Representative Western Blot of Superoxide dismutase-2 (SOD-2), catalase (CAT), peroxiredoxin-3 (PRDX-3), and Nuclear factor erythroid-related factor 2 (Nrf2) (<b>E</b>). GAPDH was used as loading control (25 μg of protein/rat/lane). Histograms represent the quantification of data. Data were normalized to the value obtained for control animals, set as 100. Values represent the mean ± SEM of 6 different rats or 3 rats in the case of PRDX-3.</p>
Full article ">Figure 4
<p>Effect of BD on gWAT levels of proteins linked to ER and cellular stress involved in ER stress and UPR<sup>ER</sup> response detected. Representative Western blots of GRP78/BiP, Calnexin and TnF-α (<b>A</b>), and total and phosphorylated (ser 78) levels of eIF2α (<b>B</b>) were performed in gWAT lysate. GAPDH was used as loading control (25 μg of protein/rat/lane). Histograms represent the quantification of data. Data were normalized to the value obtained for control animals, set as 100. Values represent the mean ± SEM of 6 different rats. * <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; 0.0001.</p>
Full article ">Figure 5
<p>Effect of BD on gWAT mitochondrial respiratory complexes abundance and cytochrome oxidase activity. (<b>A</b>) shows representative Western Blots of specific subunits of the five mitochondrial respiratory complexes, namely CI-NDUF88, CII-SDHB, CIII-UQCRC2, CIV-MTCO1, and CV-ATP VA. GAPDH was used as loading control (25 μg of protein/rat/lane). Histograms represent the quantification of data. Data were normalized to the value obtained for control animals, set as 100. Values represent the mean ± SEM of 6 different rats. (<b>B</b>,<b>C</b>) show Cytochrome oxidase/complex IV activity reported as nmoles O/min mg tissue proteins (<b>B</b>) or normalized for Complex IV levels and expressed in arbitrary units (<b>C</b>). * <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. Values represent the mean ± SEM of 8 different rats.</p>
Full article ">Figure 6
<p>Effect of administration of BD for 14 days on gWAT adipokines profiles. Representative adipokine profile detected in gWAT from C and BD 14 d rats, by an adipokine protein array (<b>A</b>). The arrowheads indicate signals with significant changes. Magnifications of the spot of proteins whose intensity resulted statistically different are reported in (<b>B</b>). Histograms represent the quantification of relative levels of adipokines with observable changes (<b>C</b>). The values represent the mean ± SEM of 3 different samples, each one obtained by a pool of two different animals. Data were normalized to the value obtained for control animals, set as 100. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">
13 pages, 675 KiB  
Article
Oxidative Stress and Performance after Training in Professional Soccer (European Football) Players
by Michele Abate, Raffaello Pellegrino, Angelo Di Iorio and Vincenzo Salini
Antioxidants 2023, 12(7), 1470; https://doi.org/10.3390/antiox12071470 - 22 Jul 2023
Viewed by 1219
Abstract
Vitamins, hormones, free radicals, and antioxidant substances significantly influence athletic performance. The aim of this study was to evaluate whether these biological mediators changed during the season and if this was associated with the rate of improvement in performance after training, assessed by [...] Read more.
Vitamins, hormones, free radicals, and antioxidant substances significantly influence athletic performance. The aim of this study was to evaluate whether these biological mediators changed during the season and if this was associated with the rate of improvement in performance after training, assessed by means of a standardized test. Professional male soccer players took part in the study. Two evaluations were performed: the first in the pre-season period and the second at the mid-point of the official season, after about 6 months of intensive training and weekly matches. Blood levels of vitamins D, B12, and folic acid, testosterone and cortisol, free radicals, and antioxidant substances were measured. Two hours after breakfast, a Yo-Yo test was performed. The relationships between the biological mediators and the rate of improvement after training (i.e., the increase in meters run in the Yo-Yo test between the pre-season and mid-season periods) were evaluated by means of a linear mixed models analysis. Results: Eighty-two paired tests were performed. The athletes showed better performance after training, with an increase in the meters run of about 20%. No significant relationships between the vitamin and hormone values and the gain in the performance test were observed. Plasmatic levels of free radicals increased significantly, as did the blood antioxidant potential. An indirect relationship between oxidative stress and the improvement in performance was observed (free radicals β ± SE: = −0.33 ± 0.10; p-value = 0.001), with lower levels of oxidative stress being associated with higher levels of performance in the Yo-Yo test. Monitoring the measures of oxidative stress could be a useful additional tool for coaches in training and/or recovery programs tailored to each player. Full article
(This article belongs to the Special Issue Exercise-Induced Antioxidant Response and Oxidative Stress)
Show Figures

Figure 1

Figure 1
<p>Linear mixed models analysis residuals versus the YoYo test according to time. The dashed line represents the pre-season test; the continuous line represents the mid-season test. The starting point of the lines refers to the athletes who had the worst performance, whereas the end point refers to the athletes who had the best performance.</p>
Full article ">Figure 2
<p>Linear mixed model analysis residuals versus age according to the time of the study. The cortisol serum levels were adjusted.</p>
Full article ">
22 pages, 2309 KiB  
Review
Biosynthesis, Deficiency, and Supplementation of Coenzyme Q
by Carmine Staiano, Laura García-Corzo, David Mantle, Nadia Turton, Lauren E. Millichap, Gloria Brea-Calvo and Iain Hargreaves
Antioxidants 2023, 12(7), 1469; https://doi.org/10.3390/antiox12071469 - 21 Jul 2023
Cited by 2 | Viewed by 3807
Abstract
Originally identified as a key component of the mitochondrial respiratory chain, Coenzyme Q (CoQ or CoQ10 for human tissues) has recently been revealed to be essential for many different redox processes, not only in the mitochondria, but elsewhere within other cellular membrane [...] Read more.
Originally identified as a key component of the mitochondrial respiratory chain, Coenzyme Q (CoQ or CoQ10 for human tissues) has recently been revealed to be essential for many different redox processes, not only in the mitochondria, but elsewhere within other cellular membrane types. Cells rely on endogenous CoQ biosynthesis, and defects in this still-not-completely understood pathway result in primary CoQ deficiencies, a group of conditions biochemically characterised by decreased tissue CoQ levels, which in turn are linked to functional defects. Secondary CoQ deficiencies may result from a wide variety of cellular dysfunctions not directly linked to primary synthesis. In this article, we review the current knowledge on CoQ biosynthesis, the defects leading to diminished CoQ10 levels in human tissues and their associated clinical manifestations. Full article
(This article belongs to the Special Issue The Ubiquitous and Multifaceted Coenzyme Q)
Show Figures

Figure 1

Figure 1
<p><b>CoQ biosynthesis pathway.</b> A compendium of the reactions carried out to synthesize CoQ in bacteria (enzymes in green), yeast (enzymes in orange), plants (enzymes in pink), and mammals (enzymes in purple) is depicted including the name of the proteins involved. Proteins whose functions are still unknown are indicated separately. For simplicity, only some proteins involved in the 4HB biosynthesis in mammals have been indicated. For an extensive revision on 4HB biosynthesis, see Fernández-del-Río and Clarke, 2021 [<a href="#B21-antioxidants-12-01469" class="html-bibr">21</a>]. PAH, phenylalanine hydrolase; TAT, tyrosine aminotransferase; AADAT, mitochondrial alpha-aminoadipate aminotransferase; ALDH3A1, aldehyde dehydrogenase 3A1; HPDL, hydroxyphenylpyruvate dioxygenase-like [<a href="#B22-antioxidants-12-01469" class="html-bibr">22</a>]; simbols ? and ?? indicate enzyme/s still not identified.</p>
Full article ">Figure 2
<p><b>Statin associated side effects.</b> SAMS—Statin associated muscle symptoms; HFpEF—Heart failure with preserved ejection fraction; BBB—Blood–brain barrier.</p>
Full article ">Figure 3
<p><b>CoQ<sub>10</sub> transport across the blood–brain barrier under normal conditions.</b> SR-B1: Scavenger receptor B1; LRP-1: Low Density Lipoprotein related protein-1; RAGE: Receptor Advanced Glycation End-products; P-gp: P-glycoprotein transporter. Although P-gp has been shown to mediate CoQ<sub>10</sub> transport in other cell lines, it does not participate in its movement across the BBB [<a href="#B147-antioxidants-12-01469" class="html-bibr">147</a>].</p>
Full article ">
17 pages, 2078 KiB  
Article
Micromolar Dihydroartemisinin Concentrations Elicit Lipoperoxidation in Plasmodium falciparum-Infected Erythrocytes
by Oleksii Skorokhod, Elena Valente, Giorgia Mandili, Daniela Ulliers and Evelin Schwarzer
Antioxidants 2023, 12(7), 1468; https://doi.org/10.3390/antiox12071468 - 21 Jul 2023
Cited by 3 | Viewed by 1469
Abstract
Malaria is still the most important parasitic infectious disease. Numerous substances are known to have antimalarial activity; among them, artemisinin is the most widely used one, and artemisinin-based combination therapy (ACT) is recommended for the treatment of Plasmodium falciparum (P.f.) malaria. Antitumor, immunomodulatory, [...] Read more.
Malaria is still the most important parasitic infectious disease. Numerous substances are known to have antimalarial activity; among them, artemisinin is the most widely used one, and artemisinin-based combination therapy (ACT) is recommended for the treatment of Plasmodium falciparum (P.f.) malaria. Antitumor, immunomodulatory, and other therapeutic applications of artemisinin are under extensive study. Several different mechanisms of action were proposed for dihydroartemisinin (DHA), the active metabolite of artemisinin, such as eliciting oxidative stress in target cells. The goal of this study is to monitor the generation of reactive oxygen species (ROS) and lipid peroxidation product 4-hydroxynonenal (4-HNE) by DHA in P.f.-infected human erythrocytes. Checking ROS and 4-HNE-protein adducts kinetics along the maturation of the parasite, we detected the highest level of 4-HNE in ring forms of P.f. due to DHA treatment. Low micromolar concentrations of DHA quickly induced levels of 4-HNE-adducts which are supposed to be damaging. Mass spectrometry identified the P.f. protein cysteine proteinase falcipain-1 as being heavily modified by 4-HNE, and plausibly, 4-HNE conjugation with vital P.f. proteins might contribute to DHA-elicited parasite death. In conclusion, significant 4-HNE accumulation was detectable after DHA treatment, though, at concentrations well above pharmacologically effective ranges in malaria treatment, but at concentrations described for antitumor activity. Thus, lipid peroxidation with consequent 4-HNE conjugation of functionally relevant proteins might be considered as a uniform mechanism for how DHA potentiates antimalarials’ action in ACT and controls the progression of tumors. Full article
(This article belongs to the Section Aberrant Oxidation of Biomolecules)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p><b>Common DHA effects on ring-stage <span class="html-italic">P.f.</span>-infected RBC.</b> (<b>A</b>) The chemical structure of DHA is shown with oxygen atoms in red, including the functional endoperoxide group (National Center for Biotechnology Information. PubChem Compound Summary for CID 540327, Dihydroartemisinin, <a href="https://pubchem.ncbi.nlm.nih.gov/compound/Dihydroartemisinin" target="_blank">https://pubchem.ncbi.nlm.nih.gov/compound/Dihydroartemisinin</a>, accessed on 30 May 2023). (<b>B</b>) Morphological damage and growth delay of <span class="html-italic">P.f</span>. parasites were assessed in synchronous in vitro cultures of asexual stages of parasites grown in erythrocytes and treated at the ring stage (time 0) with 0, 10, and 100 nM DHA for 6 and 24 h. Microscopic images taken after Diff-Quik<sup>®</sup>-stained smears show damaged and shrunken parasite forms after DHA treatment. Images were acquired using an inverted microscope Leica DM IRB (Leica Microsystems, Wetzlar, Germany) equipped with a 100× oil planar apochromatic objective with 1.32 numerical aperture and a Leica camera DFC 420 C. Leica DFC software (version 3.3.1) was applied.</p>
Full article ">Figure 2
<p><b>Inhibition of parasite growth by DHA</b>. Synchronized <span class="html-italic">P. falciparum</span> cultures were treated at the ring stage (12–16 h after reinfection, grey columns) and the trophozoite stage (26–30 h after reinfection, black columns) with 0–10,000 nM DHA. Parasite growth was assessed at 6 h after the start of DHA exposure by flow cytometry after cell staining with ethidium bromide (EtBr). Mean fluorescence intensities (MFIs) are shown as mean ± SE of 3–5 independent experiments performed with erythrocytes of different donors. The significance of differences vs. untreated control cultures (0 µM DHA) is indicated by * for <span class="html-italic">p</span> &lt; 0.01 and ** for <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 3
<p><b>Damage of ring-stage P. falciparum and decrease in multiplication rate by DHA</b>. (<b>A</b>) Synchronized <span class="html-italic">P. falciparum</span> cultures were treated at the ring stage (12–16 h after reinfection) with 0–1000 nM DHA (time 0; T = 0) and examined for parasitemia at 24 h after DHA supplementation (T = 24) by manual microscopic inspection of Diff-Quik<sup>®</sup>-stained smears. Parasites with irregular shapes (distorted, broken, pyknotic, and fragmented forms) were counted as damaged (black columns) separately from infected erythrocytes harboring normally shaped viable parasites (gray columns). (<b>B</b>) The multiplication rate was determined for <span class="html-italic">P.f.</span>, which were treated at the ring stage with DHA and calculated as the ratio of parasitemias of viable parasites after and before reinfection of freshly added erythrocytes. The parasitemia before reinfection was adjusted by supplementation with fresh non-infected erythrocytes at a schizont parasitemia of 4.93 ± 0.6% in the case of not DHA-treated synchronous control cultures (DHA 0 nM). Parasite cultures pre-treated with 1–1000 nM DHA at the ring stage were identically supplemented with fresh erythrocytes prior to reinfection, even though alive schizont parasitemias were lower due to DHA treatment as compared to untreated control culture. Parasitemia after reinfection was counted 9 h after adding non-infected donor erythrocyte suspension. (<b>C</b>) The absence of DHA-elicited hemolysis is shown as a normalized hemolysis rate. Free hemoglobin was quantified in culture supernatant after 6 h of DHA treatment. Supernatant hemoglobin was referred to as total hemoglobin in the RBC suspension to obtain the hemolysis rate. These rates were normalized by referring any measured value to the hemolysis rate assessed in respective untreated cultures with erythrocytes from the same donor. Means ± SE are shown for independent cultures with RBCs from 3 different donors. The significances of differences between treated (0–1000 nM DHA) and untreated control cultures (0 nM DHA) are indicated by * for <span class="html-italic">p</span> &lt; 0.05 for damaged parasites, § for <span class="html-italic">p</span> &lt; 0.05 for total parasitemia (viable plus damaged parasites), and # for <span class="html-italic">p</span> &lt; 0.05 for multiplication rate of parasites.</p>
Full article ">Figure 4
<p><b>ROS production by DHA in infected erythrocytes</b>. Synchronized <span class="html-italic">P. falciparum</span> cultures were adjusted to 10% parasitemia and treated with 0–10 µM DHA at the ring stage (12–16 h after reinfection). Where present, 100 µM NAC was added 30 min prior to DHA to the parasites culture. In parallel, an otherwise untreated culture was incubated with pro-oxidant xanthine/xanthine oxidase as a positive control for ROS production. Cells were stained with the ROS-sensitive fluorescent probes DCF-DA (<b>A</b>) and DHE (<b>B</b>) at 3 h after pre-incubation with DHA. The probe was separately assessed in non-infected (white columns) and infected (grey/black columns) erythrocytes distinguished by ethidium bromide using flow cytometry. The mean fluorescence intensity (MFI) of labeled cells was normalized by setting the MFI of corresponding non-infected, not-DHA-treated erythrocytes as 1. Means ± SE for 3–5 erythrocyte donors are plotted. The significance of the differences vs. untreated control cultures (0 µM DHA) is indicated by * for <span class="html-italic">p</span> &lt; 0.01 and ** for <span class="html-italic">p</span> &lt; 0.05. The significance of difference between 10 µM DHA-treated <span class="html-italic">P.f.</span> cultures pretreated with NAC or not is indicated by §.</p>
Full article ">Figure 5
<p><b>Slow increase in 4-HNE conjugate formation in infected erythrocytes by DHA</b>. Synchronized <span class="html-italic">P.f.</span> cultures were treated at the ring stage (12–16 h after reinfection, <b>A</b>,<b>B</b>) and the trophozoite stage (26–30 h after reinfection, <b>C</b>,<b>D</b>) with 0–1000 nM DHA for 3–24 h. 4-HNE-conjugate formation was assessed in intact infected (<b>A</b>,<b>C</b>) and non-infected erythrocytes from the same cultures (<b>B</b>,<b>D</b>) using flow cytometry at indicated time points after DHA addition at time 0 and expressed as MFI normalized to isotype control. Means ± SE are presented for independent cultures from 4 RBC donors (<b>A</b>,<b>B</b>) and 3 RBC donors (<b>C</b>,<b>D</b>). The significance of differences between DHA-treated and non-treated cultures (<b>A</b>–<b>D</b>) at the same time point is indicated by * for <span class="html-italic">p</span> &lt; 0.05. The significance of differences between untreated cultures (concentration 0, white columns) at different time points and the starting point of observation (time 0) is indicated by § for <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">
16 pages, 1885 KiB  
Article
Evaluation of the Antioxidant and Anti-Lipoxygenase Activity of Berberis vulgaris L. Leaves, Fruits, and Stem and Their LC MS/MS Polyphenolic Profile
by Anna Och, Marta Olech, Kamil Bąk, Sebastian Kanak, Anna Cwener, Marek Cieśla and Renata Nowak
Antioxidants 2023, 12(7), 1467; https://doi.org/10.3390/antiox12071467 - 21 Jul 2023
Cited by 4 | Viewed by 1697
Abstract
Berberis vulgaris L. is currently widely studied for its antioxidant and chemopreventive properties, especially with regard to the beneficial properties of its fruits. Although the bark and roots have been well known and used in traditional medicine since ancient times, little is known [...] Read more.
Berberis vulgaris L. is currently widely studied for its antioxidant and chemopreventive properties, especially with regard to the beneficial properties of its fruits. Although the bark and roots have been well known and used in traditional medicine since ancient times, little is known about the other parts of this plant. The aim of the research was to determine the antioxidant and LOX inhibitory activity effects of extracts obtained from the leaves, fruits, and stems. Another aim of the work was to carry out the quantitative and qualitative analysis of phenolic acids, flavonoid aglycones, and flavonoid glycosides. The extracts were obtained with the use of ASE (accelerated solvent extraction). The total content of polyphenols was determined and was found to vary depending on the organ, with the highest amount of polyphenols found in the leaf extracts. The free radical scavenging activity of the extracts was determined spectrophotometrically in relation to the DPPH (2,2-diphenyl-1-picrylhydrazyl) radical, with results ranging from 63.9 mgTE/g for the leaves to 65.2 mgTE/g for the stem. Antioxidant activity was also assessed using the ABTS test. The lowest value was recorded for the barberry fruit (117.9 mg TE/g), and the highest level was found for the barberry leaves (140.5 mgTE/g). The oxygen radical absorbance capacity test (ORAC) showed the lowest value for the stem (167.7 mgTE/g) and the highest level for the leaves (267.8 mgTE/g). The range of the percentage inhibition of LOX was determined as well. The percentage inhibition of the enzyme was positively correlated with the sum of the flavonoids, TPC, TFC, and the content of selected flavonoids. Phenolic acids, flavonoid aglycones, and flavonoid glycosides were determined qualitatively and quantitatively in individual parts of Berberis vulgaris L. The content of phenolic acids, flavonoid aglycones, and flavonoid glycosides was determined with the LC-MS/MS method. The following phenolic acids were quantitatively and qualitatively identified in individual parts of Berberis vulgaris L.: gallic acid, 3-caffeoylquinic acid, protocatechuic acid, 5-caffeoylquinic acid, 4-caffeoylquinic acid, and caffeic acid. The flavonoid glycosides determined were: eleutheroside E, Eriodictyol-7-glucopyranoside, rutin, hyperoside, isoquercitin, luteoloside, narcissoside, naringenin-7-glucoside, isorhamnetin-3-glucoside, afzeline, and quercitrin. Flavonoid aglycones such as catechin, luteolin, quercetin, and eriodictyol were also determined qualitatively and quantitatively. Full article
(This article belongs to the Section Natural and Synthetic Antioxidants)
Show Figures

Figure 1

Figure 1
<p>Dried parts of plant material obtained for investigations: (<b>a</b>) fruits, (<b>b</b>), leaves, and (<b>c</b>) stem.</p>
Full article ">Figure 1 Cont.
<p>Dried parts of plant material obtained for investigations: (<b>a</b>) fruits, (<b>b</b>), leaves, and (<b>c</b>) stem.</p>
Full article ">Figure 2
<p>Extracted ion chromatograms of phenolic acids detected in the <span class="html-italic">Berberis</span> stem, obtained in the multiple reaction monitoring (MRM) mode: 1—gallic acid; 2—3-caffeoylquinic acid; 3—protocatechuic acid; 4—5-caffeoylquinic acid; 5—4-caffeoylquinic acid; 6—caffeic acid.</p>
Full article ">Figure 3
<p>Principal component analysis of chemical components of investigated phenolic acids, flavonoid aglycones, and flavonoid glycosides in the barberry extracts. Analysis was performed using Statistica 6.0.; length of gradient of 1st axis, 1.294; cumulative percentage variance of data of 1st axis, 82.5; cumulative percentage variance of data of 2nd axis, 17.5.</p>
Full article ">
21 pages, 1709 KiB  
Review
Aldehyde Dehydrogenase and Aldo-Keto Reductase Enzymes: Basic Concepts and Emerging Roles in Diabetic Retinopathy
by Burak Mugdat Karan, Karis Little, Josy Augustine, Alan W. Stitt and Tim M. Curtis
Antioxidants 2023, 12(7), 1466; https://doi.org/10.3390/antiox12071466 - 21 Jul 2023
Cited by 2 | Viewed by 2492
Abstract
Diabetic retinopathy (DR) is a complication of diabetes mellitus that can lead to vision loss and blindness. It is driven by various biochemical processes and molecular mechanisms, including lipid peroxidation and disrupted aldehyde metabolism, which contributes to retinal tissue damage and the progression [...] Read more.
Diabetic retinopathy (DR) is a complication of diabetes mellitus that can lead to vision loss and blindness. It is driven by various biochemical processes and molecular mechanisms, including lipid peroxidation and disrupted aldehyde metabolism, which contributes to retinal tissue damage and the progression of the disease. The elimination and processing of aldehydes in the retina rely on the crucial role played by aldehyde dehydrogenase (ALDH) and aldo-keto reductase (AKR) enzymes. This review article investigates the impact of oxidative stress, lipid-derived aldehydes, and advanced lipoxidation end products (ALEs) on the advancement of DR. It also provides an overview of the ALDH and AKR enzymes expressed in the retina, emphasizing their growing importance in DR. Understanding the relationship between aldehyde metabolism and DR could guide innovative therapeutic strategies to protect the retina and preserve vision in diabetic patients. This review, therefore, also explores various approaches, such as gene therapy and pharmacological compounds that have the potential to augment the expression and activity of ALDH and AKR enzymes, underscoring their potential as effective treatment options for DR. Full article
Show Figures

Figure 1

Figure 1
<p><b>Mechanisms of neurovascular unit dysfunction in DR.</b> Under normal conditions, the neurovascular unit (NVU) comprises vascular endothelial cells, pericytes, glial cells, microglia, and neurons, which work together to maintain neurovascular coupling and the integrity of the inner blood-retinal barrier (iBRB). In diabetes, the NVU is disrupted through several mechanisms. Müller cells, crucial for supporting retinal neurons, undergo gliosis and exhibit a pro-inflammatory state. Additionally, they experience impaired regulation of glutamate and potassium levels in the extracellular space. These changes disrupt neurovascular coupling, compromise iBRB integrity, and lead to neuronal apoptosis. In parallel, microglia become pro-inflammatory and contribute to neurovascular degeneration. Increased oxidative stress and inflammation result in pericyte cell death and compromised tight junctions at the iBRB. The breakdown of iBRB leads to hemorrhage and leakage from blood vessels, facilitating the infiltration of pro-inflammatory cells from the circulation.</p>
Full article ">Figure 2
<p><b>Pathways of Oxidative Stress in DR.</b> Retinal hyperglycemia triggers multiple mechanisms that culminate in oxidative stress, as described from left to right in the figure. Dysregulated autophagy disrupts retinal antioxidant mechanisms, leading to an imbalance between the production of ROS and antioxidant activity. Hyperglycemia further enhances ROS production and affects various metabolic pathways in the retina, including the polyol pathway, hexosamine pathway, formation of advanced glycation end products (AGEs), and the protein kinase C (PKC) pathway. These pathways promote oxidative stress, and elevated levels of ROS, in turn, further activating these pathways, creating a detrimental cycle of oxidative stress in DR. Additionally, hyperglycemia contributes to mitochondrial dysfunction and endoplasmic reticulum (ER) stress, which perpetuate oxidative stress in the retina. The high levels of oxidative stress result in increased lipid peroxidation, inflammation, and apoptosis of retinal cells, which are characteristic features of DR.</p>
Full article ">
54 pages, 5917 KiB  
Review
Oxidative Stress: A Suitable Therapeutic Target for Optic Nerve Diseases?
by Francesco Buonfiglio, Elsa Wilma Böhm, Norbert Pfeiffer and Adrian Gericke
Antioxidants 2023, 12(7), 1465; https://doi.org/10.3390/antiox12071465 - 20 Jul 2023
Cited by 12 | Viewed by 2967
Abstract
Optic nerve disorders encompass a wide spectrum of conditions characterized by the loss of retinal ganglion cells (RGCs) and subsequent degeneration of the optic nerve. The etiology of these disorders can vary significantly, but emerging research highlights the crucial role of oxidative stress, [...] Read more.
Optic nerve disorders encompass a wide spectrum of conditions characterized by the loss of retinal ganglion cells (RGCs) and subsequent degeneration of the optic nerve. The etiology of these disorders can vary significantly, but emerging research highlights the crucial role of oxidative stress, an imbalance in the redox status characterized by an excess of reactive oxygen species (ROS), in driving cell death through apoptosis, autophagy, and inflammation. This review provides an overview of ROS-related processes underlying four extensively studied optic nerve diseases: glaucoma, Leber’s hereditary optic neuropathy (LHON), anterior ischemic optic neuropathy (AION), and optic neuritis (ON). Furthermore, we present preclinical findings on antioxidants, with the objective of evaluating the potential therapeutic benefits of targeting oxidative stress in the treatment of optic neuropathies. Full article
(This article belongs to the Special Issue Oxidative-Stress in Human Diseases—2nd Edition)
Show Figures

Figure 1

Figure 1
<p>Prevalence (per 100,000) in some of the most frequent optic neuropathies. LHON: Leber’s hereditary optic neuropathy; ON: optic neuritis; NA-AION: nonarteritic anterior ischemic optic neuropathy; A-AION: arteritic anterior ischemic optic neuropathy; GCA: giant cell arteritis. * We used a <span class="html-italic">y</span>-axis break in consideration of the remarkably higher prevalence of glaucoma compared to all other optic nerve disorders. ** LHON prevalence in case of incomplete penetrance is meaningfully higher than in complete penetrance due to the high frequency of the variant mutant carriers.</p>
Full article ">Figure 2
<p>Anatomy and perfusion of the visual pathway. LGN: lateral geniculate nucleus.</p>
Full article ">Figure 3
<p>Model representing the ROS impact on the retina and on the optic nerve. ROS: reactive oxidative species; NOX2: NADPH oxidase type 2; XO: xanthine oxidase; eNOS: endothelial nitric oxide synthase; RGC: retinal ganglion cell; RNFL: retinal nerve fiber layer; RPE: retinal pigment epithelium; BM: Brunch’s membrane. Up arrows mean increase or upregulation.</p>
Full article ">Figure 4
<p>Anatomic overview of aqueous humor outflow, with focus on the drainage system through the trabecular meshwork and the Schlemm’s canal to the superficial veins.</p>
Full article ">Figure 5
<p>Model of etiopathogenesis in glaucomatous optic neuropathies. AH: aqueous humor; TM: trabecular meshwork; TMC: trabecular meshwork cell; ONH: optic nerve head; OPA1: optic atrophy 1 gene; TNF-α: tumor necrosis factor alpha; NF-kB: nuclear factor “kappa-light-chain-enhancer” of activated B-cells; ATP: adenosintriphosphat; RGC: retinal ganglion cell; oxLDL: oxidized low density lipoprotein; AGE: advanced glycation end product; pJNK: c-Jun N-terminal kinase; pERK: extracellular-signal-regulated kinase.</p>
Full article ">Figure 6
<p>Role of calcium and reactive oxygen species in the interplay between ER and mitochondria. ER: endoplasmic reticulum; PERK: protein kinase RNA-like ER kinase; ATF: activating transcription factor; IRE-1: inositol-requiring protein 1; CHOP: CCAAT-enhancer-binding protein homologous protein; eIF2α: eukaryotic initiation factor 2α; sXBP1: spliced X-box binding protein-1; TNFR-2: tumor necrosis factor alpha receptor 2; ASK-1: apoptosis signal-regulating kinase 1; JNK: c-Jun N-terminal kinase; ERAD: ER-associated degradation; NF-kB: nuclear factor “kappa-light-chain-enhancer” of activated B-cells. Up arrows mean increase or upregulation. Down arrows mean decrease.</p>
Full article ">Figure 7
<p>Direct and indirect effect of idebenone on the mitochondrial oxidative metabolism. COQ<sub>10</sub>: cofactor Q 10; Cyt c: cytochrome c; GSH: glutathione; SOD: superoxide dismutase; GPX: glutathione peroxidase; NQO1: NAD(P)H quinone oxidoreductase 1; NOX2: nicotinamide adenine dinucleotide phosphate oxidase 2; Nrf2: nuclear factor erythroid-derived 2-related factor 2.</p>
Full article ">Figure 8
<p>Model of LHON pathogenesis. NAD: nicotinamide dinucleotide; ETC: electron transport chain; ATP: adenosine triphosphate; cyt c: cytochrome c; Bid: BH3 interacting-domain death agonist; Apaf-1: apoptotic protease-activating factor 1; PTP: permeability transition pore.</p>
Full article ">
37 pages, 11146 KiB  
Review
Quinones as Neuroprotective Agents
by Ángel Cores, Noelia Carmona-Zafra, José Clerigué, Mercedes Villacampa and J. Carlos Menéndez
Antioxidants 2023, 12(7), 1464; https://doi.org/10.3390/antiox12071464 - 20 Jul 2023
Cited by 17 | Viewed by 4488
Abstract
Quinones can in principle be viewed as a double-edged sword in the treatment of neurodegenerative diseases, since they are often cytoprotective but can also be cytotoxic due to covalent and redox modification of biomolecules. Nevertheless, low doses of moderately electrophilic quinones are generally [...] Read more.
Quinones can in principle be viewed as a double-edged sword in the treatment of neurodegenerative diseases, since they are often cytoprotective but can also be cytotoxic due to covalent and redox modification of biomolecules. Nevertheless, low doses of moderately electrophilic quinones are generally cytoprotective, mainly due to their ability to activate the Keap1/Nrf2 pathway and thus induce the expression of detoxifying enzymes. Some natural quinones have relevant roles in important physiological processes. One of them is coenzyme Q10, which takes part in the oxidative phosphorylation processes involved in cell energy production, as a proton and electron carrier in the mitochondrial respiratory chain, and shows neuroprotective effects relevant to Alzheimer’s and Parkinson’s diseases. Additional neuroprotective quinones that can be regarded as coenzyme Q10 analogues are idobenone, mitoquinone and plastoquinone. Other endogenous quinones with neuroprotective activities include tocopherol-derived quinones, most notably vatiquinone, and vitamin K. A final group of non-endogenous quinones with neuroprotective activity is discussed, comprising embelin, APX-3330, cannabinoid-derived quinones, asterriquinones and other indolylquinones, pyrroloquinolinequinone and its analogues, geldanamycin and its analogues, rifampicin quinone, memoquin and a number of hybrid structures combining quinones with amino acids, cholinesterase inhibitors and non-steroidal anti-inflammatory drugs. Full article
(This article belongs to the Special Issue Oxidative Stress and Nrf2 in Neuroprotection)
Show Figures

Figure 1

Figure 1
<p>Main mechanisms of action of the quinones described in this review. Abbreviations: T/B cells: the two main types of lymphocytes. BBB: blood brain barrier. AChE: acetylcholinesterase. BChE: butyrylcholinesterase. AChR: acetylcholine receptor. MMP-9: matrix metalloproteinase 9. Aβ: beta-amyloid protein. BACE-1: beta-secretase 1. GSK-3β: glycogen synthase kinase-3 beta. HSP90: Heat-shock protein 90. NQO1: NAD(P)H:quinone oxidoreductase 1. ROS: reactive oxygen species. MAO: monoamino oxidase. PTPm: mitochondrial permeability transition pore. 15-LOX: 15-lipooxygenase. NOX: nitric oxide. DAMPs: damage-associated molecular patterns. TLR4: Toll-like receptor 4. NF-κB: Nuclear factor kappa-light-chain-enhancer of activated B cells. NRLP3: NLR family pyrin domain containing 3. JAK: Janus kinase. STAT: signal transducer and activator of transcription. TNF-α: tumor necrosis factor alpha. IL: interleukins.</p>
Full article ">Figure 2
<p>General features of quinone chemistry of relevance to neuroprotection. Quinones may act as electrophiles, thereby alkylating proteins or nucleic acids. Additionally, they can accept one electron to give semiquinones, which can in turn be reduced to hydroquinones by one-electron transfer and protonation. Semiquinone formation can be reverted by molecular oxygen, leading to the generation of superoxide anion-radicals.</p>
Full article ">Figure 3
<p>Toxic species derived from the oxidative metabolism of dopamine. Formation of dopamine-<span class="html-italic">o</span>-quinone (DAQ) from the one-electron or two-electron oxidation of dopamine and its evolution by intramolecular Michael additions and additional oxidation reactions.</p>
Full article ">Figure 4
<p>Mechanisms explaining the generation of reactive oxygen species from doxorubicin.</p>
Full article ">Figure 5
<p>Main molecular mechanisms involved in chemotherapy-induced cognitive impairment.</p>
Full article ">Figure 6
<p>Cytoxic vs. cytoprotective responses to quinones, depending on their concentration and electrophilicity.</p>
Full article ">Figure 7
<p>The Keap1/Nrf2 cytoprotective pathway. Basal state: Negative regulation of Nrf2 under normal conditions (“basal state”) and its activation under pathological conditions (“induced state” in the presence of reactive oxygen species (ROS) or covalent modifiers.</p>
Full article ">Figure 8
<p>Structure of coenzyme Q<sub>10</sub> and generation of its three oxidation states by two successive one-electron reductions.</p>
Full article ">Figure 9
<p>The role of coenzyme Q<sub>10</sub> in electron and proton transfer processes in the mitochondrial respiratory chain.</p>
Full article ">Figure 10
<p>The lipid peroxidation process and mechanisms that protect against it involving the interplay of α-tocopherol and coenzyme Q<sub>10</sub>.</p>
Full article ">Figure 11
<p>Structure of ubisol-Q<sub>10</sub>, a nanomicellar formulation combining coenzyme Q<sub>10</sub> with polyoxyethanyl α-tocopheryl sebacate (PTS).</p>
Full article ">Figure 12
<p>Structures of idebenone, its active metabolite, QS-10 and the related hydroquinone D1.</p>
Full article ">Figure 13
<p>Structures of decylubiquinone and its mitochondria-targeted phosphonium analogue mitoquinone mesylate.</p>
Full article ">Figure 14
<p>Structure of plastoquinone and some of its synthetic analogues, namely the phosphonium derivative SkQ,1 designed for mitochondrial penetration and retention, and the fluorescent plastoquinone-rhodamine hybrid SkQR1.</p>
Full article ">Figure 15
<p>Mechanism of the antioxidant activity of tocopherol in cell membranes and its metabolism to yield α-tocotrienol quinone (vatiquinone) and its α-tocotrienol hydroquinone.</p>
Full article ">Figure 16
<p>Structures of neuroprotective vitamin K-related quinones.</p>
Full article ">Figure 17
<p>Structures of embelin and its analogue, the embelin-donepezil hybrid SB-1448.</p>
Full article ">Figure 18
<p>(<b>A</b>) Activation of transcription factors by the APE1/Ref-1 protein and its biological consequences. (<b>B</b>) Structures of the APE1/Ref-1 inhibitors APX-3330, APX-2009 and APX-2014. Abbreviations: APE1/Ref1: Apurinic/apyrimidinic endonuclease/redox-factor 1. TF: transcription factor. HIF-1α: Hypoxia-inducible factor 1-alpha. NF-κB: Nuclear factor kappa-light-chain-enhancer of activated B cells. STAT3: Signal transducer and activator of transcription 3. Nrf2: nuclear factor erythroid 2–related factor 2.</p>
Full article ">Figure 19
<p>Stages in the design of neuroprotective cannabinoid-derived quinones.</p>
Full article ">Figure 20
<p>Neuroprotective asterriquinones and simpler indolylquinones.</p>
Full article ">Figure 21
<p>Biosynthetic origin of PQQ and its redox equilibria with PQQ semiquinone and hydroquinone.</p>
Full article ">Figure 22
<p>Products derived from the reaction of PQQ with amino acids such as free glutamate in the brain.</p>
Full article ">Figure 23
<p>Geldanamycin and its neuroprotective analogues.</p>
Full article ">Figure 24
<p>Structures of rifampicin and its quinone.</p>
Full article ">Figure 25
<p>Miscellaneous neuroprotective benzoquinones, naphthoquinones and anthraquinones.</p>
Full article ">Figure 26
<p>Structures of some neuroprotective heterocyclic quinones.</p>
Full article ">Figure 27
<p>Structure and pharmacological profile of memoquin. Structures of a half-molecule simplified memoquin analogue <b>7</b> and memoquin-lipoic acid hybrids <b>8</b>.</p>
Full article ">Figure 28
<p>Memoquin analogues that include fragments corresponding to non-steroidal anti-inflammatory drugs.</p>
Full article ">Figure 29
<p>Multitarget hybrid compounds with cholinesterase inhibitory activity.</p>
Full article ">Figure 30
<p>Selected naphthoquinone-tryptophan and naphthoquinone-dopamine hybrids.</p>
Full article ">Figure 31
<p>An indolequinone derivative acting as a bexarotene prodrug by a process initiated by NQO1 reduction of the quinone moiety.</p>
Full article ">
22 pages, 3242 KiB  
Article
Protective Effects of Polyphenol-Rich Extracts against Neurotoxicity Elicited by Paraquat or Rotenone in Cellular Models of Parkinson’s Disease
by Mitali A. Tambe, Aurélie de Rus Jacquet, Katherine E. Strathearn, Jennifer A. Hensel, Bryce D. Colón, Aswathy Chandran, Gad G. Yousef, Mary H. Grace, Mario G. Ferruzzi, Qingli Wu, James E. Simon, Mary Ann Lila and Jean-Christophe Rochet
Antioxidants 2023, 12(7), 1463; https://doi.org/10.3390/antiox12071463 - 20 Jul 2023
Cited by 5 | Viewed by 1717
Abstract
Parkinson’s disease (PD) is a neurodegenerative disorder involving motor symptoms caused by a loss of dopaminergic neurons in the substantia nigra region of the brain. Epidemiological evidence suggests that anthocyanin (ANC) intake is associated with a low risk of PD. Previously, we reported [...] Read more.
Parkinson’s disease (PD) is a neurodegenerative disorder involving motor symptoms caused by a loss of dopaminergic neurons in the substantia nigra region of the brain. Epidemiological evidence suggests that anthocyanin (ANC) intake is associated with a low risk of PD. Previously, we reported that extracts enriched with ANC and proanthocyanidins (PAC) suppressed dopaminergic neuron death elicited by the PD-related toxin rotenone in a primary midbrain culture model. Here, we characterized botanical extracts enriched with a mixed profile of polyphenols, as well as a set of purified polyphenolic standards, in terms of their ability to mitigate dopaminergic cell death in midbrain cultures exposed to another PD-related toxicant, paraquat (PQ), and we examined underlying neuroprotective mechanisms. Extracts prepared from blueberries, black currants, grape seeds, grape skin, mulberries, and plums, as well as several ANC, were found to rescue dopaminergic neuron loss in PQ-treated cultures. Comparison of a subset of ANC-rich extracts for the ability to mitigate neurotoxicity elicited by PQ versus rotenone revealed that a hibiscus or plum extract was only neuroprotective in cultures exposed to rotenone or PQ, respectively. Several extracts or compounds with the ability to protect against PQ neurotoxicity increased the activity of the antioxidant transcription factor Nrf2 in cultured astrocytes, and PQ-induced dopaminergic cell death was attenuated in Nrf2-expressing midbrain cultures. In other studies, we found that extracts prepared from hibiscus, grape skin, or purple basil (but not plums) rescued defects in O2 consumption in neuronal cells treated with rotenone. Collectively, these findings suggest that extracts enriched with certain combinations of ANC, PAC, stilbenes, and other polyphenols could potentially slow neurodegeneration in the brains of individuals exposed to PQ or rotenone by activating cellular antioxidant mechanisms and/or alleviating mitochondrial dysfunction. Full article
(This article belongs to the Special Issue Dietary Polyphenols and Neuroprotection II)
Show Figures

Figure 1

Figure 1
<p>Polyphenol-enriched extracts with different ANC profiles have different abilities to protect against PQ neurotoxicity. Primary midbrain cultures incubated in the absence or presence of extract prepared from BB (<b>A</b>); wild BB (<b>C</b>); BC (<b>D</b>); or PB (<b>E</b>) for 72 h were exposed to PQ (2.5 μM) in the absence or presence of extract for 24 h. Alternatively, cultures incubated in the absence or presence of a BB extract (0.1 μg/mL) for 66 h were incubated in fresh media (minus extract) for 6 h and then exposed to PQ (2.5 μM) in the absence of extract for 24 h (<b>B</b>). Control cells were incubated in the absence of PQ or extract. The cells were stained with antibodies specific for MAP2 and TH and scored for relative dopaminergic cell viability. The data are presented as the mean ± SEM; <span class="html-italic">n</span> = 4 (<b>A</b>,<b>D</b>,<b>E</b>), <span class="html-italic">n</span> = 5 (<b>B</b>), or <span class="html-italic">n</span> = 6 (<b>C</b>); * <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; 0.0001, square root transformation, one-way ANOVA with Tukey’s multiple comparisons post hoc test (ns, not significant). In panels (<b>B</b>,<b>D</b>,<b>E</b>), a statistically significant neuroprotective effect is observed for cultures treated with BB (<b>B</b>) or BC (1 μg/mL; (<b>D</b>)) plus PQ versus PQ alone, and a significant neurotoxic effect is observed for cultures treated in the presence versus the absence of PQ (<b>E</b>), when the square root-transformed data are analyzed via ANOVA with the Newman-Keuls post hoc test (<span class="html-italic">p</span> ≤ 0.05).</p>
Full article ">Figure 2
<p>Extracts enriched with PAC or PA protect against PQ neurotoxicity. Primary midbrain cultures incubated in the absence or presence of a GS extract (<b>A</b>); plum extract (<b>B</b>); or curcumin (<b>C</b>) for 72 h were exposed to PQ (2.5 μM) in the absence or presence of extract or compound for 24 h. Control cells were incubated in the absence of PQ, extract, or compound. The cells were stained with antibodies specific for MAP2 and TH and scored for relative dopaminergic cell viability. The data are presented as the mean ± SEM; <span class="html-italic">n</span> = 3 (<b>B</b>,<b>C</b>) or <span class="html-italic">n</span> = 5 (<b>A</b>); * <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, square root transformation, one-way ANOVA with Tukey’s multiple comparisons post hoc test (ns, not significant).</p>
Full article ">Figure 3
<p>Stilbene-rich extracts alleviate PQ neurotoxicity. Primary midbrain cultures incubated in the absence or presence of a mulberry extract (<b>A</b>); GSK extract (<b>B</b>); or resveratrol (10 nM) (<b>C</b>) for 72 h were exposed to PQ (2.5 μM) in the absence or presence of extract or compound for 24 h. Control cells were incubated in the absence of PQ, extract, or compound. The cells were stained with antibodies specific for MAP2 and TH and scored for relative dopaminergic cell viability. The data are presented as the mean ± SEM; <span class="html-italic">n</span> = 4 (<b>A</b>,<b>C</b>) or <span class="html-italic">n</span> = 5 (<b>B</b>); * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, square root transformation, one-way ANOVA with Tukey’s multiple comparisons post hoc test (ns, not significant).</p>
Full article ">Figure 4
<p>Individual ANC (but not a hibiscus extract) protect against PQ neurotoxicity. Primary midbrain cultures incubated in the absence or presence of C3G (<b>A</b>); D3G (<b>B</b>); M3G (<b>C</b>); C3So (<b>D</b>); or a hibiscus extract (<b>E</b>) for 72 h were exposed to PQ (2.5 μM) in the absence or presence of compound or extract for 24 h. Control cells were incubated in the absence of PQ, compound, or extract. The cells were stained with antibodies specific for MAP2 and TH and scored for relative dopaminergic cell viability. The data are presented as the mean ± SEM; <span class="html-italic">n</span> = 3 (<b>E</b>), <span class="html-italic">n</span> = 4 (<b>A</b>–<b>C</b>), or <span class="html-italic">n</span> = 5 (<b>D</b>): * <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, square root transformation, one-way ANOVA with Tukey’s multiple comparisons post hoc test (ns, not significant).</p>
Full article ">Figure 5
<p>A subset of botanical extracts and all of the tested ANC increase Nrf2 transcriptional activity. Primary cortical astrocytes transduced with an ARE-EGFP reporter adenovirus for 48 h were incubated in the absence or presence of BB extract (<b>A</b>); wild BB extract (<b>B</b>); GSK extract (<b>C</b>); C3G (<b>D</b>); D3G (<b>E</b>); or M3G (<b>F</b>) for 24 h. Control astrocytes were transduced with the reporter virus and incubated in the absence of extract or compound. The cells were imaged to determine the intracellular EGFP fluorescence intensity. The data are presented as the mean ± SEM (fold change relative to control); <span class="html-italic">n</span> = 4 (<b>B</b>,<b>C</b>) or <span class="html-italic">n</span> = 6 or 7 (<b>A</b>,<b>D</b>,<b>E</b>,<b>F</b>); * <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 versus a predicted ratio of 1, log transformation followed by one-sample <span class="html-italic">t</span>-test (ns, not significant).</p>
Full article ">Figure 6
<p>Nrf2 over-expression is sufficient for protection against PQ neurotoxicity. Primary midbrain cultures (untransduced or transduced with adenovirus encoding untagged mouse Nrf2) were incubated with PQ (2.5 μM). Control cells were incubated in the absence of virus or PQ. The cells were stained with antibodies specific for MAP2 and TH and scored for relative dopaminergic cell viability. The data are presented as the mean ± SEM; <span class="html-italic">n</span> = 3; * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001, square root transformation, one-way ANOVA with Tukey’s multiple comparisons post hoc test.</p>
Full article ">Figure 7
<p>A subset of botanical extracts alleviate rotenone-induced deficits in mitochondrial respiration. Galactose-conditioned N27 cells were incubated in the absence or presence of an extract prepared from PB (<b>A</b>); plums (<b>B</b>); GSK (<b>C</b>) or hibiscus (<b>D</b>) for 21 h and then exposed to rotenone (50 nM) in the absence or presence of extract for 3 h. Control cell were incubated in the absence of rotenone or extract. Oxygen consumption was measured with a Clark-type oxygen electrode attached to a voltmeter. The data are presented as the mean ± SEM; <span class="html-italic">n</span> = 3 (<b>A</b>,<b>B</b>,<b>D</b>) or <span class="html-italic">n</span> = 4 (<b>C</b>); * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, log transformation, one-way ANOVA with Tukey’s multiple comparisons post hoc test.</p>
Full article ">
22 pages, 6944 KiB  
Article
High-Intensity Exercise Promotes Deleterious Cardiovascular Remodeling in a High-Cardiovascular-Risk Model: A Role for Oxidative Stress
by Aline Meza-Ramos, Anna Alcarraz, Marta Lazo-Rodriguez, Gemma Sangüesa, Elisenda Banon-Maneus, Jordi Rovira, Maria Jose Ramirez-Bajo, Marta Sitges, Lluís Mont, Pedro Ventura-Aguiar, Montserrat Batlle and Eduard Guasch
Antioxidants 2023, 12(7), 1462; https://doi.org/10.3390/antiox12071462 - 20 Jul 2023
Viewed by 1650
Abstract
Although the benefits of moderate exercise in patients at high cardiovascular risk are well established, the effects of strenuous exercise remain unknown. We aimed to study the impact of strenuous exercise in a very high cardiovascular risk model. Nephrectomized aged Zucker obese rats [...] Read more.
Although the benefits of moderate exercise in patients at high cardiovascular risk are well established, the effects of strenuous exercise remain unknown. We aimed to study the impact of strenuous exercise in a very high cardiovascular risk model. Nephrectomized aged Zucker obese rats were trained at a moderate (MOD) or high (INT) intensity or were kept sedentary (SED) for 10 weeks. Subsequently, echocardiography and ex vivo vascular reactivity assays were performed, and blood, aortas, perivascular adipose tissue (PVAT), and left ventricles (LVs) were harvested. An improved risk profile consisting of decreased body weight and improved response to a glucose tolerance test was noted in the trained groups. Vascular reactivity experiments in the descending thoracic aorta demonstrated increased endothelial NO release in the MOD group but not in the INT group, compared with SED; the free radical scavenger TEMPOL improved endothelial function in INT rats to a similar level as MOD. An imbalance in the expression of oxidative stress-related genes toward a pro-oxidant environment was observed in the PVAT of INT rats. In the heart, INT training promoted eccentric hypertrophy and a mild reduction in ejection fraction. Obesity was associated with LV fibrosis and a transition toward β-myosin heavy chain and the N2Ba titin isoform. Exercise reverted the myosin imbalance, but only MOD reduced the predominance of the N2Ba titin isoform. In conclusion, moderate exercise yields the most intense cardiovascular benefits in a high-cardiovascular-risk animal model, while intense training partially reverts them. Full article
(This article belongs to the Special Issue Exercise-Induced Antioxidant Response and Oxidative Stress)
Show Figures

Figure 1

Figure 1
<p>Impact of different loads of regular training on cardiovascular risk factors. (<b>A</b>) Weekly measurement (estimated marginal mean ± standard error of the mean {SEM}) of body weight. Analysis with linear mixed modeling with a repeated measures covariance structure, including week, group, and its interaction as predictors. The time point × group interaction was significant at the <span class="html-italic">p</span> &lt; 0.001 level; within each week, significant post hoc pairwise comparisons between groups (LSD tests) are shown. N = 10 per group. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 INT vs. Sed (<b>B</b>) Indexed body weight (BW) before euthanasia. Analysis with a linear trend analysis. * <span class="html-italic">p</span> &lt; 0.05. (<b>C</b>) Results (estimated marginal mean ± SEM) of the intraperitoneal glucose tolerance test. Analysis with linear mixed modeling with a repeated measures covariance structure, including time point, group, and its interaction as predictors. The time point × group interaction was significant at the <span class="html-italic">p</span> &lt; 0.001 level; within each time point, significant post hoc pairwise comparisons between groups (LSD tests) are shown. N = 10 per group. ** <span class="html-italic">p</span> &lt; 0.01 SED vs MOD; + <span class="html-italic">p</span> &lt; 0.05 SED vs INT. (<b>D</b>) Area under the curve (AUC, mean ± SD) of glucose blood concentrations during the glucose test. Analysis was performed with a one-way ANOVA which showed a significant omnibus test at the <span class="html-italic">p</span> &lt; 0.05 level. Significant pairwise LSD tests are shown. * <span class="html-italic">p</span> &lt; 0.05. (<b>E</b>) Serum insulin concentration (mean ± SD) before and 30 min after glucose administration. Analysis with linear mixed modeling with a repeated measures covariance structure, including time point (i.e., baseline and 30 min) and group and its interaction as predictors. The interaction time point × group was significant at the <span class="html-italic">p</span> &lt; 0.05 level; within each group, significant post hoc pairwise comparisons between time points (baseline vs. 30-min, LSD tests) are shown. ** <span class="html-italic">p</span> &lt; 0.01. (<b>F</b>) Triglyceride plasma concentration (mean ± SD) in all groups. Analysis was performed with a one-way ANOVA. SED, MOD, and INT (obese sedentary, moderately and intensively trained rats, respectively).</p>
Full article ">Figure 2
<p>Echocardiographic changes produced by exercise. (<b>A</b>) Ascending aortic diameter (mean ± SD). Analysis with a linear trend analysis. (<b>B</b>) Representative M-mode images of a parasternal long-axis view. The segmented blue line identifies the interventricular septum (IVS) and the green arrow identifies the left ventricular diastolic diameters (LVDd). (<b>C</b>–<b>H</b>) The mean ± SD is shown for indexed IVS (iIVS) (<b>C</b>), indexed LVDd (iLVDd) (<b>D</b>), indexed left ventricle posterior wall (iLVPW) (<b>E</b>), left ventricle ejection fraction (<b>F</b>) and fractional shortening (<b>G</b>), and transmitral E/A ratio (<b>H</b>). All analyses were performed with a one-way ANOVA which showed a significant omnibus test at the <span class="html-italic">p</span> &lt; 0.05 level for iVS, iLVDd, ejection fraction, and fractional shortening. Post hoc pairwise comparisons were performed with the LSD tests. * <span class="html-italic">p</span> &lt; 0.05. SED, MOD, and INT (obese sedentary, moderately and intensively trained rats, respectively).</p>
Full article ">Figure 3
<p>Aortic collagen deposition and turnover. (<b>A</b>) Percentage of collagen deposit in the tunica media from picrosirius red-stained thoracic aorta (mean ± SD). (<b>B</b>–<b>D</b>) Aortic mRNA expression (mean ± SD) of alpha-1 procollagen types 1 (<b>B</b>) and 3 (<b>C</b>), and matrix metalloproteinase-2. Analyses were performed with one-way ANOVA which showed a significant omnibus test at the <span class="html-italic">p</span> &lt; 0.0001 level for gene expression parameters. Post hoc pairwise comparisons (<span class="html-italic">t</span>-tests) were FDR-adjusted. * <span class="html-italic">p</span> &lt; 0.05; **** <span class="html-italic">p</span> &lt; 0.0001. SED, MOD, and INT (obese sedentary, moderately, and intensively trained rats, respectively), CTL (young lean rats).</p>
Full article ">Figure 4
<p>Relaxation response in vascular reactivity experiments in descending thoracic aorta. Dose-response relaxation curves induced by carbachol (CCH) alone (<b>A</b>) and in the presence of nitric oxide inhibitor LNMMA (<b>B</b>), sodium nitroprusside (SNP) in the presence of L-NMMA (<b>C</b>), and carbachol in the presence of the free radical scavenger TEMPOL (<b>D</b>). Shaded areas of all curves represent 95% CI. The estimated logEC50 (mean ± SEM) for each group and curve is shown in the inset. Analyses were performed with a one-way ANOVA per each individual graph, comparing fitted logEC50 in three parameter equations. Significant post hoc pairwise FDR-adjusted comparisons between groups are shown, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001. SED, MOD, and INT (obese sedentary, moderately, and intensively trained rats, respectively), CTL (young lean rats). For all experiments: n = 9 (SED), n = 10 (MOD), n = 10 (INT, except for panel 4C in which n = 9), and n = 8 (CTL).</p>
Full article ">Figure 5
<p>Contractile response in vascular reactivity experiments in descending thoracic aorta. Dose-response contraction curves induced by phenylephrine (PHE) alone (<b>A</b>) and in the presence of the nitric oxide inhibitor L-NMMA (<b>B</b>) or the free radical scavenger TEMPOL (<b>C</b>). Shaded areas of all curves represent 95% CI. Estimated LogEC50 (mean ± SEM) for each group and curve is shown. Analyses were performed with a one-way ANOVA per each individual graph, comparing fitted logEC50 in three parameter equations. Significant post hoc pairwise FDR-adjusted comparisons between groups are shown, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, and **** <span class="html-italic">p</span> &lt; 0.0001. SED, MOD, and INT (obese sedentary, moderately, and intensively trained rats, respectively), CTL (young lean rats). For all experiments: n = 6 (SED, except for panel (<b>C</b>) in which n = 5), n = 11 (MOD), n = 9 (INT), and n = 8 (CTL).</p>
Full article ">Figure 6
<p>Oxidative stress imbalance in aortic and perivascular adipose tissue. (<b>A</b>) Aortic mRNA expression (mean ± SD) of pro-oxidant markers. (<b>B</b>) Aortic mRNA expression (mean ± SD) of antioxidant markers. (<b>C</b>) Representative images of Nrf2 nuclei translocation in the aortic thoracic wall (upper panel; nuclei stained in blue {DAPI}, Nrf2 in red, when translocated to the nuclei appears pink/purple), and percentage (mean ± SD) of positive nuclei in the tunica media (lower panel). (<b>D</b>) Perivascular adipose tissue (PVAT) mRNA expression of pro-oxidant markers (mean ± SD). (<b>E</b>) PVAT mRNA expression of antioxidant markers (mean ± SD). Analyses were performed at the gene level with a one-way ANOVA, significant post hoc pairwise comparisons between groups (FDR adjustment for (<b>A</b>,<b>B</b>), LSD tests for (<b>D</b>,<b>E</b>) are shown, * <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. SED, MOD, and INT (obese sedentary, moderately and intensively trained rats, respectively), CTL (young lean rats).</p>
Full article ">Figure 7
<p>Left ventricular myocardial histological assessment. (<b>A</b>) Representative triple immunofluorescence images of myocardial triple immunofluorescence (wheat germ agglutinin in red for extracellular matrix, vimentin in blue for fibroblasts, and isolectin-GS IB4 in green for capillaries). Results (mean ± SD) for the left ventricular (LV) myocyte cross-sectional area (CSA) in the LV free wall and the interventricular septum (IVS) ((<b>B</b>,<b>C</b>), respectively), extracellular matrix content in the LV free wall and the IVS (<b>D</b>,<b>E</b>), vimentin-positive area in the LV free wall and the IVS (<b>F</b>,<b>G</b>), and capillary density in the LV free wall and the IVS (<b>H</b>,<b>I</b>). Analyses were performed with a one-way ANOVA, and significant post hoc pairwise FDR-adjusted comparisons between groups are shown, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001. SED, MOD, and INT (obese sedentary, moderately, and intensively trained rats, respectively), CTL (young lean rats).</p>
Full article ">Figure 8
<p>Expression of genes involved in cardiac remodeling in all groups. Interventricular septum mRNA expression (mean ± SD) of the α- and β-myosin heavy chain isoforms (Mhc) (<b>A</b>) and their ratio (<b>B</b>), titin isoforms N2b and N2ba (<b>C</b>), and their ratio (<b>D</b>), Bnp (<b>E</b>), Igfr1 (<b>F</b>), and Mef2d (<b>G</b>). Analyses were performed at the gene level with a one-way ANOVA, significant post hoc pairwise FDR-adjusted comparisons between groups are shown, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, and **** <span class="html-italic">p</span> &lt; 0.0001. SED, MOD, and INT (obese sedentary, moderately, and intensively trained rats, respectively), CTL (young lean rats).</p>
Full article ">Figure 9
<p>Myocardial collagen deposition. (<b>A</b>) Representative images of picrosirius red-stained samples (<b>left panel</b>) and quantification of myocardial fibrosis (mean ± SD, <b>right panel</b>) in the left ventricle free wall (LVFW), interventricular septum (IVS), and right ventricle free wall (RVFW). Analysis was performed with a linear mixed effects modeling, including group, chamber, and their interaction as predictors; both group and cardiac chamber were significant at the <span class="html-italic">p</span> &lt; 0.001 level, and significant post hoc pairwise comparisons within the group main factor are shown (<span class="html-italic">p</span>-values are FDR-adjusted). (<b>B</b>–<b>E</b>) mRNA expression (mean ± SD) of alpha-1 procollagen types 1 (<b>B</b>) and 3 (<b>C</b>), matrix metalloproteinase-2 (<b>D</b>), and tissular metalloproteinase inhibitor 1 (<b>E</b>). Analyses were performed at the gene level with a one-way ANOVA, and significant post hoc pairwise FDR-adjusted comparisons between groups are shown. *** <span class="html-italic">p</span> &lt; 0.001, and **** <span class="html-italic">p</span> &lt; 0.0001. SED, MOD, and INT (obese sedentary, moderately, and intensively trained rats, respectively), CTL (young lean rats).</p>
Full article ">
20 pages, 4195 KiB  
Article
Pepper Fruit Extracts Show Anti-Proliferative Activity against Tumor Cells Altering Their NADPH-Generating Dehydrogenase and Catalase Profiles
by Marta Rodríguez-Ruiz, María C. Ramos, María J. Campos, Inmaculada Díaz-Sánchez, Bastien Cautain, Thomas A. Mackenzie, Francisca Vicente, Francisco J. Corpas and José M. Palma
Antioxidants 2023, 12(7), 1461; https://doi.org/10.3390/antiox12071461 - 20 Jul 2023
Cited by 2 | Viewed by 2585
Abstract
Cancer is considered one of the main causes of human death worldwide, being characterized by an alteration of the oxidative metabolism. Many natural compounds from plant origin with anti-tumor attributes have been described. Among them, capsaicin, which is the molecule responsible for the [...] Read more.
Cancer is considered one of the main causes of human death worldwide, being characterized by an alteration of the oxidative metabolism. Many natural compounds from plant origin with anti-tumor attributes have been described. Among them, capsaicin, which is the molecule responsible for the pungency in hot pepper fruits, has been reported to show antioxidant, anti-inflammatory, and analgesic activities, as well as anti-proliferative properties against cancer. Thus, in this work, the potential anti-proliferative activity of pepper (Capsicum annuum L.) fruits from diverse varieties with different capsaicin contents (California < Piquillo < Padrón < Alegría riojana) against several tumor cell lines (lung, melanoma, hepatoma, colon, breast, pancreas, and prostate) has been investigated. The results showed that the capsaicin content in pepper fruits did not correspond with their anti-proliferative activity against tumor cell lines. By contrast, the greatest activity was promoted by the pepper tissues which contained the lowest capsaicin amount. This indicates that other compounds different from capsaicin have this anti-tumor potentiality in pepper fruits. Based on this, green fruits from the Alegría riojana variety, which has negligible capsaicin levels, was used to study the effect on the oxidative and redox metabolism of tumor cell lines from liver (Hep-G2) and pancreas (MIA PaCa-2). Different parameters from both lines treated with crude pepper fruit extracts were determined including protein nitration and protein S-nitrosation (two post-translational modifications (PTMs) promoted by nitric oxide), the antioxidant capacity, as well as the activity of the antioxidant enzymes superoxide dismutase (SOD), catalase, and glutathione peroxidase (GPX), among others. In addition, the activity of the NADPH-generating enzymes glucose-6-phosphate dehydrogenase (G6PDH), 6-phosphogluconate dehydrogenase (6PGDH), and NADP-isocitrate dehydrogenase (NADP-ICDH) was followed. Our data revealed that the treatment of both cell lines with pepper fruit extracts altered their antioxidant capacity, enhanced their catalase activity, and considerably reduced the activity of the NADPH-generating enzymes. As a consequence, less H2O2 and NADPH seem to be available to cells, thus avoiding cell proliferation and possibly triggering cell death in both cell lines. Full article
Show Figures

Figure 1

Figure 1
<p>Plant materials and pepper varieties used in this work, and their capsaicin content. (<b>A</b>) Different parts of a representative pepper fruit. (<b>B</b>) Phenotype of fruits from the four varieties at two ripening stages: immature green and ripe red. Whereas California-Melchor is a sweet pepper fruit type, Piquillo Padrón and Alegría riojana contain different capsaicin levels with the sequence Piquillo &lt;&lt;&lt; Padrón &lt; Alegría riojana. (<b>C</b>) Content of total capsaicin levels in placenta and pericarp from fruits of the four pepper varieties at two ripening stages. Placenta tissue was processed once seeds were discarded. Data are the means ± SEM of three replicates determined from five fruits of the four varieties and at the two ripening stages. Different letters after each value indicate that differences were statistically significant (one-way Anova and Tukey test, <span class="html-italic">p</span> &lt; 0.05). FW, fresh weight. Figure designed from data provided in Palma et al. [<a href="#B51-antioxidants-12-01461" class="html-bibr">51</a>].</p>
Full article ">Figure 2
<p>Antiproliferative activity and capsaicin content of crude extracts from fruits of four pepper varieties against seven tumor cell lines. The antiproliferative activity is expressed as % of dead cells after the treatment, and capsaicin content as μg/g fresh weight (FW), determined in the samples of pepper fruits used in the assays. Plant materials assayed in this experiment and their corresponding codes include pericarp (PE) and placenta (PL) from green (g) and red (r) pepper fruits from the varieties California (C), Piquillo (Pi), Padrón (P), and Alegría riojana (AR). The cell lines used corresponded to tumors from lung (A549), melanoma (A2058), hepatoma (Hep-G2), colon (HT-29), breast (MCF-7), pancreas (MIA PaCa-2), and prostate (PC-3). The plot shown is representative of three independent experiments. Color code of each graph is provided in the legend on the right.</p>
Full article ">Figure 3
<p>IC50 and level of the tumor marker ERK 1/2 from Hep-G2 and MIA PaCa-2 tumor cell lines after the incubation with crude pepper fruit extracts from the Alegría riojana variety. IC50 was defined as the amount of pepper fruit that reduced by 50% the cell viability. (<b>A</b>) IC50 in Hep-G2 cells. (<b>B</b>) IC50 in MIA PaCa-2 cells. The plots shown are representative of the three independent experiments performed. Each symbol at the assayed concentrations, either triangles, squares or circles, represents a replicate. Grey symbols denote outlier values once the regression curve is adjusted to all data. Square brackets in the two plots denote the IC50 for each cell line.</p>
Full article ">Figure 4
<p>Level of the tumor marker ERK 1/2 from Hep-G2 and MIA PaCa-2 tumor cell lines after the incubation with crude pepper fruit extracts from the Alegría riojana variety. (<b>A</b>) Detection of the tumor marker ERK 1/2 (related protein-serine/threonine kinases 1 and 2) in Hep-G2 and MIA PaCa-2 cells that were untreated (C) and treated (T) with crude extracts from pepper fruits. Polypeptides were separated by SDS-PAGE in 4–20% precast polyacrylamide gels, and blotting assays were performed using a monoclonal antibody against ERK1/2 proteins (dilution 1:1000). Molecular weight markers are indicated on the left. (<b>B</b>) Densitograms of the ERK 1/2 bands detected after the blotting assay, where arbitrary units (in parentheses) were assigned to each tumor marker band by the use of the program ImageJ. The blotting image shown is representative of the three independent experiments performed.</p>
Full article ">Figure 5
<p>Detection of tyrosine (NO<sub>2</sub>-Tyr, panel (<b>A</b>)) and tryptophan (NO<sub>2</sub>-Trp, panel (<b>B</b>)) nitrated polypeptides from Hep-G2 and MIA PaCa-2 tumor cell lines after the incubation with crude pepper fruit extracts from the Alegría riojana variety. Polypeptides were separated by SDS-PAGE in 4–20% precast polyacrylamide gels, and blotting assays were performed using a polyclonal antibody against nitro-tyrosine (NO<sub>2</sub>-Tyr, dilution 1:500), and a monoclonal antibody against nitro-tryptophan (NO<sub>2</sub>-Trp, dilution 1:1000). C, untreated cells. T, treated cells. PHY, <span class="underline">P</span>olypeptide from <span class="underline">H</span>ep-G2 cells with nitrated Tyr (<span class="underline">Y</span>). PMY, <span class="underline">P</span>olypeptide from <span class="underline">M</span>IA PaCa-2 cells with nitrated Tyr (<span class="underline">Y</span>). PHW, <span class="underline">P</span>olypeptide from <span class="underline">H</span>ep-G2 cells with nitrated Trp (<span class="underline">W</span>). PMT, Polypeptide from MIA PaCa-2 cells with nitrated Trp (<span class="underline">W</span>). Molecular weight markers (Mw) are indicated on both sides of the panel. The blotting shown is representative of three independent experiments.</p>
Full article ">Figure 6
<p>Detection of S-nitrosated (SNO-Cys, panel (<b>A</b>)) and glutathionylated (panel (<b>B</b>)) polypeptides from Hep-G2 and MIA PaCa-2 tumor cell lines after the incubation with crude pepper fruit extracts from the Alegría riojana variety. Polypeptides were separated by SDS-PAGE in 4–20% precast polyacrylamide gels and blotting assays were performed using a polyclonal antibody against S-nitrosocysteine (SNO-Cys, dilution 1:500), and a polyclonal antibody against glutathione (anti-GSH, dilution 1:1000). C, untreated cells. T, treated cells. PHC, <span class="underline">P</span>olypeptide from <span class="underline">H</span>ep-G2 cells with nitrosated <span class="underline">C</span>ys (C). PMC, <span class="underline">P</span>olypeptide from <span class="underline">M</span>IA PaCa-2 cells with nitrated nitrosated <span class="underline">C</span>ys (C). PHG, <span class="underline">P</span>olypeptide from <span class="underline">H</span>ep-G2 cells <span class="underline">G</span>lutathionylated. PMG, <span class="underline">P</span>olypeptide from <span class="underline">M</span>IA PaCa-2 cells <span class="underline">G</span>lutathionylated. Molecular weight markers (Mw) are indicated on both sides of the panel. The blotting shown is representative of the three independent experiments.</p>
Full article ">Figure 7
<p>Total antioxidant activity and catalase activity from Hep-G2 and MIA PaCa-2 tumor cell lines after the incubation with crude pepper fruit extracts from the Alegría riojana variety. (<b>A</b>) Total antioxidant activity expressed as ascorbate equivalents. (<b>B</b>) Catalase activity. Data are expressed as the means of at least nine measurements from three independent experiments ± SEM. Asterisks denote significant differences in comparisons of treated cells to untreated (control) ones at <span class="html-italic">p</span> &lt; 0.05 (Student <span class="html-italic">t</span>-test).</p>
Full article ">Figure 8
<p>Superoxide dismutase (SOD) isoenzymes from Hep-G2 and MIA PaCa-2 tumor cell lines after the incubation with crude pepper fruit extracts from the Alegría riojana variety. Proteins (20 μg, see <a href="#sec2dot4-antioxidants-12-01461" class="html-sec">Section 2.4</a>) were separated using non-denaturing PAGE in 10% polyacrylamide gels, and activity was detected in gels through the specific NBT staining method. C, untreated cells. T, cells treated with pepper fruit crude extracts. The zymogram shown is representative of the three experiments performed.</p>
Full article ">Figure 9
<p>Activity of NADPH-generating enzymes from Hep-G2 and MIA PaCa-2 tumor cell lines after the incubation with crude pepper fruit extracts from the Alegría riojana variety. (<b>A</b>) Glucose-6-phosphate dehydrogenase (G6PDH). (<b>B</b>) 6-Phosphogluconate dehydrogenase (6PGDH). (<b>C</b>) NADP-dependent isocitrate dehydrogenase (NADP-ICDH). Data are expressed as the means of at least nine measurements from three independent experiments ± SEM. Asterisks denote significant differences in comparisons of treated cells to untreated (control) ones at a <span class="html-italic">p</span> &lt; 0.05 (Student <span class="html-italic">t</span>-test).</p>
Full article ">Figure 10
<p>Proposed model of the effect of crude extracts from pepper fruits on the antioxidant and redox metabolism of tumor cells. As a consequence of the treatment with pepper extracts, an increase of catalase activity and a decline of the NADPH-generation enzymes occurs, thus provoking lower H<sub>2</sub>O<sub>2</sub> levels and limited NADPH availability. Under these circumstances, cell proliferation is arrested and cell death may be triggered. CAT, catalase. SOD, superoxide dismutase. GPX, glutathione peroxidase. GSH, reduced glutathione. GSSG, oxidized glutathione. GR, glutathione reductase. G6PDH, glucose-6-phosphate dehydrogenase. 6PGDH, 6-phosphogluconate dehydrogensase. ICDH, isocitrate dehydrogenase. G6P, glucose-6-phosphate. 6PG, 6-phosphogluconate. Ru5P, ribulose-5-phosphate. αKG, α-ketoglutarate. In blue ink are the enzymatic systems studied in this work.</p>
Full article ">
27 pages, 4088 KiB  
Review
Alzheimer’s Disease and Green Tea: Epigallocatechin-3-Gallate as a Modulator of Inflammation and Oxidative Stress
by Víctor Valverde-Salazar, Daniel Ruiz-Gabarre and Vega García-Escudero
Antioxidants 2023, 12(7), 1460; https://doi.org/10.3390/antiox12071460 - 20 Jul 2023
Cited by 11 | Viewed by 3745
Abstract
Alzheimer’s disease (AD) is the most common cause of dementia, characterised by a marked decline of both memory and cognition, along with pathophysiological hallmarks including amyloid beta peptide (Aβ) accumulation, tau protein hyperphosphorylation, neuronal loss and inflammation in the brain. Additionally, oxidative stress [...] Read more.
Alzheimer’s disease (AD) is the most common cause of dementia, characterised by a marked decline of both memory and cognition, along with pathophysiological hallmarks including amyloid beta peptide (Aβ) accumulation, tau protein hyperphosphorylation, neuronal loss and inflammation in the brain. Additionally, oxidative stress caused by an imbalance between free radicals and antioxidants is considered one of the main risk factors for AD, since it can result in protein, lipid and nucleic acid damage and exacerbate Aβ and tau pathology. To date, there is a lack of successful pharmacological approaches to cure or even ameliorate the terrible impact of this disease. Due to this, dietary compounds with antioxidative and anti-inflammatory properties acquire special relevance as potential therapeutic agents. In this context, green tea, and its main bioactive compound, epigallocatechin-3-gallate (EGCG), have been targeted as a plausible option for the modulation of AD. Specifically, EGCG acts as an antioxidant by regulating inflammatory processes involved in neurodegeneration such as ferroptosis and microglia-induced cytotoxicity and by inducing signalling pathways related to neuronal survival. Furthermore, it reduces tau hyperphosphorylation and aggregation and promotes the non-amyloidogenic route of APP processing, thus preventing the formation of Aβ and its subsequent accumulation. Taken together, these results suggest that EGCG may be a suitable candidate in the search for potential therapeutic compounds for neurodegenerative disorders involving inflammation and oxidative stress, including Alzheimer’s disease. Full article
(This article belongs to the Special Issue Oxidative Stress in Alzheimer's Disease)
Show Figures

Figure 1

Figure 1
<p>Amyloid β and tau pathology in Alzheimer’s disease. The main hallmarks of an AD brain are the accumulation of extracellular amyloid-β senile plates (1), the accumulation of intracellular neurofibrillary tangles of tau (2) and neuronal death (3). The areas in which these three features coexist are called neuritic plates (4). Amyloid precursor protein (APP) processing can take place through the amyloidogenic pathway (A), being cleaved by β-secretase (BACE), generating the sAPPβ and CTFβ fragments. The latter is subsequently processed by γ-secretase, generating Aβ42 and Aβ40, which induce amyloid β pathology in an AD brain (5) that ultimately affects neuronal homeostasis and induces cell death (6). On the contrary, in the non-amyloidogenic pathway (B), APP is cleaved by α- and γ-secretases, generating CTFα and sAPPα fragments that exert a neuroprotective function because a diminished APP conversion to Aβ and sAPPα inhibits BACE (7). On the other hand, tau is a microtubule-associated protein that establishes their structure. Post-translational modifications of tau, including phosphorylation, induce the detachment of tau and microtubule destabilization (8) and lead to tau aggregation (9). The accumulation of tau protein causes a failure of neuronal homeostasis whose final consequence is neuronal death (10).</p>
Full article ">Figure 2
<p>Oxidative stress in Alzheimer’s disease. There is an increase in oxidative stress in an AD brain, which induces hyperphosphorylated tau (1) and amyloid-β (2) accumulation, mitochondrial dysfunction (3) and inflammation (4). At the same time, these processes also induce oxidative stress, generating a vicious cycle. Mitochondrial dysfunction is related to Aβ accumulation and, at the same time, Aβ is able to impair mitochondrial function (5). The accumulation of Aβ also induces tau hyperphosphorylation (6), which provokes mitochondrial dysfunction (7). Either Aβ (8) or tau (9) accumulation induce inflammation, which, in the same way, favours the accumulation of Aβ and tau. The accumulation of metals is one of the main causes of ROS increasement (10) in an AD brain and it also favours Aβ pathology (11) and inflammation processes (12). All these features together induce the loss of neuronal connections and the reduction in neurons shown in an AD brain (13). Ferroptosis is increased in an AD brain due to increased levels of iron, ferririn and transferrin and diminished ferroporter and GPX4 (14) that causes neuron necroptotic death and subsequent inflammation (15). EGCG is able to block oxidative stress (16) and, thanks to its chelating activity, it counteracts the accumulation of heavy metals (17) and ferroptosis (18), protecting the brain from all these harmful effects.</p>
Full article ">Figure 3
<p>Antioxidant activity of EGCG. The antioxidant compound EGCG binds to antioxidant regulatory elements (ARE), inducing the expression of stress response genes (1) such as haem oxygenase (2) and glutathione-S-transferase (3) enzymes, counteracting oxidative stress processes (4). Additionally, EGCG enhances the activity of SOD and catalase (5), which reduce oxidative stress (4). Thanks to its hydroxyl groups, EGCG harbours chelating properties (6) that exert antioxidant activity, promoting neuroprotective effects (7). Also, EGCG can inhibit the production of ROS/RNS and 3-Hydroxykynurenine effect due to this antioxidant capacity (8), avoiding oxidative stress (9).</p>
Full article ">Figure 4
<p>Effect of metal chelation mediated by EGCG in Alzheimer’s pathology. In an AD brain, iron accumulation promotes amyloidogenic processing (1), increasing Aβ40/Aβ42 levels (2), leading to the accumulation of Aβ (3). Moreover, brain iron accumulation produces an increase in tau hyperphosphorylation (4) that results in microtubule destabilization and the accumulation of tau protein (5). As a consequence, iron produces neurodegeneration (6). The metal chelator activity of EGCG (6) inhibits iron accumulation in the brain (7), therefore diminishing the accumulation of Aβ (8) and tau protein (9). Moreover, EGCG can directly potentiate the expression of p21 (10) and p27 (11), while diminishing the expression of cyclin D1 (12) and pRB (13), abolishing cell cycle re-entry (14). On the other hand, EGCG can promote the activation of HIF-1α (15), inducing the expression of cell survival genes (16). EGCG promotes the production of SAPPα (17) and the non-amyloidogenic processing of APP (18), generating neuroprotection effects (19).</p>
Full article ">Figure 5
<p>Modulation of PKC-mediated pathways by EGCG in Alzheimer’s disease. EGCG can induce the activation of PKC pathways (1), specifically the isoform PKCζ (2) that enhances long-term memory (3), PKCα (4) that results in an increase in memory function (5) and PKCε (6) that activates BDNF factor (7), promoting synaptogenesis (8), neuronal survival (9), Ca2+ liberation (10) and changes of synaptic structures (11), altogether promoting neuroprotection (12). The generation of Aβ produces Aβ plaques (13), promoting neurodegeneration processes (14). Aβ also inhibits the PKC pathway (15) and RACK (16), whose receptors are required to activate PKC (17), and blocks BDNF (18). Moreover, the isoforms PKCα and PKCε activate α-secretase (19), promoting the generation of sAPPα (20) that inhibits Aβ production (21). PKCε activates ECE1 (22), which degrades Aβ (21). The activation of the PKC pathway inhibits GSK3β (23), which is involved in tau hyperphosphorylation (24) that finally leads to neurodegeneration processes (14). EGCG produces the reduction in protein Bax (25) and promotes the degradation of protein Bad (26) through the proteasomal system (27).</p>
Full article ">Figure 6
<p>Modulation of MAPK pathway by EGCG in Alzheimer’s disease. Upon oxidative stress, EGCG induces antioxidant defences with the activation of the Keap1/Nrf2/ARE pathway (1) and increases ERK1/2 (2), promoting cell survival (3) and neuroprotective effects (4). Conversely, EGCG inhibits the ERK (5), p38 (6) and JNK (7) pathways whose effects contribute to mitochondrial disfunction and altered dynamics (8), induce tau protein hyperphosphorylation and aggregation (9) and increase apoptosis (10) and inflammation (11), leading to cell death (12). Therefore, EGCG can avoid this cell death (13).</p>
Full article ">Figure 7
<p>Modulation of PI3K/Akt pathway mediated by EGCG. Akt/protein kinase B (PKB) is activated by Phosphatidyl Inositol 3 Kinase (PI3K) (1), which catalyses the conversion of phosphatidyl inositol (4,5) biphosphate (PIP2) into phosphatidyl inositol (3,4,5) triphosphate (PIP3), a process that is reversed by phosphatidylinositol (3,4,5)-triphosphate 3-phosphatase (PTEM) (2). PIP3 also recruits phosphoinositide-dependent kinase 1 (PDK1) to the plasma membrane, activating Akt (3). Akt phosphorylates glycogen synthase kinase 3 (GSK3), inhibiting its function (4). Active GSK3 is able to phosphorylate Mcl-1 (5), which is targeted for proteasomal degradation (6), liberating Bax and Bak proapoptotic factors (7). This causes the permeabilization of the mitochondrial outer membrane, releasing cytochrome c (Cyt C) (8) that attaches to Apaf-1, generating the apoptosome (9), leading to the activation of caspase 3 (10) that induces apoptosis (11). GSK3 also phosphorylates tau, inducing its aggregation and subsequent neurodegeneration (12). EGCG can ultimately increase PI3K and Akt activity (13) and inhibit PTEM in the presence of ROS (14), inhibiting apoptosis (15), phospho-Tau aggregation (16) and, therefore, generating neuroprotection.</p>
Full article ">
16 pages, 21608 KiB  
Article
Pharmacological and Genetic Suppression of VDAC1 Alleviates the Development of Mitochondrial Dysfunction in Endothelial and Fibroblast Cell Cultures upon Hyperglycemic Conditions
by Konstantin N. Belosludtsev, Dmitriy A. Serov, Anna I. Ilzorkina, Vlada S. Starinets, Mikhail V. Dubinin, Eugeny Yu. Talanov, Maxim N. Karagyaur, Alexandra L. Primak and Natalia V. Belosludtseva
Antioxidants 2023, 12(7), 1459; https://doi.org/10.3390/antiox12071459 - 20 Jul 2023
Cited by 3 | Viewed by 2011
Abstract
Prolonged hyperglycemia related to diabetes and its complications leads to multiple cellular disorders, the central one being the dysfunction of mitochondria. Voltage-dependent anion channels (VDAC) of the outer mitochondrial membrane control the metabolic, ionic, and energy cross-talk between mitochondria and the rest of [...] Read more.
Prolonged hyperglycemia related to diabetes and its complications leads to multiple cellular disorders, the central one being the dysfunction of mitochondria. Voltage-dependent anion channels (VDAC) of the outer mitochondrial membrane control the metabolic, ionic, and energy cross-talk between mitochondria and the rest of the cell and serve as the master regulators of mitochondrial functions. Here, we have investigated the effect of pharmacological suppression of VDAC1 by the newly developed inhibitor of its oligomerization, VBIT-4, in the primary culture of mouse lung endotheliocytes and downregulated expression of VDAC1 in human skin fibroblasts on the progression of mitochondrial dysfunction upon hyperglycemic stress. The cells were grown in high-glucose media (30 mM) for 36 h. In response to hyperglycemia, the mRNA level of VDAC1 increased in endotheliocytes and decreased in human skin fibroblasts. Hyperglycemia induced overproduction of mitochondrial ROS, an increase in the susceptibility of the organelles to mitochondrial permeability transition (MPT) pore opening and a drop in mitochondrial membrane potential, which was accompanied by a decrease in cell viability in both cultures. Treatment of endotheliocytes with 5 µM VBIT-4 abolished the hyperglycemia-induced increase in susceptibility to spontaneous opening of the MPT pore and ROS generation in mitochondria. Silencing of VDAC1 expression in human skin fibroblasts exposed to high glucose led to a less pronounced manifestation of all the signs of damage to mitochondria. Our data identify a mitochondria-related response to pharmacological and genetic suppression of VDAC activity in vascular cells in hyperglycemia and suggest the potential therapeutic value of targeting these channels for the treatment of diabetic vasculopathies. Full article
(This article belongs to the Special Issue Oxidative Stress in Metabolic Disease)
Show Figures

Figure 1

Figure 1
<p><span class="html-italic">Vdac1</span> mRNA level in primary mouse microvascular endothelial cells (EC) and human skin fibroblasts (HF) under normal (5 mM) and high (30 mM) glucose conditions. Means ± SD are shown (<span class="html-italic">n</span> = 4–5).</p>
Full article ">Figure 2
<p>Effect of VBIT-4 (5 µM) on the viability of mouse microvascular endothelial cells under normoglycemia (5 mM glucose) and high (30 mM) glucose conditions. Means ± SD are shown (<span class="html-italic">n</span> = 4).</p>
Full article ">Figure 3
<p>Effect of VBIT-4 (5 µM) on the mitochondrial membrane potential (Δψ) (<b>A</b>) and production of reactive oxygen species (<b>B</b>) in mouse microvascular endothelial cells under normo- (5 mM glucose) and hyperglycemia (30 mM glucose). Means ± SD are shown (<span class="html-italic">n</span> = 3–5).</p>
Full article ">Figure 4
<p>MPT pore opening in mouse microvascular endothelial cells. (<b>A</b>) Representative fluorescence images of calcein-AM in the presence of CoCl<sub>2</sub> in endothelial cells of the experimental groups. Scale bar—25 μm. (<b>B</b>) Intensity of calcein-AM fluorescence signals in mitochondria of the microvascular endothelial cells from four experimental groups. Means ± SD are shown (<span class="html-italic">n</span> = 4).</p>
Full article ">Figure 5
<p>mRNA level (<b>A</b>) and amount of VDAC1 protein (<b>B</b>) in cells with normal (WT) and decreased (VDAC1-/-) expression of VDAC1. (<b>C</b>) Survival of fibroblasts with normal and reduced expression of VDAC1 under normal (5 mM) and high (30 mM) glucose conditions (<b>C</b>). Means ± SD are shown (<span class="html-italic">n</span> = 4–5).</p>
Full article ">Figure 6
<p>Changes in the mitochondrial membrane potential (Δψ) (<b>A</b>) and production of reactive oxygen species (<b>B</b>) in human skin fibroblasts with normal (WT) and reduced (VDAC1-/-) expression of VDAC1 under conditions of normo- (5 mM glucose) and hyperglycemia (30 mM glucose). Means ± SD are shown (<span class="html-italic">n</span> = 3–5).</p>
Full article ">Figure 7
<p>MPT pore opening in human skin fibroblasts with normal (WT) and reduced (VDAC1-/-) expression of VDAC1. (<b>A</b>) Typical fluorescence images of mitochondrial calcein in the presence of CoCl<sub>2</sub> in fibroblasts of the experimental groups. Scale bar—25 μm. (<b>B</b>) Intensity of calcein fluorescence in fibroblasts mitochondria from four experimental groups. Means ± SD are shown (<span class="html-italic">n</span> = 4).</p>
Full article ">Figure 8
<p>The relative mRNA levels of <span class="html-italic">Drp1</span> (<b>A</b>), <span class="html-italic">Mfn2</span> (<b>B</b>), <span class="html-italic">Pink1</span> (<b>C</b>), <span class="html-italic">Parkin</span> (<b>D</b>), and <span class="html-italic">Ppargc1a</span> (<b>E</b>) in the human skin fibroblasts in the experimental groups. Means ± SD are shown (<span class="html-italic">n</span> = 5).</p>
Full article ">
11 pages, 1175 KiB  
Article
Beneficial Effects of Olive Oil Enriched with Lycopene on the Plasma Antioxidant and Anti-Inflammatory Profile of Hypercholesterolemic Patients
by Jesus Roman Martínez Álvarez, Ana Belen Lopez Jaen, Monica Cavia-Saiz, Pilar Muñiz and Victoria Valls-Belles
Antioxidants 2023, 12(7), 1458; https://doi.org/10.3390/antiox12071458 - 20 Jul 2023
Cited by 1 | Viewed by 1806
Abstract
Olive oil and lycopene are foods that have potent antioxidant activity. The objective was to determine the effects of consumption of olive oil enriched with lycopene on oxidative stress biomarkers in hypercholesterolemic subjects. We examined the effects of oil enriched with lycopene extract [...] Read more.
Olive oil and lycopene are foods that have potent antioxidant activity. The objective was to determine the effects of consumption of olive oil enriched with lycopene on oxidative stress biomarkers in hypercholesterolemic subjects. We examined the effects of oil enriched with lycopene extract daily intake during 1 month on plasma antioxidant capacity, lipids profile (triacylgycerols, total cholesterol, cHDL; cLDL, ox-LDL), biomarkers of oxidative stress, and inflammatory markers related with atherosclerosis risk (C-reactive protein (CRP), IL-6; sDC4L) in subjects hypercholesteremics (cholesterol > 220 mg/dL). In the group consuming olive oil-lycopene, significant increases (p < 0.05) in the levels of plasma lycopene concentration (0.146 ± 0.03 versus 0.202 ± 0.04 (µmol/L)), α-carotene (0.166 ± 0.064 versus 0.238 ± 0.07) and in β-carotene (0.493 ± 0.187 versus 0.713 ± 0.221) were observed. These results are linked with the increases of plasma antioxidants and decreases biomarkers of oxidative stress (carbonyl groups, malondialdehyde and 8-hydroxy-deoxiguanosine) observed in hypercholesterolemic group. In relation to lipid profile, a significant decrease was observed in the levels of ox-LDL (781 ± 302 versus 494 ± 200), remaining unchanged the levels of TG, cholesterol, HDL and LDL-c. Regarding inflammatory biomarkers, the levels of CRP and IL-6 decreased significantly. The positive results obtained in this study support the use of olive oil enriched with lycopene to reduce the risk of coronary disease. Full article
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>)—Lycopene, α-carotene, β-carotene and cryptoxanthin levels in the plasma of patients with hypercholesterolemia, at the basal time (initial value) and after 30 days of diet supplementation with lycopene-olive oil (<b>A</b>) and olive oil (<b>B</b>). Values are means ± SD. Different alphabetical letters indicate significant differences (<span class="html-italic">p</span> &lt; 0.005). * Indicates that significant different (<span class="html-italic">p</span> &lt; 0.05) between initial value and 1 month.</p>
Full article ">Figure 2
<p>Blood lipid profile levels in plasma of patients with hypercholesterolemic, at basal time (initial value) and after 30 days of diet supplementation with lycopene-olive oil (<b>A</b>) and olive oil (<b>B</b>). TG = triglycerides; HDL = high-density lipoprotein; LDL: low-density lipoprotein; LDLox: LDL oxidized low-density protein. Values are means ± SD. * Indicate that after 1 month the means are significantly different (<span class="html-italic">p</span> &lt; 0.05) when compared to initial time values.</p>
Full article ">Figure 3
<p>Biomarkers of oxidative stress (MDA, Carbonyl groups, and 8OHdG) levels in plasma of patients with hypercholesterolemic, at basal time (initial value) and after 1 month of diet supplementation with lycopene-olive oil and olive oil. Values are means ± SD. * Indicate that means values after 1 month are significantly different (<span class="html-italic">p</span> &lt; 0.05) when compared to initial time baseline.</p>
Full article ">Figure 3 Cont.
<p>Biomarkers of oxidative stress (MDA, Carbonyl groups, and 8OHdG) levels in plasma of patients with hypercholesterolemic, at basal time (initial value) and after 1 month of diet supplementation with lycopene-olive oil and olive oil. Values are means ± SD. * Indicate that means values after 1 month are significantly different (<span class="html-italic">p</span> &lt; 0.05) when compared to initial time baseline.</p>
Full article ">
19 pages, 568 KiB  
Article
Contribution of Saccharomyces and Non-Saccharomyces Yeasts on the Volatile and Phenolic Profiles of Rosehip Mead
by Alexandra-Costina Avîrvarei, Carmen Rodica Pop, Elena Mudura, Floricuța Ranga, Simona-Codruța Hegheș, Emese Gal, Haifeng Zhao, Anca Corina Fărcaș, Maria Simona Chiș and Teodora Emilia Coldea
Antioxidants 2023, 12(7), 1457; https://doi.org/10.3390/antiox12071457 - 19 Jul 2023
Cited by 4 | Viewed by 1670
Abstract
The resurgence of mead, a honey-based fermented beverage, is attributed to the increasing consumption of fermented foods and beverages, driven by its distinct flavors and perceived health benefits. This study investigates the influence of different yeast strains, namely Saccharomyces cerevisiae var. bayanus, [...] Read more.
The resurgence of mead, a honey-based fermented beverage, is attributed to the increasing consumption of fermented foods and beverages, driven by its distinct flavors and perceived health benefits. This study investigates the influence of different yeast strains, namely Saccharomyces cerevisiae var. bayanus, and Torulaspora delbrueckii, on the volatile and phenolic compounds of these beverages. Analytical techniques, including HPLC-DAD and GS/MS, were employed to analyze the chemical composition of the beverages. ANOVA analysis of variance was conducted to assess differences in the volatile and phenolic compounds. The findings reveal that yeast selection significantly impacts the chemical profiles of the beverages. Saccharomyces cerevisiae fermentation preserves rosehip-specific flavonoids and phenolic acids. Sequential fermentation with Torulaspora delbrueckii demonstrated proficiency in generating esters, contributing to fruity and floral aromas in the beverages. This study investigates the importance of yeast selection in shaping the chemical composition of rosehip mead, providing insights into the distinct characteristics conferred by different yeast strains. By optimizing yeast selection and fermentation techniques, the overall quality and diversity of these beverages can be enhanced. Full article
(This article belongs to the Special Issue Plant Materials and Their Antioxidant Potential)
Show Figures

Figure 1

Figure 1
<p>Mead production experimental design and process flow.</p>
Full article ">
15 pages, 1071 KiB  
Review
Apples and Apple By-Products: Antioxidant Properties and Food Applications
by Umme Asma, Ksenia Morozova, Giovanna Ferrentino and Matteo Scampicchio
Antioxidants 2023, 12(7), 1456; https://doi.org/10.3390/antiox12071456 - 19 Jul 2023
Cited by 14 | Viewed by 6478
Abstract
In recent years, there has been a growing interest in utilizing natural antioxidants as alternatives to synthetic additives in food products. Apples and apple by-products have gained attention as a potential source of natural antioxidants due to their rich phenolic content. However, the [...] Read more.
In recent years, there has been a growing interest in utilizing natural antioxidants as alternatives to synthetic additives in food products. Apples and apple by-products have gained attention as a potential source of natural antioxidants due to their rich phenolic content. However, the extraction techniques applied for the recovery of phenolic compounds need to be chosen carefully. Studies show that ultrasound-assisted extraction is the most promising technique. High yields of phenolic compounds with antioxidant properties have been obtained by applying ultrasound on both apples and their by-products. Promising results have also been reported for green technologies such as supercritical fluid extraction, especially when a co-solvent is used. Once extracted, recent studies also indicate the feasibility of using these compounds in food products and packaging materials. The present review aims to provide a comprehensive overview of the antioxidant properties of apples and apple by-products, their extraction techniques, and potential applications in food products because of their antioxidant or nutritional properties. The findings reported here highlight the proper utilization of apples and their by-products in food to reduce the detrimental effect on the environment and provide a positive impact on the economy. Full article
(This article belongs to the Section Extraction and Industrial Applications of Antioxidants)
Show Figures

Figure 1

Figure 1
<p>The worldwide production of apples from 2012 to 2020 (<b>a</b>) and top 11 producers of apples in 2020 (<b>b</b>) according to data provided by the Food and Agriculture Organization of the United Nations.</p>
Full article ">Figure 2
<p>Apple by-product distribution.</p>
Full article ">Figure 3
<p>Apple and apple by-product phenolic compounds.</p>
Full article ">Figure 4
<p>Application of apple phenolic compounds in different foods.</p>
Full article ">
18 pages, 3194 KiB  
Article
Effect of Brewing Conditions on Antioxidant Properties of Ginkgo biloba Leaves Infusion
by Patrycja Biernacka, Katarzyna Felisiak, Iwona Adamska, Marek Śnieg and Cezary Podsiadło
Antioxidants 2023, 12(7), 1455; https://doi.org/10.3390/antiox12071455 - 19 Jul 2023
Cited by 1 | Viewed by 1670
Abstract
Due to the growing awareness of the importance of healthy eating in society, there is an increasing interest in the use of herbs and low-processed, natural products. Ginkgo biloba is a raw material with a high pro-health potential, which is related to the [...] Read more.
Due to the growing awareness of the importance of healthy eating in society, there is an increasing interest in the use of herbs and low-processed, natural products. Ginkgo biloba is a raw material with a high pro-health potential, which is related to the high content of antioxidant compounds. The aim of the study was to determine the relationship between the antioxidant activity of Ginkgo biloba leaf infusions and the weighted amount of leaves and brewing time. In addition, a sensory analysis of the infusions obtained was carried out. The innovation is to determine the migration of micro- and macroelements to the infusion prepared from Ginkgo biloba depending on the leaves’ weight used and the brewing time. The research showed the dependence of the antioxidant activity of the infusions and the migration of microelements on the size of the dried material and the brewing time. In the publication, the main factors influencing the quality of infusions were analysed, their mutual correlations were determined, and combinations showing the highest antioxidant activity and, at the same time, the highest sensory acceptability were selected. Full article
(This article belongs to the Special Issue Phenolics as Antioxidant Agents II)
Show Figures

Figure 1

Figure 1
<p>Changes in the pH of various weights in relation to the length of brewing the infusion.</p>
Full article ">Figure 2
<p>Changes in the acidity [g/L] of various weights in relation to the length of brewing the infusion.</p>
Full article ">Figure 3
<p>PCA biplot based on the first two principal component axes for antioxidant activity, acidity and soluble dry matter (<b>A</b>) and distribution of the 20 test samples based on the first two components derived from principal constituent analysis (<b>B</b>).</p>
Full article ">Figure 4
<p>Hierarchical method of agglomeration of clusters of the tested samples in relation to the antioxidant activity (total polyphenol content of the water (W) and methanol (M) fractions, DPPH, FRAP of the water (W) and methanol (M) fractions and TEAC of the water (W) and methanol (M) fractions). Cluster analysis is expressed in Euclidean distance [binding distance/maximum distance × 100].</p>
Full article ">Figure 5
<p>HPLC profile of infusions from dried <span class="html-italic">Ginkgo biloba</span> leaves in selected samples ((<b>A</b>)—samples with 2.5 g weight—S, brewing time 10, 15 and 60 min; (<b>B</b>)—samples with 5.0 g weight—M, brewing time 10, 15 and 60 min; (<b>C</b>)—samples with 7.5 g weight—L, brewing time 10, 15 and 60 min). All samples were determined—(1) chlorogenic acid; (2) epicatechin; (3) catechin; (4) coumarin; and (5) apigenin—with detection at 280 nm.</p>
Full article ">Figure 6
<p>Comparing colour, smell, flavour and overall acceptability of samples X—the smallest weight (<b>A</b>), S—small weight (<b>B</b>), M—medium weight (<b>C</b>) and L—large weight (<b>D</b>).</p>
Full article ">Figure 7
<p>Sensory profile of smell and flavour of samples X—the smallest weight (<b>A</b>), S—small weight (<b>B</b>), M—medium weight (<b>C</b>) and L—large weight (<b>D</b>) for attributes such as herbal, acrid, sweet, bitter and sour. For the clarity of the graphs, the scale has been limited to six.</p>
Full article ">
22 pages, 3622 KiB  
Article
Interactions between Angiotensin Type-1 Antagonists, Statins, and ROCK Inhibitors in a Rat Model of L-DOPA-Induced Dyskinesia
by Andrea Lopez-Lopez, Rita Valenzuela, Ana Isabel Rodriguez-Perez, María J. Guerra, Jose Luis Labandeira-Garcia and Ana Muñoz
Antioxidants 2023, 12(7), 1454; https://doi.org/10.3390/antiox12071454 - 19 Jul 2023
Cited by 3 | Viewed by 1803
Abstract
Statins have been proposed for L-DOPA-induced dyskinesia (LID) treatment. Statin anti-dyskinetic effects were related to the inhibition of the Ras-ERK pathway. However, the mechanisms responsible for the anti-LID effect are unclear. Changes in cholesterol homeostasis and oxidative stress- and inflammation-related mechanisms such as [...] Read more.
Statins have been proposed for L-DOPA-induced dyskinesia (LID) treatment. Statin anti-dyskinetic effects were related to the inhibition of the Ras-ERK pathway. However, the mechanisms responsible for the anti-LID effect are unclear. Changes in cholesterol homeostasis and oxidative stress- and inflammation-related mechanisms such as angiotensin II and Rho-kinase (ROCK) inhibition may be involved. The nigra and striatum of dyskinetic rats showed increased levels of cholesterol, ROCK, and the inflammatory marker IL-1β, which were reduced by the angiotensin type-1 receptor (AT1) antagonist candesartan, simvastatin, and the ROCK inhibitor fasudil. As observed for LID, angiotensin II-induced, via AT1, increased levels of cholesterol and ROCK in the rat nigra and striatum. In cultured dopaminergic neurons, angiotensin II increased cholesterol biosynthesis and cholesterol efflux without changes in cholesterol uptake. In astrocytes, angiotensin induced an increase in cholesterol uptake, decrease in biosynthesis, and no change in cholesterol efflux, suggesting a neuronal accumulation of cholesterol that is reduced via transfer to astrocytes. Our data suggest mutual interactions between angiotensin/AT1, cholesterol, and ROCK pathways in LID, which are attenuated by the corresponding inhibitors. Interestingly, these three drugs have also been suggested as neuroprotective treatments against Parkinson’s disease. Therefore, they may reduce dyskinesia and the progression of the disease using common mechanisms. Full article
Show Figures

Figure 1

Figure 1
<p>Experimental design of the in vivo experiments. Groups of rats and mice and time-course of experiments: lesions, behavioral tests, treatments, and sample analysis. Abbreviations: Amph, amphetamine; Ang II, angiotensin II; MFB, medial forebrain bundle; TH, tyrosine hydroxylase.</p>
Full article ">Figure 2
<p>A maximal lesion (i.e., lack of TH-immunoreactivity) can be observed by immunohistochemistry in the right substantia nigra (<b>A</b>) and right striatum (<b>B</b>) relative to the non-lesioned side (left side) or through Western blot analysis comparing the control side and the lesioned side (6-OHDA); (<b>E</b>) Nigral areas squared in A were magnified in (<b>C</b>,<b>D</b>). Abbreviations: SN, substantia nigra compacta; ST, striatum; TH, tyrosine hydroxylase. Scale bars: 600 µm (<b>A</b>,<b>B</b>); 100 µm (<b>C</b>,<b>D</b>).</p>
Full article ">Figure 3
<p>Effect of simvastatin on the development of L-DOPA-induced dyskinesia. Rats co-treated with the HMGCR inhibitor, simvastatin (15 mg/kg, orally), and L-DOPA (6 mg/kg; white circles) for 3 weeks showed a significant reduction in the development of AIMs relative to control rats treated with vehicle and L-DOPA (dark circles). This decrease was observed in the AIMs total score (<b>A</b>) and in the different components: axial (<b>B</b>), limb (<b>C</b>), and orolingual (<b>D</b>). Total AIMs score (<b>A</b>) was estimated as the addition of limb (<b>C</b>), orolingual (<b>D</b>), and axial (<b>B</b>) components. Values are expressed as means ± SEM. Two-way ANOVA for repeated measures and Holm–Sidak post hoc test, *, <span class="html-italic">p</span> &lt; 0.05. AIMs, abnormal involuntary movements.</p>
Full article ">Figure 4
<p>Effect of simvastatin on the therapeutic effect of L-DOPA assessed via the cylinder test and turning behavior (<b>A</b>,<b>B</b>) and the effect of simvastatin and the co-treatment of simvastatin and the ROCK inhibitor fasudil in L-DOPA-primed rats (<b>C</b>). (<b>A</b>) Spontaneous forelimb use in the cylinder test revealed that simvastatin administration did not reduce the ability of L-DOPA to improve forelimb akinesia, showing a similar number of touches performed with the left paw in animals treated with simvastatin and L-DOPA (LD + simv; violet bars) than in those treated with vehicle and L-DOPA (LD; green bars). (<b>B</b>) Total contralateral turns in 90 min showed that simvastatin did not reduce contralateral rotation. (<b>C</b>) In rats with previously established L-DOPA-induced dyskinesia (L-DOPA-primed rats, treated with L-DOPA 6 mg/kg for 3 weeks), treatment with oral simvastatin (15 mg/kg; white circles) and L-DOPA did not induce any significant difference in AIMs score with respect to control rats treated with vehicle and L-DOPA (dark circles). Co-treatment with simvastatin and fasudil (10 or 20 mg/kg) did not induce any significant improvement in dyskinetic behavior. In A and B, data are means ± SEM. * <span class="html-italic">p</span> &lt; 0.05, significant differences relative to baseline levels, off L-DOPA, Student <span class="html-italic">t</span>-test. In (<b>C</b>), two-way ANOVA for repeated measures and Holm–Sidak post hoc test, * <span class="html-italic">p</span> &lt; 0.05. AIMs, abnormal involuntary movements.</p>
Full article ">Figure 5
<p>Effect of L-DOPA-induced dyskinesia and simvastatin on HMGCR, ROCK, and interleukin-1β. Dyskinetic rats chronically treated with L-DOPA 6 mg/kg (6-OHDA + LD, green bars) showed a significant increase in cholesterol levels (<b>A</b>,<b>F</b>) and in HMGCR protein levels (<b>B</b>,<b>G</b>), relative to 6-OHDA-lesioned rats (6-OHDA, orange bars) in the substantia nigra (<b>A</b>,<b>B</b>) and striatum (<b>F</b>,<b>G</b>). Moreover, dyskinetic animals showed a significant increase in ROCK protein levels (<b>C</b>,<b>H</b>), ROCK activity (<b>D</b>,<b>I</b>), and interleukin-1β (IL-1β) protein expression (<b>E</b>,<b>J</b>) in both regions. The inhibition of cholesterol biosynthesis by simvastatin (6-OHDA + LD + simv, blue bars) reduces dyskinesia, ROCK levels (<b>C</b>,<b>H</b>), ROCK activity (<b>D</b>,<b>I</b>), and IL-1β levels (<b>E</b>,<b>J</b>) in the striatum and substantia nigra. Data are means ± SEM. The results were normalized to the values of 6-OHDA-lesioned rats treated with vehicle. * <span class="html-italic">p</span> &lt; 0.05, significant differences relative to 6-OHDA-lesioned rats; # <span class="html-italic">p</span> &lt; 0.05 significant differences relative to L-DOPA-treated rats. One-way ANOVA and Holm–Sidak post hoc tests were used.</p>
Full article ">Figure 6
<p>Effects of ROCK inhibitors and AT1 receptor blockers in the cholesterol biosynthesis marker HMGCR. The increase in HMGCR protein levels induced by dyskinesia (6-OHDA + LD, green bars) is reduced via the co-administration of L-DOPA and the ROCK inhibitor fasudil, (6-OHDA + LD + fasudil, blue bars) in the substantia nigra (<b>A</b>) and the striatum (<b>C</b>). Co-treatment with L-DOPA and the AT1 receptor antagonist candesartan (6-OHDA + LD + candesartan, blue bars) also produces a significant reduction in HMGCR protein levels induced through dyskinesia in the substantia nigra (<b>B</b>) and the striatum (<b>D</b>). The results were normalized to the values of 6-OHDA-lesioned animals treated with saline. Data are mean ± standard error of the mean (SEM); * <span class="html-italic">p</span> &lt; 0.05, significant differences relative to 6-OHDA-lesioned rats; <span>$</span> <span class="html-italic">p</span> &lt; 0.05 significant differences relative to L-DOPA-treated rats. One-way ANOVA and Holm–Sidak post hoc test were used.</p>
Full article ">Figure 7
<p>Effects of angiotensin, AT1 inhibition or AT1 or AT2 receptor deletion, and ROCK inhibition on cholesterol levels. Rats treated with an intraventricular injection of Angiotensin II (Ang II, pink bars) showed significantly higher levels of cholesterol in both substantia nigra (<b>A</b>) and striatum (<b>B</b>) compared to control saline-injected rats (green bars). The administration of the AT1 receptor antagonist candesartan (orange bars) induced a significant reduction in cholesterol levels in both regions (<b>C</b>,<b>D</b>). Moreover, AT1 KO mice (AT1KO, purple bars) showed significantly lower cholesterol levels than WT mice in the substantia nigra (<b>E</b>) and striatum (<b>F</b>), while AT2 KO mice (AT2KO, violet bars) showed significantly higher cholesterol content than in WT mice (<b>G</b>,<b>H</b>) in both regions. ROCK inhibition by fasudil (yellow bars) also induced a significant reduction in cholesterol levels in the nigra (<b>I</b>) and striatum (<b>J</b>). The results were normalized to the values of control animals treated with saline or WT mice. Data are mean ± standard error of the mean (SEM). * <span class="html-italic">p</span> &lt; 0.05 compared to control (Student’s <span class="html-italic">t</span>-test).</p>
Full article ">Figure 8
<p>Effects of angiotensin, AT1 inhibition, or AT1 or AT2 receptor deletion on ROCK expression. Rats treated with intraventricular angiotensin II (Ang II, pink bars) showed a significant increase in ROCK protein expression levels in the substantia nigra (<b>A</b>) and striatum (<b>B</b>) relative to control rats injected with saline (green bars). However, treatment with the AT1 antagonist candesartan (orange bars) showed a significant reduction in ROCK levels in the substantia nigra (<b>C</b>) and the striatum (<b>D</b>). Consistent with this, AT1 KO mice (AT1KO, purple bars) showed a significant decrease in ROCK levels in comparison with control WT mice in both regions (<b>E</b>,<b>F</b>). The results were normalized to the values of control animals treated with saline or WT mice. Data are mean ± standard error of the mean (SEM). * <span class="html-italic">p</span> &lt; 0.05 compared to control (Student’s <span class="html-italic">t</span>-test).</p>
Full article ">Figure 9
<p>Effect of angiotensin treatment on N27 dopaminergic neuron culture. The treatment of N27 dopaminergic cell line with angiotensin II (Ang II) for 24 h produced an increase in the mRNA (<b>A</b>) and protein expression (<b>D</b>) of cholesterol biosynthesis enzyme HMGCR, an increase in cholesterol in efflux-related molecule ABCA1 (<b>B</b>,<b>E</b>), and no changes in cholesterol uptake-related molecule LDL receptor (LDLR) expression (<b>C</b>,<b>F</b>). Data are mean ± standard error of the mean (SEM). * <span class="html-italic">p</span> &lt; 0.05 compared to control (Student’s <span class="html-italic">t</span>-test). HMGCR, 3-hydroxy-3-methylglutaryl CoA reductase; ABCA1, ATP-binding cassette transporter-1.</p>
Full article ">Figure 10
<p>Effect of angiotensin treatment on C6 astroglial culture. Treatment of C6 astroglial cell line with angiotensin II (Ang II) for 24 h produced an increase in mRNA (<b>A</b>) and protein (<b>D</b>) cholesterol uptake-related molecule LDL receptor (LDLR) expression, a decrease in cholesterol biosynthesis enzyme HMGCR (<b>B</b>,<b>E</b>), and no changes in cholesterol efflux-related molecule ABCA1 (<b>C</b>,<b>F</b>). Data are mean ± standard error of the mean (SEM). * <span class="html-italic">p</span> &lt; 0.05 compared to control (Student’s <span class="html-italic">t</span>-test). HMGCR, 3-hydroxy-3-methylglutaryl CoA reductase; ABCA1, ATP-binding cassette transporter-1.</p>
Full article ">
34 pages, 2091 KiB  
Review
A Review of the Changes Produced by Extrusion Cooking on the Bioactive Compounds from Vegetal Sources
by Silvia Mironeasa, Ionica Coţovanu, Costel Mironeasa and Mădălina Ungureanu-Iuga
Antioxidants 2023, 12(7), 1453; https://doi.org/10.3390/antiox12071453 - 19 Jul 2023
Cited by 6 | Viewed by 2010
Abstract
The demand for healthy ready-to-eat foods like snacks is increasing. Physical modification of vegetal food matrices through extrusion generates significant changes in the chemical composition of the final product. There is a great variety of food matrices that can be used in extrusion, [...] Read more.
The demand for healthy ready-to-eat foods like snacks is increasing. Physical modification of vegetal food matrices through extrusion generates significant changes in the chemical composition of the final product. There is a great variety of food matrices that can be used in extrusion, most of them being based on cereals, legumes, fruits, vegetables, or seeds. The aim of this review was to summarize the main effects of the extrusion process on the bioactive compounds content, namely phenolics, terpenes, vitamins, minerals, and fibers of vegetal mixes, as well as on their biological activity. The literature reported contradictory results regarding the changes in bioactive compounds after extrusion, mainly due to the differences in the processing conditions, chemical composition, physicochemical properties, and nutritional value of the extruded material and quantification methods. The thermolabile phenolics and vitamins were negatively affected by extrusion, while the fiber content was proved to be enhanced. Further research is needed regarding the interactions between bioactive components during extrusion, as well as a more detailed analysis of the impact of extrusion on the terpenes since there are few papers dealing with this aspect. Full article
(This article belongs to the Special Issue Impact of Processing on Antioxidant Rich Foods - 2nd Edition)
Show Figures

Figure 1

Figure 1
<p>Extrusion processing of food ingredients, reprinted with permission from Ek and Ganjyal [<a href="#B15-antioxidants-12-01453" class="html-bibr">15</a>], published by Elsevier in 2020, copyright year 2023.</p>
Full article ">Figure 2
<p>Structures of flavonoid, phenolic acids, and other non-flavonoid phenolic compounds reprinted with permission from de la Rosa et al. [<a href="#B23-antioxidants-12-01453" class="html-bibr">23</a>], published by Elsevier in 2019, copyright year 2023.</p>
Full article ">Figure 3
<p>Structures of some terpenes found in vegetal sources, reprinted with permission from Ninkuu et al. [<a href="#B37-antioxidants-12-01453" class="html-bibr">37</a>], published by Elsevier in 2021, copyright year 2023.</p>
Full article ">Figure 4
<p>Structures of vitamins, reprinted with permission from Asensi-Fabado and Munne-Bosch [<a href="#B43-antioxidants-12-01453" class="html-bibr">43</a>], published by Elsevier in 2010, copyright year 2023.</p>
Full article ">
31 pages, 1438 KiB  
Review
Exploring the Potential of Bee-Derived Antioxidants for Maintaining Oral Hygiene and Dental Health: A Comprehensive Review
by Poonam Choudhary, Surya Tushir, Manju Bala, Sanjula Sharma, Manjeet Kaur Sangha, Heena Rani, Nileshwari Raju Yewle, Parminder Kumar, Diksha Singla, Deepak Chandran, Manoj Kumar and Mohamed Mekhemar
Antioxidants 2023, 12(7), 1452; https://doi.org/10.3390/antiox12071452 - 19 Jul 2023
Cited by 3 | Viewed by 3434
Abstract
Honey bee products comprise various compounds, including honey, propolis, royal jelly, bee pollen, bee wax and bee venom, which have long been recognized for their pharmacological and health-promoting benefits. Scientists have discovered that periodontal disorders stem from dental biofilm, an inflammatory response to [...] Read more.
Honey bee products comprise various compounds, including honey, propolis, royal jelly, bee pollen, bee wax and bee venom, which have long been recognized for their pharmacological and health-promoting benefits. Scientists have discovered that periodontal disorders stem from dental biofilm, an inflammatory response to bacterial overgrowth produced by dysbiosis in the oral microbiome. The bee products have been investigated for their role in prevention of oral diseases, which are attributed to a myriad of biologically active compounds including flavonoids (pinocembrin, catechin, caffeic acid phenethyl ester (CAPE) and galangin), phenolic acids (hydroxybenzoic acid, hydroxycinnamic acid, p-coumaric, ellagic, caffeic and ferulic acids) and terpenoids. This review aims to update the current understanding of role of selected bee products, namely, honey, propolis and royal jelly, in preventing oral diseases as well as their potential biological activities and mechanism of action in relation to oral health have been discussed. Furthermore, the safety of incorporation of bee products is also critically discussed. To summarize, bee products could potentially serve as a therapy option for people suffering from a variety of oral disorders. Full article
(This article belongs to the Special Issue Antioxidant Activity of Honey Bee Products)
Show Figures

Figure 1

Figure 1
<p>Various components discussed in current review.</p>
Full article ">Figure 2
<p>Importance of polyphenols in bee products for oral health.</p>
Full article ">
22 pages, 4826 KiB  
Article
24-Epibrassinolide Facilitates Adventitious Root Formation by Coordinating Cell-Wall Polyamine Oxidase- and Plasma Membrane Respiratory Burst Oxidase Homologue-Derived Reactive Oxygen Species in Capsicum annuum L.
by Zhengyang Wen, Zhifeng Chen, Xinyan Liu, Jingbo Sun, Feng Zhang, Mengxia Zhang and Chunjuan Dong
Antioxidants 2023, 12(7), 1451; https://doi.org/10.3390/antiox12071451 - 19 Jul 2023
Cited by 1 | Viewed by 1447
Abstract
Adventitious root (AR) formation is a critical process in cutting propagation of horticultural plants. Brassinosteroids (BRs) have been shown to regulate AR formation in several plant species; however, little is known about their exact effects on pepper AR formation, and the downstream signaling [...] Read more.
Adventitious root (AR) formation is a critical process in cutting propagation of horticultural plants. Brassinosteroids (BRs) have been shown to regulate AR formation in several plant species; however, little is known about their exact effects on pepper AR formation, and the downstream signaling of BRs also remains elusive. In this study, we showed that treatment of 24-Epibrassinolide (EBL, an active BR) at the concentrations of 20–100 nM promoted AR formation in pepper (Capsicum annuum). Furthermore, we investigated the roles of apoplastic reactive oxygen species (ROS), including hydrogen peroxide (H2O2) and superoxide radical (O2•−), in EBL-promoted AR formation, by using physiological, histochemical, bioinformatic, and biochemical approaches. EBL promoted AR formation by modulating cell-wall-located polyamine oxidase (PAO)-dependent H2O2 production and respiratory burst oxidase homologue (RBOH)-dependent O2•− production, respectively. Screening of CaPAO and CaRBOH gene families combined with gene expression analysis suggested that EBL-promoted AR formation correlated with the upregulation of CaPAO1, CaRBOH2, CaRBOH5, and CaRBOH6 in the AR zone. Transient expression analysis confirmed that CaPAO1 was able to produce H2O2, and CaRBOH2, CaRBOH5, and CaRBOH6 were capable of producing O2•−. The silencing of CaPAO1, CaRBOH2, CaRBOH5, and CaRBOH6 in pepper decreased the ROS accumulation and abolished the EBL-induced AR formation. Overall, these results uncover one of the regulatory pathways for BR-regulated AR formation, and extend our knowledge of the functions of BRs and of the BRs-ROS crosstalk in plant development. Full article
(This article belongs to the Special Issue ROS Homeostasis during Plant Growth and Development)
Show Figures

Figure 1

Figure 1
<p>EBL promotes AR formation in the hypocotyls of pepper explants. (<b>A</b>–<b>C</b>) Effects of EBL on AR formation of pepper. Pepper explants with primary root excision were treated with EBL at different concentrations (0–5000 nM) for 10 days, followed by photographing (<b>A</b>), counting of AR number (<b>B</b>), and determination of AR average length (<b>C</b>). (<b>D</b>–<b>F</b>) Effects of BRz on AR formation of pepper. Pepper explants were treated with BRz at concentrations of 1.5 and 5.0 μM for 10 days. Each treatment has 15 pepper explants. Different lowercase letters indicate that the values were significantly different among different treatments (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 2
<p>Kinetics of changes in H<sub>2</sub>O<sub>2</sub> and O<sub>2</sub><sup>•−</sup> contents during AR formation in EBL- or BRz-treated pepper explants. (<b>A</b>,<b>B</b>) Contents of H<sub>2</sub>O<sub>2</sub> (<b>A</b>) and O<sub>2</sub><sup>•−</sup> (<b>B</b>) in the AR zone of pepper hypocotyls after different durations of EBL or BRz treatments. (<b>C</b>,<b>D</b>) Histochemical staining of H<sub>2</sub>O<sub>2</sub> (<b>C</b>) and O<sub>2</sub><sup>•−</sup> (<b>D</b>) in the AR zone after different durations of EBL or BRz treatments. Once primary roots were cut, pepper explants were treated with water (Control), EBL (50 nM), or BRz (5 μM), and used for AR formation. Hypocotyl samples were harvested at indicated hours (h) after treatment. One asterisk (*) and two asterisks (**) in (<b>A</b>,<b>B</b>) indicate that the mean values of three replicates were significantly different between control and treatment at each time point at <span class="html-italic">p</span> &lt; 0.05 and <span class="html-italic">p</span> &lt; 0.01, respectively. FW, fresh weight. The scale bar in (<b>C</b>,<b>D</b>) is 5 mm.</p>
Full article ">Figure 3
<p>Involvement of ROS in EBL-promoted AR formation in pepper explants. (<b>A</b>) Effects of KI and DMTU on EBL-induced AR formation. Once primary roots were cut, pepper explants were used for AR formation in water without (Control) or with EBL (50 nM), EBL + KI (0.5 mM), or EBL + DMTU (2 mM) for 10 days, followed by photographing phenotype and counting AR number. KI and DMTU were used as ROS scavengers. KI, potassium iodide; DMTU, <span class="html-italic">N</span>,<span class="html-italic">N</span>’-Dimethylthiourea. (<b>B</b>,<b>C</b>) Contents of H<sub>2</sub>O<sub>2</sub> (<b>B</b>) and O<sub>2</sub><sup>•−</sup> (<b>C</b>) in the AR zone of pepper explants treated with EBL, EBL + KI, or EBL + DMTU for 6, 24, 48, and 120 h. (<b>D</b>) Effects of CAT on EBL-induced AR formation. AR formation in pepper explants treated with EBL (50 nM) or EBL + CAT (200 unit/mg) for 10 days, followed by photographing phenotype and counting AR number. Each treatment has 15 pepper explants. Different lowercase letters indicated that the values were significantly different among different treatments (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 4
<p>Involvement of PAO-dependent H<sub>2</sub>O<sub>2</sub> and NADPH oxidase-dependent O<sub>2</sub><sup>•−</sup> in EBL-promoted AR formation in pepper explants: (<b>A</b>) Effects of AG, MDL72527, 2-HEH, and DPI on EBL-induced AR formation. Once primary roots were cut, pepper explants were used for AR formation in water with EBL (50 nM), EBL + AG (100 μM), EBL + MDL72527 (MDL, 100 μM), EBL + 2-HEH (100 μM), or EBL + DPI (10 μM) for 10 days, followed by photographing phenotype and counting AR number. DPI and AG were inhibitors of copper amine oxidase and NADPH oxidase, respectively; MDL and 2-HEH were PAO inhibitors. AG, aminoguanidine; 2-HEH, 2-hydroxyethylhydrazine; DPI, diphenyleneiodonium. (<b>B</b>) Effects of EBL on the activities of cell-wall PAO (CW-PAO) during AR formation. (<b>C</b>,<b>D</b>) Effects of MDL72527 and 2-HEH on EBL-induced CW-PAO activity (<b>C</b>) and H<sub>2</sub>O<sub>2</sub> content (<b>D</b>) at 24 h of AR formation. (<b>E</b>) Effects of EBL on the activities of plasma membrane NADPH oxidase (PM-NADPH oxidase) during AR formation. (<b>F</b>,<b>G</b>) Effects of DPI on EBL-induced PM-NADPH oxidase activity (<b>F</b>) and O<sub>2</sub><sup>•−</sup> content (<b>G</b>) at 72 h of AR formation. (<b>H</b>) Expression of <span class="html-italic">CaLBD</span>, <span class="html-italic">CaCYCLIN</span>, and <span class="html-italic">CaCDK</span> genes in response to EBL, EBL + MDL, and EBL + DPI treatments at 72 h of AR formation. The expression level for each gene in the mock plants at 0 dpe was normalized to 1.0. The accession numbers for these genes are listed in <a href="#app1-antioxidants-12-01451" class="html-app">Table S4</a>. Each treatment contains three biological replicates, and each replicate has 15 explants. Different lowercase letters in (<b>A</b>,<b>C</b>,<b>D</b>,<b>F</b>–<b>H</b>) indicate that the mean values of three replicates are significantly different among different treatments (<span class="html-italic">p</span> &lt; 0.05). Two asterisks (**) in (<b>B</b>,<b>E</b>) indicate significant differences between control and EBL treatment at <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 5
<p>Phylogenetic analysis of CaPAO family and EBL-induced expression of <span class="html-italic">CaPAO</span>s during AR formation in pepper explants: (<b>A</b>) Neighbor-joining phylogenetic tree of the CaPAO, AtPAO, and SlPAO proteins. The accession numbers for these proteins are listed in <a href="#app1-antioxidants-12-01451" class="html-app">Table S1</a>. (<b>B</b>) The secretory signal peptide in the N-terminal of CaPAO1 and SlPAO1 proteins. The green dashed square indicates the secretory signal peptide. Invariant residues are shaded in blue boxes, with residues that are conserved colored red, and variable residues shown in black. (<b>C</b>) Expression levels of <span class="html-italic">CaPAO</span> genes in the base of pepper hypocotyls before AR formation. The expression level of each <span class="html-italic">CaPAO</span> gene was normalized to <span class="html-italic">CaUBI3</span> expression. (<b>D</b>) Effects of EBL and BRz on the expression of <span class="html-italic">CaPAO</span> genes during AR formation. The expression level at 0 h was normalized to 1.0. Each treatment contains three biological replicates, and each replicate has 15 explants. Different lowercase letters in (<b>C</b>) indicate that the mean values of three replicates are significantly different among different genes (<span class="html-italic">p</span> &lt; 0.05). Two asterisks (**) in (<b>D</b>) indicate significant differences between control and EBL or BRz treatment at <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 6
<p>Phylogenetic analysis of CaRBOH family and EBL-induced expression of <span class="html-italic">CaRBOH</span>s during AR formation in pepper explants. (<b>A</b>) Neighbor-joining phylogenetic tree of CaRBOH, AtRBOH, and SlRBOH proteins. The accession numbers of these proteins are listed in <a href="#app1-antioxidants-12-01451" class="html-app">Supplementary Table S1</a>. (<b>B</b>) Expression levels of <span class="html-italic">CaRBOH</span> genes in the base of pepper hypocotyls before AR formation. The expression level of each gene was normalized to <span class="html-italic">CaUBI3</span> expression. (<b>C</b>) Effects of EBL and BRz on the expression of <span class="html-italic">CaRBOH</span> genes during AR formation in pepper hypocotyls. The expression level for each gene at 0 h was normalized to 1.0. Each treatment contains three biological replicates, and each replicate has 15 explants. Different lowercase letters in (<b>B</b>) indicate that the mean values of three replicates are significantly different among different genes (<span class="html-italic">p</span> &lt; 0.05). One asterisk (*) and two asterisks (**) in (<b>C</b>) indicate significant differences between control and EBL or BRz treatment at <span class="html-italic">p</span> &lt; 0.05 and <span class="html-italic">p</span> &lt; 0.01, respectively.</p>
Full article ">Figure 7
<p>Capability of CaPAO1, CaRBOH2, CaRBOH5, and CaRBOH6 in ROS production based on transient expression analysis. (<b>A</b>,<b>B</b>) Relative expression level of <span class="html-italic">CaPAO1</span> (<b>A</b>) and detection of endogenous H<sub>2</sub>O<sub>2</sub> by DAB staining (<b>B</b>) in pepper leaves expressing <span class="html-italic">CaPAO1</span>. (<b>C</b>,<b>D</b>) Relative expression levels of <span class="html-italic">CaRBOH2</span>, <span class="html-italic">CaRBOH5</span>, and <span class="html-italic">CaRBOH6</span> (<b>C</b>) and detection of endogenous O<sub>2</sub><sup>•−</sup> by NBT staining (<b>D</b>) in pepper leaves expressing <span class="html-italic">CaRBOH2</span>, <span class="html-italic">CaRBOH5</span>, and <span class="html-italic">CaRBOH6</span>. Control indicates the leaves without infiltration, and EV indicates infiltration of leaves with <span class="html-italic">Agrobacterium</span> carrying the empty vector. Each treatment has 15 pepper explants. In (<b>B</b>,<b>D</b>), the results show similar trends, and a representative result is shown. Different lowercase letters in (<b>A</b>,<b>C</b>) indicate that the mean values of three replicates were significantly different among different treatments (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 8
<p>Silencing of <span class="html-italic">CaPAO1</span> and <span class="html-italic">CaRBOH</span>s decreases AR formation in pepper. (<b>A</b>) Expression levels of <span class="html-italic">CaPAO1</span> and <span class="html-italic">CaRBOH</span>s in the epicotyls of control and VIGS plants. (<b>B</b>) H<sub>2</sub>O<sub>2</sub> contents in the AR zone of <span class="html-italic">CaPAO1</span>-silenced explants at 24 h of AR formation. (<b>C</b>) O<sub>2</sub><sup>•−</sup> contents in the AR zone of control and <span class="html-italic">CaRBOH</span>-silenced explants at 72 h of AR formation. (<b>D</b>) AR formation in the control and VIGS explants treated with or without EBL. Each treatment has 15 pepper explants. Different lowercase letters indicate that the mean values of ten replicates are significantly different among different treatments (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 9
<p>Schematic model for EBL-induced AR formation by coordinating ROS generation in pepper. EBL induces CaPAO1-derived H<sub>2</sub>O<sub>2</sub> generation and CaRBOH2/5/6-derived O<sub>2</sub><sup>•−</sup> generation in apoplasts. O<sub>2</sub><sup>•−</sup> and H<sub>2</sub>O<sub>2</sub> trigger signaling transduction to regulate the expression of <span class="html-italic">CaLBD</span>, <span class="html-italic">CaCYC</span>, and <span class="html-italic">CaCDK</span> genes, and further stimulate AR formation. PAs, polyamines. Put, putrescine. PM, plasma membrane.</p>
Full article ">
Previous Issue
Next Issue
Back to TopTop